1
|
Deng Q, Parker E, Duan R, Yang L. Preconditioning and Posttreatment Strategies in Neonatal Hypoxic-Ischemic Encephalopathy: Recent Advances and Clinical Challenges. Mol Neurobiol 2025:10.1007/s12035-025-04896-4. [PMID: 40178781 DOI: 10.1007/s12035-025-04896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a severe neurological disorder caused by impaired cerebral blood flow and brain hypoxia, resulting in high morbidity and mortality rates. While therapeutic hypothermia remains the standard treatment and has been shown to reduce mortality to some extent, its therapeutic efficacy is limited, and it applies only to a select group of neonates who meet stringent inclusion criteria. Advances in our understanding of the pathophysiology of HIE have led to the identification of several promising neuroprotective strategies designed to mitigate or prevent the neurological damage induced by hypoxia-ischemia. Among these, preconditioning has emerged as a potent neuroprotective approach, enhancing cellular resilience to subsequent injury and potentially reducing treatment complexity and healthcare costs. Preconditioning/pretreatment and posttreatment offer significant promise in attenuating the neurological damage associated with HIE. Thus, exploring early intervention strategies for neonatal HIE, focusing on the comparative mechanisms and therapeutic targets of preconditioning and postconditioning, is critical to developing more effective treatment modalities. This review summarizes the current understanding of the pathophysiological mechanisms underlying neonatal HIE and its prevention and treatment strategies, providing new perspectives and a theoretical foundation for future neuroprotective interventions.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, College of Physical Education and Sport Science, South China Normal University, Guangzhou, 510006, GD, China
| | - Emily Parker
- Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Rui Duan
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, College of Physical Education and Sport Science, South China Normal University, Guangzhou, 510006, GD, China.
| |
Collapse
|
2
|
Toczylowska B, Kalinowski P, Kacka-Piotrowska A, Duda P, Grąt M, Zieminska E. Metabolic Pattern of Brain Death-NMR-Based Metabolomics of Cerebrospinal Fluid. Int J Mol Sci 2025; 26:2719. [PMID: 40141360 PMCID: PMC11942502 DOI: 10.3390/ijms26062719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 03/28/2025] Open
Abstract
The aim of this study was to gain insight into the biochemical status of cerebrospinal fluid in the presence of brain death in life-supported patients. The biochemical status was determined via in vitro NMR spectroscopy of cerebrospinal fluid (CSF) obtained by lumbar puncture from 22 patients with confirmed brain death and compared with that of 34 control patients (without neurological diseases). Forty-one NMR signals from raw CSF samples and 20 signals from lipid extracts were analyzed using univariate and multivariate statistical methods. ANOVA revealed significant differences in all analyzed signals. No single biochemical marker was found to predict brain death. The CSF metabolic profiles of patients who died differed significantly from those of patients in the control group. There were many statistically significantly different compounds, including amino acids, ketone bodies, lactate, pyruvate, citrate, guanidinoacetate, choline, and glycerophosphocholine. Analysis of lipids revealed significant differences in cholesterol, estriol, and phosphoethanolamine. Discriminant analysis allows the analysis of metabolic profiles instead of single biomarkers of cerebrospinal fluid compounds. The results of our analysis allowed us to split the groups-the control group, which consisted of patients with a normal biochemical CSF composition, and the brain death group-with confirmed brain death.
Collapse
Affiliation(s)
- Beata Toczylowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 4 Trojdena Street, 02-109 Warsaw, Poland; (B.T.); (P.D.)
| | - Piotr Kalinowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland; (P.K.); (M.G.)
| | | | - Paulina Duda
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 4 Trojdena Street, 02-109 Warsaw, Poland; (B.T.); (P.D.)
| | - Michał Grąt
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland; (P.K.); (M.G.)
| | - Elzbieta Zieminska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| |
Collapse
|
3
|
Olajide OJ, Batallán Burrowes AA, da Silva IF, Bergdahl A, Chapman CA. Reduced 17β-estradiol following ovariectomy induces mitochondrial dysfunction and degradation of synaptic proteins in the entorhinal cortex. Neuroscience 2025; 565:479-486. [PMID: 39617168 DOI: 10.1016/j.neuroscience.2024.11.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Reductions in circulating estrogens can contribute to cognitive decline, in part by impairing mitochondrial function within the hippocampal region. The entorhinal cortex provides the hippocampus with its main cortical inputs. To assess the impact of estrogen deficiency on mitochondrial respiration and synaptic proteins in the entorhinal cortex, female wildtype rats received either sham surgery, bilateral ovariectomy, or ovariectomy with implantation of a subdermal capsule to maintain low levels of circulating 17β-estradiol (E2). Mitochondrial respiration in the entorhinal cortex was not significantly affected two weeks following ovariectomy, but there was a reduction in oxygen consumption four weeks after ovariectomy that was prevented by E2 supplementation. The expression of mitochondrial membrane integrity element voltage-dependent anion channel protein (VDAC1) was also reduced four weeks after ovariectomy, suggesting that respiration was reduced due to a decline in mitochondrial density. Ovariectomy also increased mitochondrial and cytoplasmic cytochrome c and upregulated superoxide dismutase 2 (SOD2) both two and four weeks after ovariectomy, reflecting mitochondrial electron leakage and oxidative redox imbalance. Further, the ovariectomy-induced changes in mitochondrial proteins were associated with reductions in postsynaptic density protein 95 (PSD95) and the presynaptic protein synaptophysin. There were no changes in mitochondrial or synaptic proteins in ovariectomized animals that received E2 supplementation. Our findings indicate that reductions in circulating 17β-estradiol induced by ovariectomy disrupt mitochondrial functions in the entorhinal cortex, and suggest that a resulting increase in oxidative stress contributes to the degradation in synaptic proteins that may affect cognitive functions mediated by the hippocampal region.
Collapse
Affiliation(s)
- Olayemi Joseph Olajide
- Department of Psychology, Concordia University, Montreal, Canada; Division of Neurobiology, Department of Anatomy, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | | | - Igor Ferraz da Silva
- Department of Psychology, Concordia University, Montreal, Canada; Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - Andreas Bergdahl
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
| | - C Andrew Chapman
- Department of Psychology, Concordia University, Montreal, Canada.
| |
Collapse
|
4
|
Oviedo JM, Cortes-Selva D, Marchetti M, Gordon L, Gibbs L, Maschek JA, Cox J, Frietze S, Amiel E, Fairfax KC. Schistosoma mansoni antigen induced innate immune memory features mitochondrial biogenesis and can be inhibited by ovarian produced hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.632838. [PMID: 39868249 PMCID: PMC11761400 DOI: 10.1101/2025.01.14.632838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
We have previously identified that S. mansoni infection induces a unique form of myeloid training that protects male but not female mice from high fat diet induced disease. Here we demonstrate that ovarian derived hormones account for this sex specific difference. Ovariectomy of females prior to infection permits metabolic reprogramming of the myeloid lineage, with BMDM exhibiting carbon source flexibility for cellular respiration, and mice protected from systemic metabolic disease. The innate training phenotype of infection can be replicated by in vivo injection of SEA, and by exposure of bone marrow to SEA in culture prior to macrophage differentiation (Day 0). This protective phenotype is linked to increased chromatin accessibility of lipid and mitochondrial pathways in BMDM including Nrf1 and Tfam, as well as mitochondrial biogenesis. This work provides evidence that S. mansoni antigens induce a unique form of innate training inhibited by ovarian-derived hormones in females.
Collapse
Affiliation(s)
- Juan Marcos Oviedo
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| | - Diana Cortes-Selva
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| | - Marco Marchetti
- Department of Human Genetics, Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, 84112, USA
| | | | - Lisa Gibbs
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| | - J. Alan Maschek
- Metabolomics, Proteomics and Mass Spectrometry Cores, University of Utah, Salt Lake City, UT, 84112
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112
| | - James Cox
- Department of Biochemistry, University of Utah, Salt Lake City UT, 84112
- Metabolomics, Proteomics and Mass Spectrometry Cores, University of Utah, Salt Lake City, UT, 84112
| | - Seth Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405
| | - Keke C. Fairfax
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City UT, 84112
| |
Collapse
|
5
|
Arjmand S, Ilaghi M, Sisakht AK, Guldager MB, Wegener G, Landau AM, Gjedde A. Regulation of mitochondrial dysfunction by estrogens and estrogen receptors in Alzheimer's disease: A focused review. Basic Clin Pharmacol Toxicol 2024; 135:115-132. [PMID: 38801027 DOI: 10.1111/bcpt.14035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that primarily manifests itself by progressive memory loss and cognitive decline, thus significantly affecting memory functions and quality of life. In this review, we proceed from the understanding that the canonical amyloid-β hypothesis, while significant, has faced setbacks, highlighting the need to adopt a broader perspective considering the intricate interplay of diverse pathological pathways for effective AD treatments. Sex differences in AD offer valuable insights into a better understanding of its pathophysiology. Fluctuation of the levels of ovarian sex hormones during perimenopause is associated with changes in glucose metabolism, as a possible window of opportunity to further understand the roles of sex steroid hormones and their associated receptors in the pathophysiology of AD. We review these dimensions, emphasizing the potential of estrogen receptors (ERs) to reveal mitochondrial functions in the search for further research and therapeutic strategies for AD pharmacotherapy. Understanding and addressing the intricate interactions of mitochondrial dysfunction and ERs potentially pave the way for more effective approaches to AD therapy.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Karimi Sisakht
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Matti Bock Guldager
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Padamsey Z, Katsanevaki D, Maeso P, Rizzi M, Osterweil EE, Rochefort NL. Sex-specific resilience of neocortex to food restriction. eLife 2024; 12:RP93052. [PMID: 38976495 PMCID: PMC11230624 DOI: 10.7554/elife.93052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024] Open
Abstract
Mammals have evolved sex-specific adaptations to reduce energy usage in times of food scarcity. These adaptations are well described for peripheral tissue, though much less is known about how the energy-expensive brain adapts to food restriction, and how such adaptations differ across the sexes. Here, we examined how food restriction impacts energy usage and function in the primary visual cortex (V1) of adult male and female mice. Molecular analysis and RNA sequencing in V1 revealed that in males, but not in females, food restriction significantly modulated canonical, energy-regulating pathways, including pathways associated waith AMP-activated protein kinase, peroxisome proliferator-activated receptor alpha, mammalian target of rapamycin, and oxidative phosphorylation. Moreover, we found that in contrast to males, food restriction in females did not significantly affect V1 ATP usage or visual coding precision (assessed by orientation selectivity). Decreased serum leptin is known to be necessary for triggering energy-saving changes in V1 during food restriction. Consistent with this, we found significantly decreased serum leptin in food-restricted males but no significant change in food-restricted females. Collectively, our findings demonstrate that cortical function and energy usage in female mice are more resilient to food restriction than in males. The neocortex, therefore, contributes to sex-specific, energy-saving adaptations in response to food restriction.
