1
|
Chapman NH, Navas PA, Dorschner MO, Mehaffey M, Wigg KG, Price KM, Naumova OY, Kerr EN, Guger SL, Lovett MW, Grigorenko EL, Berninger V, Barr CL, Wijsman EM, Raskind WH. Targeted analysis of dyslexia-associated regions on chromosomes 6, 12 and 15 in large multigenerational cohorts. PLoS One 2025; 20:e0324006. [PMID: 40424442 PMCID: PMC12112411 DOI: 10.1371/journal.pone.0324006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
Dyslexia is a common learning impairment with a genetic basis that affects word reading and spelling. An increasing list of loci and genes have been implicated, but analyses to-date have investigated only limited genomic variation within each locus with no confirmed pathogenic variants identified. Our study is the first to comprehensively sequence both coding and cis-acting regulatory regions of such genes in a large study sample. In a collection of >2000 participants in families from three independent sites, we performed targeted capture and comprehensive sequencing of all exons and some regulatory elements of five candidate risk genes (DNAAF4, CYP19A1, DCDC2, KIAA0319 and GRIN2B) for which prior evidence for a role in dyslexia exists from more than one sample. We evaluated evidence for association in each of six dyslexia-related quantitative phenotypes (traits) using both individual common single nucleotide polymorphisms and aggregated rare variants. We detected no promoter alterations and few deleterious variants in the coding exons, none of which showed evidence of association with any trait. Single variant and aggregate testing of DNAAF4 failed to detect significant evidence of association with any of the traits. The other four genes provided evidence of association with one or more traits. A common variant downstream of CYP19A1 showed significant evidence of association with multiple traits with or without verbal IQ (VIQ) adjustment. A haplotype that stretches from the downstream region of KIAA0319 to the second intron of DCDC2 was associated with reduced performance on timed real word reading. Finally, rare exonic variants in GRIN2B were associated with performance on spelling, with or without adjustment for VIQ. Our findings from this large-scale sequencing study complement those from genome-wide association studies, argue against the causative involvement of large-effect coding variants in these five candidate genes, support a multigenic etiology, and suggest a role of transcriptional regulation.
Collapse
Affiliation(s)
- Nicola H. Chapman
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Patrick A. Navas
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Michael O. Dorschner
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Michele Mehaffey
- Department of Pediatrics, Division of Pediatric Genetics, Pediatric Genetics, University of Washington, Seattle, Washington, United States of America
| | - Karen G. Wigg
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kaitlyn M. Price
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Oxana Y. Naumova
- Department of Psychology, University of Houston, Houston, Texas, United States of America
| | - Elizabeth N. Kerr
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Sharon L. Guger
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maureen W. Lovett
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Elena L. Grigorenko
- Department of Psychology, University of Houston, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States of America
| | - Virginia Berninger
- Department of Educational Psychology, University of Washington, Seattle, Washington, United States of America
| | - Cathy L. Barr
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Educational Psychology, University of Washington, Seattle, Washington, United States of America
- Division of Experimental and Translational Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Ellen M. Wijsman
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Wendy H. Raskind
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
2
|
Demetriou A, Lindqvist B, Ali HG, Shamekh MM, Varshney M, Inzunza J, Maioli S, Nilsson P, Nalvarte I. ERβ mediates sex-specific protection in the App-NL-G-F mouse model of Alzheimer's disease. Biol Sex Differ 2025; 16:29. [PMID: 40302001 PMCID: PMC12039102 DOI: 10.1186/s13293-025-00711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Menopausal loss of neuroprotective estrogen is thought to contribute to the sex differences in Alzheimer's disease (AD). Activation of estrogen receptor beta (ERβ) can be clinically relevant since it avoids the adverse systemic effects of ERα activation. However, very few studies have explored ERβ-mediated neuroprotection in AD, and no information on its contribution to the sex differences in AD exists. In the present study, we specifically explored the role of ERβ in mediating sex-specific protection against AD pathology in the AppNL-G-F knock-in mouse model of amyloidosis, and if surgical menopause (ovariectomy) modulates pathology in this model. METHODS We treated male and female AppNL-G-F knock-in mice with the clinically relevant and selective ERβ agonist LY500307. A subset of the females was ovariectomized prior to treatment. Y-maze and contextual fear conditioning tests were used to assess memory performance, and biochemical assays such as qPCR, immunohistochemistry, Western blot, and multiplex immunoassays, were used to evaluate amyloid pathology. RESULTS We found that Female AppNL-G-F mice had higher soluble Aβ levels in cortex and hippocampus than males and more activated microglia. ERβ activation protected against amyloid pathology and cognitive decline in both male and female AppNL-G-F mice. Although ovariectomy increased soluble amyloid beta (Aβ) in cortex and insoluble Aβ in hippocampus, as well as sustained neuroinflammation after ERβ activation, it had otherwise limited effects on pathology. We further identified that ERβ did not alter APP processing, but rather exerted its protection at least partly via microglia activation in a sex-specific manner. CONCLUSION Combined, we provide new understanding to the sex differences in AD by demonstrating that ERβ protects against AD pathology differently in males and females, warranting reassessment of ERβ in combating AD.
Collapse
Affiliation(s)
- Aphrodite Demetriou
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
| | - Birgitta Lindqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Heba G Ali
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mohamed M Shamekh
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
| | - Ivan Nalvarte
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden.
| |
Collapse
|
3
|
Piao YR, Li MR, Sun MZ, Liu Y, Chen CY, Chu CP, Todo Y, Tang Z, Wang CY, Jin WZ, Qiu DL. Estradiol Enhances Cerebellar Molecular Layer Interneuron-Purkinje Cell Synaptic Transmission and Improves Motor Learning Through ER-β in Vivo in Mice. CEREBELLUM (LONDON, ENGLAND) 2025; 24:51. [PMID: 39979512 DOI: 10.1007/s12311-025-01805-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
In the cerebellar cortex, 17β-estradiol (E2) binds to estrogen receptors (ERs) and plays a role in regulating cerebellar synaptic plasticity and motor learning behaviors. However, the underlying mechanisms remain unclear. In this study, we investigated the effects of E2 on synaptic transmission between cerebellar molecular layer interneurons (MLIs) and Purkinje cells (PCs) in urethane-anesthetized mice. Using in vivo cell-attached and whole-cell recordings combined with immunohistochemistry, we examined MLI-PC synaptic responses elicited by facial air-puff stimulation. Cell-attached recordings from PCs demonstrated that air-puff stimulation of the ipsilateral whisker pad elicited MLI-PC synaptic currents (P1), which were significantly enhanced by local micro-application of E2 to the cerebellar molecular layer. The E2-induced potentiation of P1 amplitude exhibited dose dependency, with a 50% effective concentration (EC50) of 30 nM. The effects of E2 on amplitude of P1 and pause of simple spike firing were completely prevented by blockade of ERs or ERβ, but not by blockade of ERα or a G-protein coupled receptor (GPER). Application of a selective ERβ agonist mimicked and overwhelmed the E2-induced enhancement of the MLI-PC synaptic transmission. Whole-cell recording with biocytin staining showing that E2 does not change the spontaneous and the evoked spike firing properties of basket-type MLIs. Rotarod test indicated that microinjection of E2 onto the cerebellar surface significantly promotes initial motor learning ability, which is abolished by blockade of ERβ. ERβ immunoreactivity was expressed in the ML and PC layer, especially around the PC somata in the mouse cerebellar cortex. These results indicate that E2 binds to ERβ, resulting in an enhance in the cerebellar MLI-PC synaptic transmission and an improvement of initial motor learning ability in vivo in mice.
Collapse
Affiliation(s)
- Yong-Rui Piao
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province, 133002, China
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China
| | - Mei-Rui Li
- Faculty of Electrical, Information and Communication Engineering, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Ming-Ze Sun
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, Jilin Province, 132013, China
| | - Yang Liu
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, Jilin Province, 132013, China
| | - Chao-Yue Chen
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, Jilin Province, 132013, China
| | - Chun-Ping Chu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province, 133002, China
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, Jilin Province, 132013, China
| | - Yuki Todo
- Faculty of Electrical, Information and Communication Engineering, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Zheng Tang
- School of Computer Engineering and Science, Shanghai University, Shanghai, 200444, China
| | - Chun-Yan Wang
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, Jilin Province, 132013, China
| | - Wen-Zhe Jin
- Department of Pain, Affiliated Hospital of Yanbian University, 1325, JuZi Street, Yanji City, Jilin Province, 133000, China.
| | - De-Lai Qiu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji City, Jilin Province, 133002, China.
- Brain Science Institute, Jilin Medical University, Jilin City, Jilin Province, 132013, China.