Collapse
Affiliation(s)
- Zahid Padamsey
- Wellcome-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Danai Katsanevaki
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
| | - Patricia Maeso
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Manuela Rizzi
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Emily E Osterweil
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
- Rosamund Stone Zander Translational Neuroscience Center, F.M. Kirby Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Nathalie L Rochefort
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
7
|
Rishabh, Rohilla M, Bansal S, Bansal N, Chauhan S, Sharma S, Goyal N, Gupta S. Estrogen signalling and Alzheimer's disease: Decoding molecular mechanisms for therapeutic breakthrough. Eur J Neurosci 2024; 60:3466-3490. [PMID: 38726764 DOI: 10.1111/ejn.16360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 07/06/2024]
Abstract
In females, Alzheimer's disease (AD) incidences increases as compared to males due to estrogen deficiency after menopause. Estrogen therapy is the mainstay therapy for menopause and associated complications. Estrogen, a hormone with multifaceted physiological functions, has been implicated in AD pathophysiology. Estrogen plays a crucial role in amyloid precursor protein (APP) processing and overall neuronal health by regulating various factors such as brain-derived neurotrophic factor (BDNF), intracellular calcium signalling, death domain-associated protein (Daxx) translocation, glutamatergic excitotoxicity, Voltage-Dependent Anion Channel, Insulin-Like Growth Factor 1 Receptor, estrogen-metabolising enzymes and apolipoprotein E (ApoE) protein polymorphisms. All these factors impact the physiology of postmenopausal women. Estrogen replacement therapies play an important treatment strategy to prevent AD after menopause. However, use of these therapies may lead to increased risks of breast cancer, venous thromboembolism and cardiovascular disease. Various therapeutic approaches have been used to mitigate the effects of estrogen on AD. These include hormone replacement therapy, Selective Estrogen Receptor Modulators (SERMs), Estrogen Receptor Beta (ERβ)-Selective Agonists, Transdermal Estrogen Delivery, Localised Estrogen Delivery, Combination Therapies, Estrogen Metabolism Modulation and Alternative Estrogenic Compounds like genistein from soy, a notable phytoestrogen from plant sources. However, mechanism via which these approaches modulate AD in postmenopausal women has not been explained earlier thoroughly. Present review will enlighten all the molecular mechanisms of estrogen and estrogen replacement therapies in AD. Along-with this, the association between estrogen, estrogen-metabolising enzymes and ApoE protein polymorphisms will also be discussed in postmenopausal AD.
Collapse
Affiliation(s)
- Rishabh
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Manni Rohilla
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Seema Bansal
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Nitin Bansal
- Department of Pharmacy, Chaudhary Bansilal University, Bhiwani, India
| | - Samrat Chauhan
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sheenam Sharma
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Navjyoti Goyal
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Sumeet Gupta
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| |
Collapse
|
8
|
Yousif A, Ebeid A, Kacsoh B, Bazzaro M, Chefetz I. The Ovary-Brain Connection. Cells 2024; 13:94. [PMID: 38201298 PMCID: PMC10778337 DOI: 10.3390/cells13010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The brain and the ovaries are in a state of continuous communication [...].
Collapse
Affiliation(s)
- Abdelrahman Yousif
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Ahmed Ebeid
- Department of Obstetrics and Gynecology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Balint Kacsoh
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Biomedical and Clinical Science, Linköping University, SE-581 85 Linköping, Sweden
| | - Ilana Chefetz
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|
9
|
Ekanayake A, Peiris S, Ahmed B, Kanekar S, Grove C, Kalra D, Eslinger P, Yang Q, Karunanayaka P. A Review of the Role of Estrogens in Olfaction, Sleep and Glymphatic Functionality in Relation to Sex Disparity in Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2024; 39:15333175241272025. [PMID: 39116421 PMCID: PMC11311174 DOI: 10.1177/15333175241272025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Several risk factors contribute to the development of Alzheimer's disease (AD), including genetics, metabolic health, cardiovascular history, and diet. It has been observed that women appear to face a higher risk of developing AD. Among the various hypotheses surrounding the gender disparity in AD, one pertains to the potential neuroprotective properties of estrogen. Compared to men, women are believed to be more susceptible to neuropathology due to the significant decline in circulating estrogen levels following menopause. Studies have shown, however, that estrogen replacement therapies in post-menopausal women do not consistently reduce the risk of AD. While menopause and estrogen levels are potential factors in the elevated incidence rates of AD among women, this review highlights the possible roles estrogen has in other pathways that may also contribute to the sex disparity observed in AD such as olfaction, sleep, and glymphatic functionality.
Collapse
Affiliation(s)
- Anupa Ekanayake
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
- Grodno State Medical University, Grodno, Belarus
| | - Senal Peiris
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Biyar Ahmed
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Sangam Kanekar
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Cooper Grove
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| | - Deepak Kalra
- Department of Neurology, Penn State University College of Medicine, Hershey, PA, USA
| | - Paul Eslinger
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
- Department of Neurology, Penn State University College of Medicine, Hershey, PA, USA
| | - Qing Yang
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
- Department of Neurosurgery, Penn State University College of Medicine, Hershey, PA, USA
| | - Prasanna Karunanayaka
- Department of Radiology, Penn State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
10
|
Zhu J, Zhou Y, Jin B, Shu J. Role of estrogen in the regulation of central and peripheral energy homeostasis: from a menopausal perspective. Ther Adv Endocrinol Metab 2023; 14:20420188231199359. [PMID: 37719789 PMCID: PMC10504839 DOI: 10.1177/20420188231199359] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Estrogen plays a prominent role in regulating and coordinating energy homeostasis throughout the growth, development, reproduction, and aging of women. Estrogen receptors (ERs) are widely expressed in the brain and nearly all tissues of the body. Within the brain, central estrogen via ER regulates appetite and energy expenditure and maintains cell glucose metabolism, including glucose transport, aerobic glycolysis, and mitochondrial function. In the whole body, estrogen has shown beneficial effects on weight control, fat distribution, glucose and insulin resistance, and adipokine secretion. As demonstrated by multiple in vitro and in vivo studies, menopause-related decline of circulating estrogen may induce the disturbance of metabolic signals and a significant decrease in bioenergetics, which could trigger an increased incidence of late-onset Alzheimer's disease, type 2 diabetes mellitus, hypertension, and cardiovascular diseases in postmenopausal women. In this article, we have systematically reviewed the role of estrogen and ERs in body composition and lipid/glucose profile variation occurring with menopause, which may provide a better insight into the efficacy of hormone therapy in maintaining energy metabolic homeostasis and hold a clue for development of novel therapeutic approaches for target tissue diseases.
Collapse
Affiliation(s)
- Jing Zhu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yier Zhou
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bihui Jin
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jing Shu
- Reproductive Medicine Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
11
|
Valencia-Olvera AC, Maldonado Weng J, Christensen A, LaDu MJ, Pike CJ. Role of estrogen in women's Alzheimer's disease risk as modified by APOE. J Neuroendocrinol 2023; 35:e13209. [PMID: 36420620 PMCID: PMC10049970 DOI: 10.1111/jne.13209] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by numerous sexual dimorphisms that impact the development, progression, and probably the strategies to prevent and treat the most common form of dementia. In this review, we consider this topic from a female perspective with a specific focus on how women's vulnerability to the disease is affected by the individual and interactive effects of estrogens and apolipoprotein E (APOE) genotype. Importantly, APOE appears to modulate systemic and neural outcomes of both menopause and estrogen-based hormone therapy. In the brain, dementia risk is greater in APOE4 carriers, and the impacts of hormone therapy on cognitive decline and dementia risk vary according to both outcome measure and APOE genotype. Beyond the CNS, estrogen and APOE genotype affect vulnerability to menopause-associated bone loss, dyslipidemia and cardiovascular disease risk. An emerging concept that may link these relationships is the possibility that the effects of APOE in women interact with estrogen status by mechanisms that may include modulation of estrogen responsiveness. This review highlights the need to consider the key AD risk factors of advancing age in a sex-specific manner to optimize development of therapeutic approaches for AD, a view aligned with the principle of personalized medicine.
Collapse
Affiliation(s)
- AC Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - J Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - A Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| | - MJ LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - CJ Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|
12
|
Blackwell JA, Silva JF, Louis EM, Savu A, Largent-Milnes TM, Brooks HL, Pires PW. Cerebral arteriolar and neurovascular dysfunction after chemically induced menopause in mice. Am J Physiol Heart Circ Physiol 2022; 323:H845-H860. [PMID: 36149767 PMCID: PMC9602916 DOI: 10.1152/ajpheart.00276.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 12/14/2022]
Abstract
Cognitive decline is linked to decreased cerebral blood flow, particularly in women after menopause. Impaired cerebrovascular function precedes the onset of dementia, possibly because of reduced functional dilation in parenchymal arterioles. These vessels are bottlenecks of the cerebral microcirculation, and dysfunction can limit functional hyperemia in the brain. Large-conductance Ca2+-activated K+ channels (BKCa) are the final effectors of several pathways responsible for functional hyperemia, and their expression is modulated by estrogen. However, it remains unknown whether BKCa function is altered in cerebral parenchymal arterioles after menopause. Using a chemically induced model of menopause, the 4-vinylcyclohexene diepoxide (VCD) model, which depletes follicles while maintaining intact ovaries, we hypothesized that menopause would be associated with reduced functional vasodilatory responses in cerebral parenchymal arterioles of wild-type mice via reduced BKCa function. Using pressure myography of isolated parenchymal arterioles, we observed that menopause (Meno) induced a significant increase in spontaneous myogenic tone. Endothelial function, assessed as nitric oxide production and dilation after cholinergic stimulation or endothelium-dependent hyperpolarization pathways, was unaffected by Meno. BKCa function was significantly impaired in Meno compared with control, without changes in voltage-gated K+ channel activity. Cerebral functional hyperemia, measured by laser-speckle contrast imaging during whisker stimulation, was significantly blunted in Meno mice, without detectable changes in basal perfusion. However, behavioral testing identified no change in cognition. These findings suggest that menopause induces cerebral microvascular and neurovascular deficits.NEW & NOTEWORTHY Cerebral parenchymal arterioles from menopause mice showed increased myogenic tone. We identified an impairment in smooth muscle cell BKCa channel activity, without a reduction in endothelium-dependent dilation or nitric oxide production. Microvascular dysfunction was associated with a reduction in neurovascular responses after somatosensory stimulation. Despite the neurovascular impairment, cognitive abilities were maintained in menopausal mice.