- Department of Physiology, College of Basic Medicine, Jilin Medical University, Jilin City, Jilin Province, 132013, China.
| |
Collapse
|
4
|
Viswanath M, Peter MCS. Thyroid hormones activate TH/E 2 receptor/regulator system and drive Na +/K +-ATPase in the ovarian wall of hypothyroid air-breathing fish (Anabas testudineus Bloch). Gen Comp Endocrinol 2025; 360:114640. [PMID: 39536982 DOI: 10.1016/j.ygcen.2024.114640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 11/09/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
In fish, as in other vertebrates, thyroid hormones (THs) act on many biological processes including growth and reproduction. Primary THs such as thyroxine (T4) and triiodothyronine (T3) are known for their direct action on osmoregulatory organs regulating ion osmotic homeostasis in many teleost fishes. However, it is unclear how these hormones interact with estradiol-17β (E2), an ovarian hormone that regulates the development of oocytes. We thus examined the short-term in vivo action of varied THs such as T4, T3 and T2, a potent TH metabolite diiodothyronine, on the expression pattern of receptors of THs and E2 in the ovarian wall of the hypothyroid climbing perch to identify the interactive pattern of TH/E2 receptor system and the molecular dynamics of Na+/K+-ATPase (NKA) subunits in the ovarian wall that provides structural and functional support to ovary. We found differential pattern of transcript abundance of NKA subunits isoforms such as nkaα1a, nkaα1b, nkaα1c atp1b1, atp1b2 and fxyd3, fxyd5, fxyd6, TH receptor isoforms (tr<, trβ, tr
Collapse
Affiliation(s)
- Meenu Viswanath
- Inter-University Centre for Evolutionary and Integrative Biology-iCEIB, School of Life Sciences, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Department of Zoology, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India
| | - M C Subhash Peter
- Inter-University Centre for Evolutionary and Integrative Biology-iCEIB, School of Life Sciences, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Department of Zoology, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Sastrajeevan Integrative Bioresearch and Education-SIBE, F17-Gandhipuram, Sreekariyam, Thiruvananthapuram 695017, Kerala, India.
| |
Collapse
|
5
|
Martínez-Martos JM, Cantón-Habas V, Rich-Ruíz M, Reyes-Medina MJ, Ramírez-Expósito MJ, Carrera-González MDP. Sexual and Metabolic Differences in Hippocampal Evolution: Alzheimer's Disease Implications. Life (Basel) 2024; 14:1547. [PMID: 39768255 PMCID: PMC11677427 DOI: 10.3390/life14121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Sex differences in brain metabolism and their relationship to neurodegenerative diseases like Alzheimer's are an important emerging topic in neuroscience. Intrinsic anatomic and metabolic differences related to male and female physiology have been described, underscoring the importance of considering biological sex in studying brain metabolism and associated pathologies. The hippocampus is a key structure exhibiting sex differences in volume and connectivity. Adult neurogenesis in the dentate gyrus, dendritic spine density, and electrophysiological plasticity contribute to the hippocampus' remarkable plasticity. Glucose transporters GLUT3 and GLUT4 are expressed in human hippocampal neurons, with proper glucose metabolism being crucial for learning and memory. Sex hormones play a major role, with the aromatase enzyme that generates estradiol increasing in neurons and astrocytes as an endogenous neuroprotective mechanism. Inhibition of aromatase increases gliosis and neurodegeneration after brain injury. Genetic variants of aromatase may confer higher Alzheimer's risk. Estrogen replacement therapy in postmenopausal women prevents hippocampal hypometabolism and preserves memory. Insulin is also a key regulator of hippocampal glucose metabolism and cognitive processes. Dysregulation of the insulin-sensitive glucose transporter GLUT4 may explain the comorbidity between type II diabetes and Alzheimer's. GLUT4 colocalizes with the insulin-regulated aminopeptidase IRAP in neuronal vesicles, suggesting an activity-dependent glucose uptake mechanism. Sex differences in brain metabolism are an important factor in understanding neurodegenerative diseases, and future research must elucidate the underlying mechanisms and potential therapeutic implications of these differences.
Collapse
Affiliation(s)
- José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
| | - Vanesa Cantón-Habas
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| | - Manuel Rich-Ruíz
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| | - María José Reyes-Medina
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, University of Córdoba, 14004 Córdoba, Spain; (V.C.-H.); (M.R.-R.); (M.J.R.-M.)
| | - María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
| | - María del Pilar Carrera-González
- Experimental and Clinical Physiopathology Research Group CTS-1039, Department of Health Sciences, Faculty of Health Sciences, University of Jaen, Las Lagunillas University Campus, 23009 Jaen, Spain; (J.M.M.-M.); (M.J.R.-E.)
- Maimonides Institute of Biomedical Research of Córdoba (IMIBIC) IMIBIC Building, Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14004 Cordoba, Spain
| |
Collapse
|
6
|
Scuto M, Majzúnová M, Torcitto G, Antonuzzo S, Rampulla F, Di Fatta E, Trovato Salinaro A. Functional Food Nutrients, Redox Resilience Signaling and Neurosteroids for Brain Health. Int J Mol Sci 2024; 25:12155. [PMID: 39596221 PMCID: PMC11594618 DOI: 10.3390/ijms252212155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
The interplay between functional food nutrients and neurosteroids has garnered significant attention for its potential to enhance stress resilience in health and/or disease. Several bioactive nutrients, including medicinal herbs, flavonoids, and bioavailable polyphenol-combined nanoparticles, as well as probiotics, vitamin D and omega-3 fatty acids, have been shown to improve blood-brain barrier (BBB) dysfunction, endogenous neurosteroid homeostasis and brain function. These nutrients can inhibit oxidative stress and neuroinflammation, which are linked to the pathogenesis of various neurological disorders. Interestingly, flavonoids exhibit dose-dependent effects, activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway at the physiological/low dose (neurohormesis). This leads to the upregulation of antioxidant phase II genes and proteins such as heme oxygenase-1 (HO-1) and sirtuin-1 (Sirt1), which are activated by curcumin and resveratrol, respectively. These adaptive neuronal response mechanisms help protect against reactive oxygen species (ROS) and neurotoxicity. Impaired Nrf2 and neurosteroid hormone signaling in the brain can exacerbate selective vulnerability to neuroinflammatory conditions, contributing to the onset and progression of neurodegenerative and psychiatric disorders, including Alzheimer's disease, anxiety and depression and other neurological disorders, due to the vulnerability of neurons to stress. This review focuses on functional food nutrients targeting Nrf2 antioxidant pathway and redox resilience genes to regulate the neurosteroid homeostasis and BBB damage associated with altered GABAergic neurotransmission. By exploring the underlying molecular mechanisms using innovative technologies, we aim to develop promising neuroprotective strategies and personalized nutritional and neuroregenerative therapies to prevent or attenuate oxidative stress and neuroinflammation, ultimately promoting brain health.
Collapse
Affiliation(s)
- Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | - Miroslava Majzúnová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 84215 Bratislava, Slovakia;
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovakia
| | - Gessica Torcitto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | - Silvia Antonuzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | - Francesco Rampulla
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| | | | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (G.T.); (S.A.); (F.R.)
| |
Collapse
|
7
|
Cioffi L, Diviccaro S, Chrostek G, Caruso D, Garcia-Segura LM, Melcangi RC, Giatti S. Neuroactive steroids fluctuate with regional specificity in the central and peripheral nervous system across the rat estrous cycle. J Steroid Biochem Mol Biol 2024; 243:106590. [PMID: 39053702 DOI: 10.1016/j.jsbmb.2024.106590] [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: 06/25/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Neuroactive steroids (i.e., sex steroid hormones and neurosteroids) are important physiological regulators of nervous function and potential neuroprotective agents for neurodegenerative and psychiatric disorders. Sex is an important component of such effects. However, even if fluctuations in sex steroid hormone level during the menstrual cycle are associated with neuropathological events in some women, the neuroactive steroid pattern in the brain across the ovarian cycle has been poorly explored. Therefore, we assessed the levels of pregnenolone, progesterone, and its metabolites (i.e., dihydroprogesterone, allopregnanolone and isoallopregnanolone), dehydroepiandrosterone, testosterone and its metabolites (i.e., dihydrotestosterone, 3α-diol and 17β-estradiol) across the rat ovarian cycle to determine whether their plasma fluctuations are similar to those occurring in the central (i.e., hippocampus and cerebral cortex) and peripheral (i.e., sciatic nerve) nervous system. Data obtained indicate that the plasma pattern of these molecules generally does not fully reflect the events occurring in the nervous system. In addition, for some neuroactive steroid levels, the pattern is not identical between the two brain regions and between the brain and peripheral nerves. Indeed, with the exception of progesterone, all other neuroactive steroids assessed here showed peculiar regional differences in their pattern of fluctuation in the nervous system during the estrous cycle. These observations may have important diagnostic and therapeutic consequences for neuropathological events influenced by the menstrual cycle.