Collapse
Affiliation(s)
- Jade A Blackwell
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Josiane F Silva
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Emma M Louis
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Andrea Savu
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Tally M Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Paulo W Pires
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
13
|
Álvarez-Delgado C. The role of mitochondria and mitochondrial hormone receptors on the bioenergetic adaptations to lactation. Mol Cell Endocrinol 2022; 551:111661. [PMID: 35483518 DOI: 10.1016/j.mce.2022.111661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/18/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022]
Abstract
The most recognized role of mitochondria is producing more than 90% of the total cellular energy in the form of ATP. In addition, mitochondrial function encompasses the maintenance of antioxidant balance, the regulation of intracellular calcium concentrations, the progression of cell death, and the biosynthesis of purines, hemes, lipids, amino acids and steroid hormones. Mitochondria are also important hormone targets. Estrogens, progestagens, and prolactin, are among the hormones that can impact mitochondrial function and modulate the underlying adaptations to changing bioenergetic and metabolic needs. Lactation represents a metabolic challenge with significant increases in energy requirements and fluctuating levels of hormones. To meet these bioenergetic demands, liver mitochondria increase their state 3 and 4 respiration, adjust superoxide dismutase activity, and elevate succinate dehydrogenase-related respiration. Skeletal muscle mitochondria respond by increasing their respiratory control ratio and adjusting catalase activity. In this review, these adaptations are described considering the lactation hormonal milieu.
Collapse
Affiliation(s)
- Carolina Álvarez-Delgado
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
| |
Collapse
|
14
|
Snäkä T, Bekkar A, Desponds C, Prével F, Claudinot S, Isorce N, Teixeira F, Grasset C, Xenarios I, Lopez-Mejia IC, Fajas L, Fasel N. Sex-Biased Control of Inflammation and Metabolism by a Mitochondrial Nod-Like Receptor. Front Immunol 2022; 13:882867. [PMID: 35651602 PMCID: PMC9150262 DOI: 10.3389/fimmu.2022.882867] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/05/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondria regulate steroid hormone synthesis, and in turn sex hormones regulate mitochondrial function for maintaining cellular homeostasis and controlling inflammation. This crosstalk can explain sex differences observed in several pathologies such as in metabolic or inflammatory disorders. Nod-like receptor X1 (NLRX1) is a mitochondria-associated innate receptor that could modulate metabolic functions and attenuates inflammatory responses. Here, we showed that in an infectious model with the human protozoan parasite, Leishmania guyanensis, NLRX1 attenuated inflammation in females but not in male mice. Analysis of infected female and male bone marrow derived macrophages showed both sex- and genotype-specific differences in both inflammatory and metabolic profiles with increased type I interferon production, mitochondrial respiration, and glycolytic rate in Nlrx1-deficient female BMDMs in comparison to wild-type cells, while no differences were observed between males. Transcriptomics of female and male BMDMs revealed an altered steroid hormone signaling in Nlrx1-deficient cells, and a “masculinization” of Nlrx1-deficient female BMDMs. Thus, our findings suggest that NLRX1 prevents uncontrolled inflammation and metabolism in females and therefore may contribute to the sex differences observed in infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Tiia Snäkä
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Amel Bekkar
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Chantal Desponds
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Florence Prével
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Nathalie Isorce
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Filipa Teixeira
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Coline Grasset
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Ioannis Xenarios
- Agora Center, Center Hospitalier Universitaire (CHUV), Lausanne, Switzerland.,Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
15
|
Ren Y, Zheng J, Wang H. Transiently gene-modulated cell reporter for ultrasensitive detection of estrogen-like compounds in tap water. CHEMOSPHERE 2022; 289:133161. [PMID: 34883127 DOI: 10.1016/j.chemosphere.2021.133161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/13/2021] [Accepted: 12/02/2021] [Indexed: 06/13/2023]
Abstract
Abnormal elevation of indispensable steroid hormone estrogens and exposure to exogenous estrogen-like compounds pose adverse health effects to aquatic animals and human alike. These compounds generally display functionally important estrogenic activity even at extremely low picomolar concentrations. In this study we identified one critical but lethal gene (TAF1) that remarkably represses estrogenic activity. This gene is selected as a candidate for genetically modulating an estrogen-responding cell line. To overcome its lethality, instead of adopting a gene knockout strategy, we developed a transient TAF1 depletion strategy using a designed small interfering RNA. By the transient knockdown of TAF1 in the estrogen-responding reporter cell line, the maximum induction signals for endogenous estrogen 17β-estradiol (E2) and environmental estrogens 17α-ethynyl estradiol (EE2) and bisphenol compounds were enhanced by 4.8-13.3 folds. The limit of detection for EE2 is about 8 × 10-15 mol/L. Moreover, by the established method, trace estrogenic activity (14.7-24.2 pg E2 equivalents (E2Eq)/L) can be detected in a portion of Tap water samples.
Collapse
Affiliation(s)
- Yun Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Zheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hailin Wang
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Institute of Environment and Health, Jianghan University, Wuhan, 430056, China.
| |
Collapse
|
16
|
Kuwahara N, Nicholson K, Isaacs L, MacLusky NJ. Androgen Effects on Neural Plasticity. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:216-230. [PMID: 35024693 PMCID: PMC8744448 DOI: 10.1089/andro.2021.0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Androgens are synthesized in the brain, gonads, and adrenal glands, in both sexes, exerting physiologically important effects on the structure and function of the central nervous system. These effects may contribute to the incidence and progression of neurological disorders such as autism spectrum disorder, schizophrenia, and Alzheimer's disease, which occur at different rates in males and females. This review briefly summarizes the current state of knowledge with respect to the neuroplastic effects of androgens, with particular emphasis on the hippocampus, which has been the focus of much of the research in this field.
Collapse
Affiliation(s)
- Nariko Kuwahara
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kate Nicholson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Lauren Isaacs
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Neil J. MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
17
|
Demetrius LA, Eckert A, Grimm A. Sex differences in Alzheimer's disease: metabolic reprogramming and therapeutic intervention. Trends Endocrinol Metab 2021; 32:963-979. [PMID: 34654630 DOI: 10.1016/j.tem.2021.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/05/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Studies on the sporadic form of Alzheimer's disease (AD) have revealed three classes of risk factor: age, genetics, and sex. These risk factors point to a metabolic dysregulation as the origin of AD. Adaptive alterations in cerebral metabolism are the rationale for the Metabolic Reprogramming (MR) Theory of the origin of AD. The theory contends that the progression toward AD involves three adaptive events: a hypermetabolic phase, a prolonged prodromal phase, and a metabolic collapse. This article exploits the MR Theory to elucidate the effect of hormonal changes on the origin and progression of AD in women. The theory invokes bioenergetic signatures of the menopausal transition to propose sex-specific diagnostic program and therapeutic strategies.
Collapse
Affiliation(s)
- Lloyd A Demetrius
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anne Eckert
- University of Basel, Transfaculty Research Platform Molecular and Cognitive Neuroscience, 4002 Basel, Switzerland; Neurobiology Lab for Brain Aging and Mental Health, Psychiatric University Clinics, 4002 Basel, Switzerland
| | - Amandine Grimm
- University of Basel, Transfaculty Research Platform Molecular and Cognitive Neuroscience, 4002 Basel, Switzerland; Neurobiology Lab for Brain Aging and Mental Health, Psychiatric University Clinics, 4002 Basel, Switzerland; University of Basel, Life Sciences Training Facility, 4055 Basel, Switzerland.
| |
Collapse
|
18
|
Dewsbury LS, Lim CK, Steiner GZ. The Efficacy of Ketogenic Therapies in the Clinical Management of People with Neurodegenerative Disease: A Systematic Review. Adv Nutr 2021; 12:1571-1593. [PMID: 33621313 PMCID: PMC8321843 DOI: 10.1093/advances/nmaa180] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/10/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Ketone bodies have potential disease-modifying activity that represent a novel therapeutic approach for neurodegenerative diseases (NDD). The aim of this systematic review was to summarize and evaluate the evidence for the application of ketogenic therapies (dietary or exogenous ketogenic agents) for NDD and provide recommendations for future research. Eight databases were electronically searched for articles reporting on controlled trials (≥4 wk duration) that induced ketosis or elevated serum ketone concentrations in people with NDD. Of 4498 records identified, 17 articles met the inclusion criteria with a total of 979 participants including studies on mild cognitive impairment (MCI; n = 6), multiple sclerosis (n = 4), Alzheimer's disease (n = 5), Parkinson's disease (n = 1), and MCI secondary to Parkinson's disease (n = 1). Of 17 studies, 7 were randomized double-blind placebo-controlled trials. Most studies used dietary interventions (n = 9), followed by medium-chain triglycerides (n = 7) and a fasting protocol (n = 1). Generally, trials were 6 wk in duration and assessed cognition as the primary outcome. Studies were heterogeneous in type and severity of NDD, interventions used, and outcomes assessed. Overall, 3/17 studies carried a low risk of bias. Based on available evidence, exogenous ketogenic agents may be more feasible than dietary interventions in NDD from a compliance and adherence perspective; more research is required to confirm this. Recommendations for future research include improving exogenous formulations to reduce adverse effects, exploring interindividual factors affecting response-to-treatment, and establishing a "minimum required dose" for clinically meaningful improvements in disease-specific symptoms, such as cognition or motor function.