Collapse
Affiliation(s)
- Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Gabriela Chrostek
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Luis Miguel Garcia-Segura
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain and Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy.
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| |
Collapse
|
8
|
Demetriou A, Lindqvist B, Ali HG, Shamekh MM, Maioli S, Inzunza J, Varshney M, Nilsson P, Nalvarte I. ERβ mediates sex-specific protection in the App-NL-G-F mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604543. [PMID: 39091856 PMCID: PMC11291054 DOI: 10.1101/2024.07.22.604543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Menopausal loss of neuroprotective estrogen is thought to contribute to the sex differences in Alzheimer's disease (AD). Activation of estrogen receptor beta (ERβ) can be clinically relevant since it avoids the negative systemic effects of ERα activation. However, very few studies have explored ERβ-mediated neuroprotection in AD, and no information on its contribution to the sex differences in AD exists. In the present study we specifically explored the role of ERβ in mediating sex-specific protection against AD pathology in the clinically relevant App NL-G-F knock-in mouse model of amyloidosis, and if surgical menopause (ovariectomy) modulates pathology in this model. We treated male and female App NL-G-F mice with the selective ERβ agonist LY500307 and subset of the females was ovariectomized prior to treatment. Memory performance was assessed and a battery of biochemical assays were used to evaluate amyloid pathology and neuroinflammation. Primary microglial cultures from male and female wild-type and ERβ-knockout mice were used to assess ERβ's effect on microglial activation and phagocytosis. We find that ERβ activation protects against amyloid pathology and cognitive decline in male and female App NL-G-F mice. Ovariectomy increased soluble amyloid beta (Aβ) in cortex and insoluble Aβ in hippocampus, but had otherwise limited effects on pathology. We further identify that ERβ does not alter APP processing, but rather exerts its protection through amyloid scavenging that at least in part is mediated via microglia in a sex-specific manner. Combined, we provide new understanding to the sex differences in AD by demonstrating that ERβ protects against AD pathology differently in males and females, warranting reassessment of ERβ in combating AD.
Collapse
Affiliation(s)
- Aphrodite Demetriou
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Birgitta Lindqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Heba G. Ali
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Mohamed M. Shamekh
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| |
Collapse
|
9
|
De Michele G, Maione L, Cocozza S, Tranfa M, Pane C, Galatolo D, De Rosa A, De Michele G, Saccà F, Filla A. Ataxia and Hypogonadism: a Review of the Associated Genes and Syndromes. CEREBELLUM (LONDON, ENGLAND) 2024; 23:688-701. [PMID: 36997834 DOI: 10.1007/s12311-023-01549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
The association of hypogonadism and cerebellar ataxia was first recognized in 1908 by Gordon Holmes. Since the seminal description, several heterogeneous phenotypes have been reported, differing for age at onset, associated features, and gonadotropins levels. In the last decade, the genetic bases of these disorders are being progressively uncovered. Here, we review the diseases associating ataxia and hypogonadism and the corresponding causative genes. In the first part of this study, we focus on clinical syndromes and genes (RNF216, STUB1, PNPLA6, AARS2, SIL1, SETX) predominantly associated with ataxia and hypogonadism as cardinal features. In the second part, we mention clinical syndromes and genes (POLR3A, CLPP, ERAL1, HARS, HSD17B4, LARS2, TWNK, POLG, ATM, WFS1, PMM2, FMR1) linked to complex phenotypes that include, among other features, ataxia and hypogonadism. We propose a diagnostic algorithm for patients with ataxia and hypogonadism, and we discuss the possible common etiopathogenetic mechanisms.