Collapse
Affiliation(s)
- Lauren S Dewsbury
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, Australia
| | - Chai K Lim
- Department of Biomedical Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Macquarie Park, New South Wales, Australia
| | - Genevieve Z Steiner
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, Australia
- Translational Health Research Institute (THRI), Western Sydney University, Penrith, New South Wales, Australia
| |
Collapse
|
19
|
Di Cesare F, Tenori L, Meoni G, Gori AM, Marcucci R, Giusti B, Molino-Lova R, Macchi C, Pancani S, Luchinat C, Saccenti E. Lipid and metabolite correlation networks specific to clinical and biochemical covariate show differences associated with sexual dimorphism in a cohort of nonagenarians. GeroScience 2021; 44:1109-1128. [PMID: 34324142 PMCID: PMC9135919 DOI: 10.1007/s11357-021-00404-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/13/2021] [Indexed: 12/26/2022] Open
Abstract
This study defines and estimates the metabolite-lipidic component association networks constructed from an array of 20 metabolites and 114 lipids identified and quantified via NMR spectroscopy in the serum of a cohort of 355 Italian nonagenarians and ultra-nonagenarian. Metabolite-lipid association networks were built for men and women and related to an array of 101 clinical and biochemical parameters, including the presence of diseases, bio-humoral parameters, familiarity diseases, drugs treatments, and risk factors. Different connectivity patterns were observed in lipids, branched chains amino acids, alanine, and ketone bodies, suggesting their association with the sex-related and sex-clinical condition-related intrinsic metabolic changes. Furthermore, our results demonstrate, using a holistic system biology approach, that the characterization of metabolic structures and their dynamic inter-connections is a promising tool to shed light on the dimorphic pathophysiological mechanisms of aging at the molecular level.
Collapse
Affiliation(s)
- Francesca Di Cesare
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy
| | - Leonardo Tenori
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | | | - Anna Maria Gori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Atherothrombotic Unit, Careggi University Hospital, Florence, Italy
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Atherothrombotic Unit, Careggi University Hospital, Florence, Italy
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Atherothrombotic Unit, Careggi University Hospital, Florence, Italy
| | | | - Claudio Macchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | | | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Sesto Fiorentino, Italy
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, the Netherlands.
| |
Collapse
|
20
|
Zhao W, Hou Y, Song X, Wang L, Zhang F, Zhang H, Yu H, Zhou Y. Estrogen Deficiency Induces Mitochondrial Damage Prior to Emergence of Cognitive Deficits in a Postmenopausal Mouse Model. Front Aging Neurosci 2021; 13:713819. [PMID: 34335235 PMCID: PMC8319728 DOI: 10.3389/fnagi.2021.713819] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Estrogen deficiency contributes to the development of Alzheimer's disease (AD) in menopausal women. In the current study, we examined the impact of estrogen deficiency on mitochondrial function and cognition using a postmenopausal mouse model. Methods: Bilateral ovariectomy was conducted in adult females C57BL/6J. Cognitive function was examined using the Morris water maze (MWM) test at 2 weeks, 1, 2, and 3 months after ovariectomy. Neurodegeneration was assessed using an immunofluorescence assay of microtubule-associated protein 2 (MAP2) in the hippocampus and immunoblotting against postsynaptic density-95 (PSD95). Mitochondrial function in the hippocampus was assessed using immunoblotting for NDUFB8, SDHB, UQCRC2, MTCO1, and ATP5A1. Mitochondrial biogenesis was examined using immunoblotting for PGC-1α, NRF1, and mtTFA. Mitochondrion fission was assessed with immunoblotting for Drp1, whereas mitochondrion fusion was analyzed with immunoblotting for OPA1 and Mfn2. Mitophagy was examined with immunoblotting for PINK1 and LC3B. Mice receiving sham surgery were used as controls. Results: Ovariectomy resulted in significant learning and memory deficits in the MWM test at 3 months, but not at any earlier time points. At 2 weeks after ovariectomy, levels of Drp1 phosphorylated at Ser637 decreased in the hippocampus. At 1 month after ovariectomy, hippocampal levels of NDUFB8, SDHB, PGC-1α, mtTFA, OPA1, and Mfn2 were significantly reduced. At 2 months after ovariectomy, hippocampal levels of MAP2, PSD95, MTCO1, NRF1, and Pink1 were also reduced. At 3 months, levels of LC3B-II were reduced. Conclusions: The cognitive decline associated with estrogen deficiency is preceded by mitochondrial dysfunction, abnormal mitochondrial biogenesis, irregular mitochondrial dynamics, and decreased mitophagy. Thus, mitochondrial damage may contribute to cognitive impairment associated with estrogen deficiency.
Collapse
Affiliation(s)
- Wei Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yue Hou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Lei Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Fangfang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Hanting Zhang
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Haiyang Yu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| |
Collapse
|
21
|
Kim YJ, Soto M, Branigan GL, Rodgers K, Brinton RD. Association between menopausal hormone therapy and risk of neurodegenerative diseases: Implications for precision hormone therapy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12174. [PMID: 34027024 PMCID: PMC8118114 DOI: 10.1002/trc2.12174] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The impact of menopausal hormone therapy (HT) on age-associated Alzheimer's and neurodegenerative diseases (NDDs) remains unresolved. To determine the effect of HT, formulation, type, and duration on risk of NDDs, a retrospective analysis was performed using a 10-year Humana claims dataset. METHODS Study population included women aged 45 years or older with or without claim records of HT medications. Patients diagnosed with NDDs including Alzheimer's disease (AD), Parkinson's disease (PD), dementia, multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS) were identified. Relative risk (RR) ratios and 95% confidence intervals (CI) for combined NDDs, or AD, PD, dementia, MS, and ALS were determined. Cumulative hazard ratios were determined to investigate the association between HT and NDDs at different age groups. RESULTS In 379,352 women with or without claim records of HT, use of HT was associated with significantly reduced risk for combined NDDs (RR 0.42, 95% CI 0.40-0.43, P < 0.001). Average follow-up time was 5.1 [2.3] years. Formulations containing natural steroids 17β-estradiol and/or progesterone were associated with greater reduction in NDD risk. Oral- HT users showed significantly reduced RRs (0.42, 0.41-0.44, P < 0.001) for combined NDDs compared to non-HT users. The RRs for transdermal-HT users were significantly decreased for all-cause dementia (0.73, 0.60-0.88, P = 0.001) and MS (0.55, 0.36-0.84, P = 0.005). Greatest reduction in risk of NDD, AD, and dementia emerged in patients aged 65 years or older. Further, the protective effect of long-term therapy (>1 year) on combined NDDs, AD, PD, and dementia was greater compared to short-term therapy (≤1 year). DISCUSSION HT was associated with reduced risk of all NDDs including AD and dementia, with greater duration of therapy and natural steroid formulations associated with greater efficacy. These findings advance precision HT to prevent NDDs including AD.
Collapse
Affiliation(s)
- Yu Jin Kim
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | - Maira Soto
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Gregory L Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of MedicineMD‐PhD Training ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen Rodgers
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of NeurologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
22
|
Liu M, Shen L, Xu M, Wang DQH, Tso P. Estradiol Enhances Anorectic Effect of Apolipoprotein A-IV through ERα-PI3K Pathway in the Nucleus Tractus Solitarius. Genes (Basel) 2020; 11:E1494. [PMID: 33322656 PMCID: PMC7764025 DOI: 10.3390/genes11121494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 01/09/2023] Open
Abstract
Estradiol (E2) enhances the anorectic action of apolipoprotein A-IV (apoA-IV), however, the intracellular mechanisms are largely unclear. Here we reported that the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway was significantly activated by E2 and apoA-IV, respectively, in primary neuronal cells isolated from rat embryonic brainstem. Importantly, the combination of E2 and apoA-IV at their subthreshold doses synergistically activated the PI3K/Akt signaling pathway. These effects, however, were significantly diminished by the pretreatment with LY294002, a selective PI3K inhibitor. E2-induced activation of the PI3K/Akt pathway was through membrane-associated ERα, because the phosphorylation of Akt was significantly increased by PPT, an ERα agonist, and by E2-BSA (E2 conjugated to bovine serum albumin) which activates estrogen receptor on the membrane. Centrally administered apoA-IV at a low dose (0.5 µg) significantly suppressed food intake and increased the phosphorylation of Akt in the nucleus tractus solitarius (NTS) of ovariectomized (OVX) rats treated with E2, but not in OVX rats treated with vehicle. These effects were blunted by pretreatment with LY294002. These results indicate that E2's regulatory role in apoA-IV's anorectic action is through the ERα-PI3K pathway in the NTS. Manipulation of the PI3K/Akt signaling activation in the NTS may provide a novel therapeutic approach for the prevention and the treatment of obesity-related disorders in females.
Collapse
Affiliation(s)
- Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| | - Ling Shen
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| |
Collapse
|
23
|
Priyanka HP, Nair RS. Neuroimmunomodulation by estrogen in health and disease. AIMS Neurosci 2020; 7:401-417. [PMID: 33263078 PMCID: PMC7701372 DOI: 10.3934/neuroscience.2020025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Systemic homeostasis is maintained by the robust bidirectional regulation of the neuroendocrine-immune network by the active involvement of neural, endocrine and immune mediators. Throughout female reproductive life, gonadal hormones undergo cyclic variations and mediate concomitant modulations of the neuroendocrine-immune network. Dysregulation of the neuroendocrine-immune network occurs during aging as a cumulative effect of declining neural, endocrine and immune functions and loss of compensatory mechanisms including antioxidant enzymes, growth factors and co-factors. This leads to disruption of homeostasis and sets the stage for the development of female-specific age-associated diseases such as autoimmunity, osteoporosis, cardiovascular diseases and hormone-dependent cancers. Ovarian hormones especially estrogen, play a key role in the maintenance of health and homeostasis by modulating the nervous, endocrine and immune functions and thereby altering neuroendocrine-immune homeostasis. Immunologically estrogen's role in the modulation of Th1/Th2 immune functions and contributing to pro-inflammatory conditions and autoimmunity has been widely studied. Centrally, hypothalamic and pituitary hormones influence gonadal hormone secretion in murine models during onset of estrous cycles and are implicated in reproductive aging-associated acyclicity. Loss of estrogen affects neuronal plasticity and the ensuing decline in cognitive functions during reproductive aging in females implicates estrogen in the incidence and progression of neurodegenerative diseases. Peripherally, sympathetic noradrenergic (NA) innervations of lymphoid organs and the presence of both adrenergic (AR) and estrogen receptors (ER) on lymphocytes poise estrogen as a potent neuroimmunomodulator during health and disease. Cyclic variations in estrogen levels throughout reproductive life, perimenopausal surge in estrogen levels followed by its precipitous decline, concomitant with decline in central hypothalamic catecholaminergic activity, peripheral sympathetic NA innervation and associated immunosuppression present an interesting study to explore female-specific age-associated diseases in a new light.
Collapse
Affiliation(s)
- Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai-600002, India
| | | |
Collapse
|
24
|
McCarthy M, Raval AP. The peri-menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 2020; 17:317. [PMID: 33097048 PMCID: PMC7585188 DOI: 10.1186/s12974-020-01998-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
The peri-menopause or menopausal transition—the time period that surrounds the final years of a woman’s reproductive life—is associated with profound reproductive and hormonal changes in a woman’s body and exponentially increases a woman’s risk of cerebral ischemia and Alzheimer’s disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-β). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-β-mediated mechanisms.
Collapse
Affiliation(s)
- Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA. .,Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
25
|
Kirshner ZZ, Yao JK, Li J, Long T, Nelson D, Gibbs RB. Impact of estrogen receptor agonists and model of menopause on enzymes involved in brain metabolism, acetyl-CoA production and cholinergic function. Life Sci 2020; 256:117975. [PMID: 32565251 PMCID: PMC7448522 DOI: 10.1016/j.lfs.2020.117975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022]
Abstract
Our goal is to understand how loss of circulating estrogens and estrogen replacement affect brain physiology and function, particularly in brain regions involved in cognitive processes. We recently conducted a large metabolomics study characterizing the effects of rodent models of menopause and treatment with estrogen receptor (ER) agonists on neurochemical targets in hippocampus, frontal cortex, and striatum. Here we characterize effects on levels of several key enzymes involved in glucose utilization and energy production, specifically phosphofructokinase, glyceraldehyde 3-phosphate dehydrogenase, and pyruvate dehydrogenase. We also evaluated effects on levels of β-actin and α-tubulin, choline acetyltransferase (ChAT) activity, and levels of ATP citrate lyase. All experiments were conducted in young adult rats. Experiment 1 compared the effects of ovariectomy (OVX), a model of surgical menopause, and 4-vinylcyclohexene diepoxide (VCD)-treatments, a model of transitional menopause, with tissues collected at proestrus and at diestrus. Experiment 2 used a separate cohort of rats to evaluate the same targets in OVX and VCD-treated rats treated with estradiol or with selective ER agonists. Differences in the expression of metabolic enzymes between cycling animals and models of surgical and transitional menopause were detected. These differences were model-, region- and time- dependent, and were modulated by selective ER agonists. Collectively, the findings demonstrate that loss of ovarian function and ER agonist treatments have differing effects in OVX vs. VCD-treated rats. Differences may help to explain differences in the effects of estrogen treatments on brain function and cognition in women who have experienced surgical vs. transitional menopause.
Collapse
Affiliation(s)
- Z Z Kirshner
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Jeffrey K Yao
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Junyi Li
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Tao Long
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Doug Nelson
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - R B Gibbs
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
26
|
Torromino G, Maggi A, De Leonibus E. Estrogen-dependent hippocampal wiring as a risk factor for age-related dementia in women. Prog Neurobiol 2020; 197:101895. [PMID: 32781107 DOI: 10.1016/j.pneurobio.2020.101895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.
Collapse
Affiliation(s)
- Giulia Torromino
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy.
| |
Collapse
|
27
|
Barth C, de Lange AMG. Towards an understanding of women's brain aging: the immunology of pregnancy and menopause. Front Neuroendocrinol 2020; 58:100850. [PMID: 32504632 DOI: 10.1016/j.yfrne.2020.100850] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/23/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Women are at significantly greater risk of developing Alzheimer's disease and show higher prevalence of autoimmune conditions relative to men. Women's brain health is historically understudied, and little is therefore known about the mechanisms underlying epidemiological sex differences in neurodegenerative diseases, and how female-specific factors may influence women's brain health across the lifespan. In this review, we summarize recent studies on the immunology of pregnancy and menopause, emphasizing that these major immunoendocrine transition phases may play a critical part in women's brain aging trajectories.
Collapse
Affiliation(s)
- Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Ann-Marie G de Lange
- Department of Psychology, University of Oslo, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.
| |
Collapse
|
28
|
Mariani MM, Mojziszek K, Curley E, Thornton JE. Lowering luteinizing hormone (LH) reverses spatial memory deficits associated with neurotoxin infusion into the hippocampus of ovx rats. Horm Behav 2020; 119:104631. [PMID: 31759942 DOI: 10.1016/j.yhbeh.2019.104631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/24/2019] [Accepted: 10/28/2019] [Indexed: 01/14/2023]
Affiliation(s)
- Monica M Mariani
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA.
| | - Kirsten Mojziszek
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Emily Curley
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| | - Janice E Thornton
- Neuroscience Department, Oberlin College, 119 Woodland Street, Oberlin, OH 44074, USA
| |
Collapse
|
29
|
Wang Y, Mishra A, Brinton RD. Transitions in metabolic and immune systems from pre-menopause to post-menopause: implications for age-associated neurodegenerative diseases. F1000Res 2020; 9. [PMID: 32047612 PMCID: PMC6993821 DOI: 10.12688/f1000research.21599.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
The brain undergoes two aging programs: chronological and endocrinological. This is particularly evident in the female brain, which undergoes programs of aging associated with reproductive competency. Comprehensive understanding of the dynamic metabolic and neuroinflammatory aging process in the female brain can illuminate windows of opportunities to promote healthy brain aging. Bioenergetic crisis and chronic low-grade inflammation are hallmarks of brain aging and menopause and have been implicated as a unifying factor causally connecting genetic risk factors for Alzheimer's disease and other neurodegenerative diseases. In this review, we discuss metabolic phenotypes of pre-menopausal, peri-menopausal, and post-menopausal aging and their consequent impact on the neuroinflammatory profile during each transition state. A critical aspect of the aging process is the dynamic metabolic neuro-inflammatory profiles that emerge during chronological and endocrinological aging. These dynamic systems of biology are relevant to multiple age-associated neurodegenerative diseases and provide a therapeutic framework for prevention and delay of neurodegenerative diseases of aging. While these findings are based on investigations of the female brain, they have a broader fundamental systems of biology strategy for investigating the aging male brain. Molecular characterization of alterations in fuel utilization and neuroinflammatory mechanisms during these neuro-endocrine transition states can inform therapeutic strategies to mitigate the risk of Alzheimer's disease in women. We further discuss a precision hormone replacement therapy approach to target symptom profiles during endocrine and chronological aging to reduce risk for age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
30
|
Guo H, Liu M, Zhang L, Wang L, Hou W, Ma Y, Ma Y. The Critical Period for Neuroprotection by Estrogen Replacement Therapy and the Potential Underlying Mechanisms. Curr Neuropharmacol 2020; 18:485-500. [PMID: 31976839 PMCID: PMC7457406 DOI: 10.2174/1570159x18666200123165652] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/03/2019] [Accepted: 01/14/2020] [Indexed: 01/13/2023] Open
Abstract
17β-Estradiol (estradiol or E2) is a steroid hormone that has been broadly applied as a neuroprotective therapy for a variety of neurodegenerative and cerebrovascular disorders such as ischemic stroke, Alzheimer's disease, and Parkinson's disease. Several laboratory and clinical studies have reported that Estrogen Replacement Therapy (ERT) had no effect against these diseases in elderly postmenopausal women, and at worst, increased their risk of onset and mortality. This review focuses on the growing body of data from in vitro and animal models characterizing the potential underlying mechanisms and signaling pathways that govern successful neuroprotection by ERT, including the roles of E2 receptors in mediating neuroprotection, E2 genomic regulation of apoptosis- related pathways, membrane-bound receptor-mediated non-genomic signaling pathways, and the antioxidant mechanisms of E2. Also discussed is the current evidence for a critical period of effective treatment with estrogen following natural or surgical menopause and the outcomes of E2 administration within an advantageous time period. The known mechanisms governing the duration of the critical period include depletion of E2 receptors, the switch to a ketogenic metabolic profile by neuronal mitochondria, and a decrease in acetylcholine that accompanies E2 deficiency. Also the major clinical trials and observational studies concerning postmenopausal Hormone Therapy (HT) are summarized to compare their outcomes with respect to neurological disease and discuss their relevance to the critical period hypothesis. Finally, potential controversies and future directions for this field are discussed throughout the review.
Collapse
Affiliation(s)
| | | | | | | | | | - Yaqun Ma
- Address correspondence to these authors at the Anesthesia and Operation Center, The First Medical Center to Chinese PLA General Hospital, Beijing 100853, China; Tel: +86 010 66938152; E-mail: and Department of Anesthesiology, The Seventh Medical Center to Chinese PLA General Hospital, Beijing 100700, China; E-mail:
| | - Yulong Ma
- Address correspondence to these authors at the Anesthesia and Operation Center, The First Medical Center to Chinese PLA General Hospital, Beijing 100853, China; Tel: +86 010 66938152; E-mail: and Department of Anesthesiology, The Seventh Medical Center to Chinese PLA General Hospital, Beijing 100700, China; E-mail:
| |
Collapse
|
31
|
Laouafa S, Roussel D, Marcouiller F, Soliz J, Gozal D, Bairam A, Joseph V. Roles of oestradiol receptor alpha and beta against hypertension and brain mitochondrial dysfunction under intermittent hypoxia in female rats. Acta Physiol (Oxf) 2019; 226:e13255. [PMID: 30635990 DOI: 10.1111/apha.13255] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/20/2022]
Abstract
AIM Chronic intermittent hypoxia (CIH) induces systemic (hypertension) and central alterations (mitochondrial dysfunction underlying cognitive deficits). We hypothesized that agonists of oestradiol receptors (ER) α and β prevent CIH-induced hypertension and brain mitochondrial dysfunction. METHODS Ovariectomized female rats were implanted with osmotic pumps delivering vehicle (Veh), the ERα agonist propylpyraoletriol (PPT - 30 μg/kg/day) or the ERβ agonist diarylpropionitril (DPN - 100 μg/kg/day). Animals were exposed to CIH (21%-10% FI O2 - 10 cycles/hour - 8 hours/day - 7 days) or normoxia. Arterial blood pressure was measured after CIH or normoxia exposures. Mitochondrial respiration and H2 O2 production were measured in brain cortex with high-resolution respirometry, as well as activity of complex I and IV of the electron transport chain, citrate synthase, pyruvate, and lactate dehydrogenase (PDH and LDH). RESULTS Propylpyraoletriol but not DPN prevented the rise of arterial pressure induced by CIH. CIH exposures decreased O2 consumption, complex I activity, and increased H2 O2 production. CIH had no effect on citrate synthase activity, but decreased PDH activity and increased LDH activity indicating higher anaerobic glycolysis. Propylpyraoletriol and DPN treatments prevented all these alterations. CONCLUSIONS We conclude that in OVX female rats, the ERα agonist prevents from CIH-induced hypertension while both ERα and ERβ agonists prevent the brain mitochondrial dysfunction and metabolic switch induced by CIH. These findings may have implications for menopausal women suffering of sleep apnoea regarding hormonal therapy.
Collapse
Affiliation(s)
- Sofien Laouafa
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - Damien Roussel
- CNRS, UMR 5023 Université Claude Bernard Lyon 1 Villeurbanne France
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - Jorge Soliz
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - David Gozal
- Department of Child Health University of Missouri School of Medicine Columbia Missouri
| | - Aida Bairam
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| |
Collapse
|
32
|
Kurt AH, Yuksel KZ, Uremis N, Uremis MM, Altun I, Bosnak M, Kilicaslan D, Alli B. Protective Effects of G Protein-Coupled Estrogen Receptor 1 (GPER1) on β-Amyloid-Induced Neurotoxicity: Implications for Alzheimer’s Disease. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419010148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
|
34
|
Abstract
This article reviews the role of endogenous estrogen in neural and cognitive processing, followed by an examination of longitudinal cognitive data captured in various stages of the menopausal transition. The remaining text reviews the contradictory results from major hormone therapy trials to date, evidence for the "timing hypothesis," and closes with recommendations for future research and for practicing clinicians.
Collapse
|
35
|
Nemeth VL, Must A, Horvath S, Király A, Kincses ZT, Vécsei L. Gender-Specific Degeneration of Dementia-Related Subcortical Structures Throughout the Lifespan. J Alzheimers Dis 2018; 55:865-880. [PMID: 27792015 DOI: 10.3233/jad-160812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Age-related changes in brain structure are a question of interest to a broad field of research. Structural decline has been consistently, but not unambiguously, linked to functional consequences, including cognitive impairment and dementia. One of the areas considered of crucial importance throughout this process is the medial temporal lobe, and primarily the hippocampal region. Gender also has a considerable effect on volume deterioration of subcortical grey matter (GM) structures, such as the hippocampus. The influence of age×gender interaction on disproportionate GM volume changes might be mediated by hormonal effects on the brain. Hippocampal volume loss appears to become accelerated in the postmenopausal period. This decline might have significant influences on neuroplasticity in the CA1 region of the hippocampus highly vulnerable to pathological influences. Additionally, menopause has been associated with critical pathobiochemical changes involved in neurodegeneration. The micro- and macrostructural alterations and consequent functional deterioration of critical hippocampal regions might result in clinical cognitive impairment-especially if there already is a decline in the cognitive reserve capacity. Several lines of potential vulnerability factors appear to interact in the menopausal period eventually leading to cognitive decline, mild cognitive impairment, or Alzheimer's disease. This focused review aims to delineate the influence of unmodifiable risk factors of neurodegenerative processes, i.e., age and gender, on critical subcortical GM structures in the light of brain derived estrogen effects. The menopausal period appears to be of key importance for the risk of cognitive decline representing a time of special vulnerability for molecular, structural, and functional influences and offering only a narrow window for potential protective effects.
Collapse
Affiliation(s)
- Viola Luca Nemeth
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Anita Must
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Szatmar Horvath
- Department of Psychiatry, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Andras Király
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zsigmond Tamas Kincses
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
36
|
Rutkowsky JM, Lee LL, Puchowicz M, Golub MS, Befroy DE, Wilson DW, Anderson S, Cline G, Bini J, Borkowski K, Knotts TA, Rutledge JC. Reduced cognitive function, increased blood-brain-barrier transport and inflammatory responses, and altered brain metabolites in LDLr -/-and C57BL/6 mice fed a western diet. PLoS One 2018; 13:e0191909. [PMID: 29444171 PMCID: PMC5812615 DOI: 10.1371/journal.pone.0191909] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/12/2018] [Indexed: 12/20/2022] Open
Abstract
Recent work suggests that diet affects brain metabolism thereby impacting cognitive function. Our objective was to determine if a western diet altered brain metabolism, increased blood-brain barrier (BBB) transport and inflammation, and induced cognitive impairment in C57BL/6 (WT) mice and low-density lipoprotein receptor null (LDLr -/-) mice, a model of hyperlipidemia and cognitive decline. We show that a western diet and LDLr -/- moderately influence cognitive processes as assessed by Y-maze and radial arm water maze. Also, western diet significantly increased BBB transport, as well as microvessel factor VIII in LDLr -/- and microglia IBA1 staining in WT, both indicators of activation and neuroinflammation. Interestingly, LDLr -/- mice had a significant increase in 18F- fluorodeoxyglucose uptake irrespective of diet and brain 1H-magnetic resonance spectroscopy showed increased lactate and lipid moieties. Metabolic assessments of whole mouse brain by GC/MS and LC/MS/MS showed that a western diet altered brain TCA cycle and β-oxidation intermediates, levels of amino acids, and complex lipid levels and elevated proinflammatory lipid mediators. Our study reveals that the western diet has multiple impacts on brain metabolism, physiology, and altered cognitive function that likely manifest via multiple cellular pathways.
Collapse
Affiliation(s)
- Jennifer M. Rutkowsky
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California, United States of America
- * E-mail:
| | - Linda L. Lee
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California, United States of America
| | - Michelle Puchowicz
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mari S. Golub
- Department of Environmental Toxicology, University of California, Davis, California, United States of America
| | - Douglas E. Befroy
- Magnetic Resonance Research Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Dennis W. Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Steven Anderson
- Department of Physiology and Membrane Biology, University of California, Davis, California, United States of America
| | - Gary Cline
- Department of Endocrinology, Yale University, New Haven, Connecticut, United States of America
| | - Jason Bini
- Yale PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut, United States of America
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California, Davis, California, United States of America
| | - Trina A. Knotts
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - John C. Rutledge
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | | |
Collapse
|
37
|
Somayaji MR, Przekwas AJ, Gupta RK. Combination Therapy for Multi-Target Manipulation of Secondary Brain Injury Mechanisms. Curr Neuropharmacol 2018; 16:484-504. [PMID: 28847295 PMCID: PMC6018188 DOI: 10.2174/1570159x15666170828165711] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a major healthcare problem that affects millions of people worldwide. Despite advances in understanding and developing preventative and treatment strategies using preclinical animal models, clinical trials to date have failed, and a 'magic bullet' for effectively treating TBI-induced damage does not exist. Thus, novel pharmacological strategies to effectively manipulate the complex and heterogeneous pathophysiology of secondary injury mechanisms are needed. Given that goal, this paper discusses the relevance and advantages of combination therapies (COMTs) for 'multi-target manipulation' of the secondary injury cascade by administering multiple drugs to achieve an optimal therapeutic window of opportunity (e.g., temporally broad window) and compares these regimens to monotherapies that manipulate a single target with a single drug at a given time. Furthermore, we posit that integrated mechanistic multiscale models that combine primary injury biomechanics, secondary injury mechanobiology/neurobiology, physiology, pharmacology and mathematical programming techniques could account for vast differences in the biological space and time scales and help to accelerate drug development, to optimize pharmacological COMT protocols and to improve treatment outcomes.
Collapse
Affiliation(s)
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
38
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
39
|
Neuroketotherapeutics: A modern review of a century-old therapy. Neurochem Int 2017; 117:114-125. [PMID: 28579059 PMCID: PMC5711637 DOI: 10.1016/j.neuint.2017.05.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/18/2023]
Abstract
Neuroketotherapeutics represent a class of bioenergetic medicine therapies that feature the induction of ketosis. These therapies include medium-chain triglyceride supplements, ketone esters, fasting, strenuous exercise, the modified Atkins diet, and the classic ketogenic diet. Extended experience reveals persons with epilepsy, especially pediatric epilepsy, benefit from ketogenic diets although the mechanisms that underlie its effects remain unclear. Data indicate ketotherapeutics enhance mitochondrial respiration, promote neuronal long-term potentiation, increase BDNF expression, increase GPR signaling, attenuate oxidative stress, reduce inflammation, and alter protein post-translational modifications via lysine acetylation and β-hydroxybutyrylation. These properties have further downstream implications involving Akt, PLCγ, CREB, Sirtuin, and mTORC pathways. Further studies of neuroketotherapeutics will enhance our understanding of ketone body molecular biology, and reveal novel central nervous system therapeutic applications.
Collapse
|
40
|
Worsening of memory deficit induced by energy-dense diet in a rat model of early-Alzheimer's disease is associated to neurotoxic Aβ species and independent of neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2017; 1863:731-743. [DOI: 10.1016/j.bbadis.2016.12.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/07/2016] [Accepted: 12/23/2016] [Indexed: 01/20/2023]
|
41
|
Manshack LK, Conard CM, Bryan SJ, Deem SL, Holliday DK, Bivens NJ, Givan SA, Rosenfeld CS. Transcriptomic alterations in the brain of painted turtles ( Chrysemys picta) developmentally exposed to bisphenol A or ethinyl estradiol. Physiol Genomics 2017; 49:201-215. [PMID: 28159858 DOI: 10.1152/physiolgenomics.00103.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 12/25/2022] Open
Abstract
Developmental exposure of turtles and other reptiles to endocrine-disrupting chemicals (EDCs), including bisphenol A (BPA) and ethinyl estradiol (EE), can stimulate partial to full gonadal sex-reversal in males. We have also recently shown that in ovo exposure to either EDC can induce similar sex-dependent behavioral changes typified by improved spatial learning and memory or possibly feminized brain responses. Observed behavioral changes are presumed to be due to BPA- and EE-induced brain transcriptomic alterations during development. To test this hypothesis, we treated painted turtles (Chrysemys picta) at developmental stage 17, incubated at 26°C (male-inducing temperature), with 1) BPA (1 ng/µl), 2) EE (4 ng/µl), or 3) vehicle ethanol (control group). Ten months after hatching and completion of the behavioral tests, juvenile turtles were euthanized, brains were collected and frozen in liquid nitrogen, and RNA was isolated for RNA-Seq analysis. Turtles exposed to BPA clustered separately from EE-exposed and control individuals. More transcripts and gene pathways were altered in BPA vs. EE individuals. The one transcript upregulated in both BPA- and EE-exposed individuals was the mitochondrial-associated gene, ND5, which is involved in oxidative phosphorylation. Early exposure of turtles to BPA increases transcripts linked with ribosomal and mitochondrial functions, especially bioenergetics, which has been previously linked with improved cognitive performance. In summary, even though both BPA and EE resulted in similar behavioral alterations, they diverge in the pattern of neural transcript alterations with early BPA significantly upregulating several genes involved in oxidative phosphorylation, mitochondrial activity, and ribosomal function, which could enhance cognitive performance.
Collapse
Affiliation(s)
- Lindsey K Manshack
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Caroline M Conard
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Sara J Bryan
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri
| | - Sharon L Deem
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri.,Saint Louis Zoo Institute for Conservation Medicine, St. Louis, Missouri
| | - Dawn K Holliday
- Pathology and Anatomical Sciences, School of Medicine, University of Missouri, Columbia, Missouri.,Department of Biology and Environmental Sciences, Westminster College, Fulton, Missouri
| | - Nathan J Bivens
- DNA Core Facility, University of Missouri, Columbia, Missouri
| | - Scott A Givan
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,Informatics Research Core Facility, University of Missouri, Columbia, Missouri.,Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri
| | - Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri; .,Biomedical Sciences, University of Missouri, Columbia, Missouri.,Genetics Area Program, University of Missouri, Columbia, Missouri; and.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri
| |
Collapse
|
42
|
Imtiaz B, Taipale H, Tanskanen A, Tiihonen M, Kivipelto M, Heikkinen AM, Tiihonen J, Soininen H, Hartikainen S, Tolppanen AM. Risk of Alzheimer's disease among users of postmenopausal hormone therapy: A nationwide case-control study. Maturitas 2017; 98:7-13. [PMID: 28274328 DOI: 10.1016/j.maturitas.2017.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To examine the association between postmenopausal hormone therapy (HT) and Alzheimer's disease (AD). METHODS Medicine and Alzheimer's disease (MEDALZ) is a nested case-control study of the entire Finnish population with clinically verified AD from 2005 to 2011 and up to 4 matched controls per case. This study comprises 230,580 women (46,117 cases and 184,463 controls). Data on HT use from 1995 to 2011 was extracted from the national prescription register using following ATC codes: G03C (estrogen), G03D (progestogen) and G03F (estrogen and progestogen in combination). Only systemic HT (oral or transdermal) was considered. RESULTS Use of systemic estrogen and progestogen was associated with an increased risk of AD, with ORs (95% CI) of 1.10 (1.06-1.12) and 1.13 (1.10-1.17) respectively, but use of systemic estrogen HT for >10years (OR, 95% CI: 0.91, 0.84-0.99) was protective against AD. Long-term (>10years) use of progestogen and combination HT was not related to AD risk (OR, 95% CI: 1.0, 0.90-1.2). CONCLUSION Our findings do not suggest HT is an important determinant of AD risk.
Collapse
Affiliation(s)
- Bushra Imtiaz
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.
| | - Heidi Taipale
- Kuopio Research Center of Geriatric Care, University of Eastern Finland, Kuopio, Finland; Research Center for Comparative Effectiveness and Patient Safety (RECEPS) and School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Antti Tanskanen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Miia Tiihonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Miia Kivipelto
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland; Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | - Jari Tiihonen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland; Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Sirpa Hartikainen
- Kuopio Research Center of Geriatric Care, University of Eastern Finland, Kuopio, Finland; School of Pharmacy, Social Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Anna-Maija Tolppanen
- Research Center for Comparative Effectiveness and Patient Safety (RECEPS) and School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
43
|
Crider A, Pillai A. Estrogen Signaling as a Therapeutic Target in Neurodevelopmental Disorders. J Pharmacol Exp Ther 2017; 360:48-58. [PMID: 27789681 PMCID: PMC5193073 DOI: 10.1124/jpet.116.237412] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022] Open
Abstract
Estrogens, the primary female sex hormones, were originally characterized through their important role in sexual maturation and reproduction. However, recent studies have shown that estrogens play critical roles in a number of brain functions, including cognition, learning and memory, neurodevelopment, and adult neuroplasticity. A number of studies from both clinical as well as preclinical research suggest a protective role of estrogen in neurodevelopmental disorders including autism spectrum disorder (ASD) and schizophrenia. Alterations in the levels of estrogen receptors have been found in subjects with ASD or schizophrenia, and adjunctive estrogen therapy has been shown to be effective in enhancing the treatment of schizophrenia. This review summarizes the findings on the role of estrogen in the pathophysiology of neurodevelopmental disorders with a focus on ASD and schizophrenia. We also discuss the potential of estrogen as a therapeutic target in the above disorders.
Collapse
Affiliation(s)
- Amanda Crider
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Anilkumar Pillai
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
44
|
Uchoa MF, Moser VA, Pike CJ. Interactions between inflammation, sex steroids, and Alzheimer's disease risk factors. Front Neuroendocrinol 2016; 43:60-82. [PMID: 27651175 PMCID: PMC5123957 DOI: 10.1016/j.yfrne.2016.09.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/10/2016] [Accepted: 09/14/2016] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder for which there are no effective strategies to prevent or slow its progression. Because AD is multifactorial, recent research has focused on understanding interactions among the numerous risk factors and mechanisms underlying the disease. One mechanism through which several risk factors may be acting is inflammation. AD is characterized by chronic inflammation that is observed before clinical onset of dementia. Several genetic and environmental risk factors for AD increase inflammation, including apolipoprotein E4, obesity, and air pollution. Additionally, sex steroid hormones appear to contribute to AD risk, with age-related losses of estrogens in women and androgens in men associated with increased risk. Importantly, sex steroid hormones have anti-inflammatory actions and can interact with several other AD risk factors. This review examines the individual and interactive roles of inflammation and sex steroid hormones in AD, as well as their relationships with the AD risk factors apolipoprotein E4, obesity, and air pollution.
Collapse
Affiliation(s)
- Mariana F Uchoa
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - V Alexandra Moser
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Christian J Pike
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
45
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
46
|
Tskitishvili E, Pequeux C, Munaut C, Viellevoye R, Nisolle M, Noël A, Foidart JM. Use of estetrol with other steroids for attenuation of neonatal hypoxic-ischemic brain injury: to combine or not to combine? Oncotarget 2016; 7:33722-43. [PMID: 27231853 PMCID: PMC5085115 DOI: 10.18632/oncotarget.9591] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Estetrol (E4), estradiol (E2) and progesterone (P4) have important antioxidative and neuroprotective effects in neuronal system. We aimed to study the consequence of combined steroid therapy in neonatal hypoxic-ischemic encephalopathy (HIE). In vitro the effect of E4 combined with other steroids on oxidative stress and the cell viability in primary hippocampal cultures was evaluated by lactate dehydrogenase and cell survival assays. In vivo neuroprotective and therapeutic efficacy of E4 combined with other steroids was studied in HIE model of immature rats. The rat pups rectal temperature, body and brain weights were evaluated.The hippocampus and the cortex were investigated by histo/immunohistochemistry: intact cell number counting, expressions of markers for early gray matter lose, neuro- and angiogenesis were studied. Glial fibrillary acidic protein was evaluated by ELISA in blood samples. In vitro E4 and combinations of high doses of E4 with P4 and/or E2 significantly diminished the LDH activity and upregulated the cell survival.In vivopretreatment or treatment by different combinations of E4 with other steroids had unalike effects on body and brain weight, neuro- and angiogenesis, and GFAP expression in blood. The combined use of E4 with other steroids has no benefit over the single use of E4.
Collapse
Affiliation(s)
- Ekaterine Tskitishvili
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Christel Pequeux
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Carine Munaut
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Renaud Viellevoye
- Neonatal Intensive Care Unit, Department of Pediatrics, University of Liege, Liege 1, Belgium
| | - Michelle Nisolle
- Department of Obstetrics and Gynecology, University of Liege, Liege1, Belgium
| | - Agnes Noël
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Jean-Michel Foidart
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| |
Collapse
|
47
|
Pratap UP, Patil A, Sharma HR, Hima L, Chockalingam R, Hariharan MM, Shitoot S, Priyanka HP, ThyagaRajan S. Estrogen-induced neuroprotective and anti-inflammatory effects are dependent on the brain areas of middle-aged female rats. Brain Res Bull 2016; 124:238-53. [PMID: 27242078 DOI: 10.1016/j.brainresbull.2016.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Reproductive aging in females is characterized by fluctuations and precipitous decline in estrogen levels, which may lead to reduction in cognitive function and age-associated neurodegenerative disorders. The nature of estrogen-mediated neuronal plasticity is unknown during reproductive aging. We hypothesize that estrogen treatment of early middle-aged ovariectomized rats may exert specific effects in the brain by modulating signaling pathways regulating metabolic enzymes, inflammatory markers, antioxidant status, cholinergic function and survival signals. PURPOSE To investigate the mechanisms of estrogen-induced effects on neuroprotection and neuroinflammation through the involvement of intracellular signaling pathways in brain areas of ovariectomized (OVX) middle-aged (MA) female rats. METHODS Ovariectomized early MA female Sprague-Dawley rats (n=8/group) were implanted with 17β-estradiol (E2) 30-day release pellets (0.6μg and 300μg). At the end of the treatment period, frontal cortex (FC), striatum (STR), medial basal hypothalamus (MBH), and hippocampus (HP) were isolated and examined for the expression of tyrosine hydroxylase (p-TH), nerve growth factor (NGF), p-NF-κB (p50 and p65)and p-ERK, p-CREB, p-Akt, and activities of cholinesterases and antioxidant enzymes, key regulatory enzymes of metabolic pathways, and nitric oxide production. RESULTS E2 enhanced p-TH expression in FC and HP, reduced NGF expression in HP, and suppressed p-NF-κB expression in FC and STR. It also increased the expression of molecular markers (p-ERK, p-CREB and p-Akt), and nitric oxide production in various brain areas, while differentially regulating the activities of metabolic enzymes and cholinesterases. CONCLUSION Estrogen modulates the neural and inflammatory factors, and intracellular markers depending on the brain areas that may influence differential remodeling of neuronal circuitry which can be used to develop therapeutic strategies in cognitive impairment and neurodegenerative disorders in aging.
Collapse
Affiliation(s)
- Uday P Pratap
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Anushree Patil
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Himanshu R Sharma
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Lalgi Hima
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Ramanathan Chockalingam
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Murali M Hariharan
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Sushrut Shitoot
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Hannah P Priyanka
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India
| | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
48
|
Vaňková M, Hill M, Velíková M, Včelák J, Vacínová G, Dvořáková K, Lukášová P, Vejražková D, Rusina R, Holmerová I, Jarolímová E, Vaňková H, Kancheva R, Bendlová B, Stárka L. Preliminary evidence of altered steroidogenesis in women with Alzheimer's disease: Have the patients "OLDER" adrenal zona reticularis? J Steroid Biochem Mol Biol 2016; 158:157-177. [PMID: 26704533 DOI: 10.1016/j.jsbmb.2015.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) represents more than half of total dementias. Various factors including altered steroid biosynthesis may participate in its pathophysiology. We investigated how the circulating steroids (measured by GC-MS and RIA) may be altered in the presence of AD. Sixteen women with AD and 22 age- and BMI-corresponding controls aged over 65 years were enrolled in the study. The steroid levels (47 steroids and steroid polar conjugates) and their ratios in AD female patients indicated increased CYP11A1 activity, weakened activity of the CYP17A1C17,20 lyase metabolic step and attenuated sulfotransferase SULT2A1 activity at higher activity of the CYP17A1 17-hydroxylase step. The patients showed diminished HSD3B2 activity for C21 steroids, abated conversion of 17-hydroxyprogesterone to cortisol, and significantly elevated cortisol. The women with AD had also attenuated steroid 7α-hydroxylation forming immunoprotective Δ(5)-C19 steroids, attenuated aromatase activity forming estradiol that induces autoimmunity and a shift from the 3β-hydroxy-5α/β-reduced C19 steroids to their neuroinhibitory and antiinflammatory GABAergic 3α-hydroxy- counterparts and showed higher levels of the 3α-hydroxy-5α/β-reduced C21 steroids and pregnenolone sulfate (improves cognitive abilities but may be both protective and excitotoxic). Our preliminary data indicated functioning of alternative "backdoor" pathway in women with AD showing higher levels of both 5α/β-reduced C21 steroids but reduced levels of both 5α/β-reduced C21 steroids, which implied that the alternative "backdoor" pathway might include both 5α- and 5β-reduced steroids. Our study suggested relationships between AD status in women based on the age of subjects and levels of 10 steroids measured by GC-MS.
Collapse
Affiliation(s)
- Markéta Vaňková
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Martin Hill
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Marta Velíková
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Josef Včelák
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Gabriela Vacínová
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | | | - Petra Lukášová
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | | | - Robert Rusina
- Department of Neurology, Thomayer's Hospital, Vídeňská 800, Prague 140 59, Czech Republic.
| | - Iva Holmerová
- Faculty of Humanities, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic.
| | - Eva Jarolímová
- Faculty of Humanities, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic.
| | - Hana Vaňková
- Faculty of Humanities, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic; Third Faculty of Medicine, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic.
| | - Radmila Kancheva
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Běla Bendlová
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Luboslav Stárka
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| |
Collapse
|
49
|
Klosinski LP, Yao J, Yin F, Fonteh AN, Harrington MG, Christensen TA, Trushina E, Brinton RD. White Matter Lipids as a Ketogenic Fuel Supply in Aging Female Brain: Implications for Alzheimer's Disease. EBioMedicine 2015; 2:1888-904. [PMID: 26844268 PMCID: PMC4703712 DOI: 10.1016/j.ebiom.2015.11.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/24/2015] [Accepted: 11/02/2015] [Indexed: 01/28/2023] Open
Abstract
White matter degeneration is a pathological hallmark of neurodegenerative diseases including Alzheimer's. Age remains the greatest risk factor for Alzheimer's and the prevalence of age-related late onset Alzheimer's is greatest in females. We investigated mechanisms underlying white matter degeneration in an animal model consistent with the sex at greatest Alzheimer's risk. Results of these analyses demonstrated decline in mitochondrial respiration, increased mitochondrial hydrogen peroxide production and cytosolic-phospholipase-A2 sphingomyelinase pathway activation during female brain aging. Electron microscopic and lipidomic analyses confirmed myelin degeneration. An increase in fatty acids and mitochondrial fatty acid metabolism machinery was coincident with a rise in brain ketone bodies and decline in plasma ketone bodies. This mechanistic pathway and its chronologically phased activation, links mitochondrial dysfunction early in aging with later age development of white matter degeneration. The catabolism of myelin lipids to generate ketone bodies can be viewed as a systems level adaptive response to address brain fuel and energy demand. Elucidation of the initiating factors and the mechanistic pathway leading to white matter catabolism in the aging female brain provides potential therapeutic targets to prevent and treat demyelinating diseases such as Alzheimer's and multiple sclerosis. Targeting stages of disease and associated mechanisms will be critical. Mitochondrial dysfunction activates mechanisms for catabolism of myelin lipids to generate ketone bodies for ATP production. Mechanisms leading to ketone body driven energy production in brain coincide with stages of reproductive aging in females. Sequential activation of myelin catabolism pathway during aging provides multiple therapeutic targets and windows of efficacy.
The mechanisms underlying white matter degeneration, a hallmark of multiple neurodegenerative diseases including Alzheimer's, remain unclear. Herein we provide a mechanistic pathway, spanning multiple transitions of aging, that links mitochondrial dysfunction early in aging with later age white matter degeneration. Catabolism of myelin lipids to generate ketone bodies can be viewed as an adaptive survival response to address brain fuel and energy demand. Women are at greatest risk of late-onset-AD, thus, our analyses in female brain address mechanisms of AD pathology and therapeutic targets to prevent, delay and treat AD in the sex most affected with potential relevance to men.
Collapse
Key Words
- ABAD, Aβ-binding alcohol dehydrogenase
- ABAD, Aβ-binding-alcohol-dehydrogenase
- ACER3, alkaline ceramidase
- AD, Alzheimer's disease
- APO-ε4, apolipoprotein ε4
- APP, amyloid precursor protein
- Aging oxidative stress
- Alzheimer's disease
- BACE1, beta-secretase 1
- BBB, blood brain barrier
- CC, corpus callosum
- CMRglu, cerebral glucose metabolic rate
- COX, complex IV cytochrome c oxidase
- CPT1, carnitine palmitoyltransferase 1
- Cldn11, claudin 11
- Cyp2j6, arachidonic acid epoxygenase
- Cytosolic phospholipase A2
- DHA, docosahexaesnoic acid
- Erbb3, Erb-B2 receptor tyrosine kinase 3
- FDG-PET, 2-[18F]fluoro-2-deoxy-d-glucose
- GFAP, glial fibrillary acidic protein
- H2O2, hydrogen peroxide
- HADHA, hydroxyacyl-CoA dehydrogenase
- HK, hexokinase
- Ketone bodies
- LC MS, liquid chromatography mass spectrometer
- MAG, myelin associated glycoprotein
- MBP, myelin basic protein
- MCT1, monocarboxylate transporter 1
- MIB, mitochondrial isolation buffer
- MOG, myelin oligodendrocyte glycoprotein
- MTL, medial temporal lobe
- Mitochondria
- NEFA, nonesterified fatty acids
- Neurodegeneration
- OCR, oxygen consumption rate
- Olig2, oligodendrocyte transcription factor
- PB, phosphate buffer
- PCC, posterior cingulate
- PCR, polymerase chain reaction
- PDH, pyruvate dehydrogenase
- PEI, polyethyleneimine
- RCR, respiratory control ratio
- ROS, reactive oxygen species
- S1P, sphingosine
- TLDA, TaqMan low density array
- WM, white matter
- WT, wild type
- White matter
- cPLA2, cytosolic phospholipase A2
Collapse
Affiliation(s)
- Lauren P Klosinski
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, 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
| | - Fei Yin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Eugenia Trushina
- Department of Neurology, Mayo Clinic Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA; 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
|
50
|
Yin F, Sancheti H, Liu Z, Cadenas E. Mitochondrial function in ageing: coordination with signalling and transcriptional pathways. J Physiol 2015; 594:2025-42. [PMID: 26293414 DOI: 10.1113/jp270541] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/13/2015] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction entailing decreased energy-transducing capacity and perturbed redox homeostasis is an early and sometimes initiating event in ageing and age-related disorders involving tissues with high metabolic rate such as brain, liver and heart. In the central nervous system (CNS), recent findings from our and other groups suggest that the mitochondrion-centred hypometabolism is a key feature of ageing brains and Alzheimer's disease. This hypometabolic state is manifested by lowered neuronal glucose uptake, metabolic shift in the astrocytes, and alternations in mitochondrial tricarboxylic acid cycle function. Similarly, in liver and adipose tissue, mitochondrial capacity around glucose and fatty acid metabolism and thermogenesis is found to decline with age and is implicated in age-related metabolic disorders such as obesity and type 2 diabetes mellitus. These mitochondrion-related disorders in peripheral tissues can impact on brain functions through metabolic, hormonal and inflammatory signals. At the cellular level, studies in CNS and non-CNS tissues support the notion that instead of being viewed as autonomous organelles, mitochondria are part of a dynamic network with close interactions with other cellular components through energy- or redox-sensitive cytosolic kinase signalling and transcriptional pathways. Hence, it would be critical to further understand the molecular mechanisms involved in the communication between mitochondria and the rest of the cell. Therapeutic strategies that effectively preserves or improve mitochondrial function by targeting key component of these signalling cascades could represent a novel direction for numerous mitochondrion-implicated, age-related disorders.
Collapse
Affiliation(s)
- Fei Yin
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Harsh Sancheti
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Zhigang Liu
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Enrique Cadenas
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| |
Collapse
|