Collapse
Affiliation(s)
- Giovanna De Michele
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy.
| | - Luigi Maione
- Department of Endocrinology and Reproductive Diseases, Paris-Saclay University, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicetre, Paris, France
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Mario Tranfa
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Chiara Pane
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Daniele Galatolo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Stella Maris, Pisa, Italy
| | - Anna De Rosa
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Giuseppe De Michele
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Francesco Saccà
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Alessandro Filla
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Via Sergio Pansini 5, 80131, Naples, Italy
| |
Collapse
|
10
|
Torgerson C, Ahmadi H, Choupan J, Fan CC, Blosnich JR, Herting MM. Sex, gender diversity, and brain structure in early adolescence. Hum Brain Mapp 2024; 45:e26671. [PMID: 38590252 PMCID: PMC11002534 DOI: 10.1002/hbm.26671] [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: 07/28/2023] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
There remains little consensus about the relationship between sex and brain structure, particularly in early adolescence. Moreover, few pediatric neuroimaging studies have analyzed both sex and gender as variables of interest-many of which included small sample sizes and relied on binary definitions of gender. The current study examined gender diversity with a continuous felt-gender score and categorized sex based on X and Y allele frequency in a large sample of children ages 9-11 years old (N = 7195). Then, a statistical model-building approach was employed to determine whether gender diversity and sex independently or jointly relate to brain morphology, including subcortical volume, cortical thickness, gyrification, and white matter microstructure. Additional sensitivity analyses found that male versus female differences in gyrification and white matter were largely accounted for by total brain volume, rather than sex per se. The model with sex, but not gender diversity, was the best-fitting model in 60.1% of gray matter regions and 61.9% of white matter regions after adjusting for brain volume. The proportion of variance accounted for by sex was negligible to small in all cases. While models including felt-gender explained a greater amount of variance in a few regions, the felt-gender score alone was not a significant predictor on its own for any white or gray matter regions examined. Overall, these findings demonstrate that at ages 9-11 years old, sex accounts for a small proportion of variance in brain structure, while gender diversity is not directly associated with neurostructural diversity.
Collapse
Affiliation(s)
- Carinna Torgerson
- Department of Population and Public Health SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Mark and Mary Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Hedyeh Ahmadi
- Department of Population and Public Health SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jeiran Choupan
- Mark and Mary Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Chun Chieh Fan
- Center for Population Neuroscience and GeneticsLaureate Institute for Brain ResearchTulsaOklahomaUSA
- Department of Radiology, School of MedicineUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - John R. Blosnich
- Suzanne Dworak‐Peck School of Social WorkUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Megan M. Herting
- Department of Population and Public Health SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
11
|
Fernández-Vargas M, Macedo-Lima M, Remage-Healey L. Acute Aromatase Inhibition Impairs Neural and Behavioral Auditory Scene Analysis in Zebra Finches. eNeuro 2024; 11:ENEURO.0423-23.2024. [PMID: 38467426 PMCID: PMC10960633 DOI: 10.1523/eneuro.0423-23.2024] [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: 10/18/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 03/13/2024] Open
Abstract
Auditory perception can be significantly disrupted by noise. To discriminate sounds from noise, auditory scene analysis (ASA) extracts the functionally relevant sounds from acoustic input. The zebra finch communicates in noisy environments. Neurons in their secondary auditory pallial cortex (caudomedial nidopallium, NCM) can encode song from background chorus, or scenes, and this capacity may aid behavioral ASA. Furthermore, song processing is modulated by the rapid synthesis of neuroestrogens when hearing conspecific song. To examine whether neuroestrogens support neural and behavioral ASA in both sexes, we retrodialyzed fadrozole (aromatase inhibitor, FAD) and recorded in vivo awake extracellular NCM responses to songs and scenes. We found that FAD affected neural encoding of songs by decreasing responsiveness and timing reliability in inhibitory (narrow-spiking), but not in excitatory (broad-spiking) neurons. Congruently, FAD decreased neural encoding of songs in scenes for both cell types, particularly in females. Behaviorally, we trained birds using operant conditioning and tested their ability to detect songs in scenes after administering FAD orally or injected bilaterally into NCM. Oral FAD increased response bias and decreased correct rejections in females, but not in males. FAD in NCM did not affect performance. Thus, FAD in the NCM impaired neuronal ASA but that did not lead to behavioral disruption suggesting the existence of resilience or compensatory responses. Moreover, impaired performance after systemic FAD suggests involvement of other aromatase-rich networks outside the auditory pathway in ASA. This work highlights how transient estrogen synthesis disruption can modulate higher-order processing in an animal model of vocal communication.
Collapse
Affiliation(s)
- Marcela Fernández-Vargas
- Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Matheus Macedo-Lima
- Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Luke Remage-Healey
- Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
12
|
Kalakh S, Mouihate A. The Effects of Neuroactive Steroids on Myelin in Health and Disease. Med Princ Pract 2024; 33:198-214. [PMID: 38350432 PMCID: PMC11175611 DOI: 10.1159/000537794] [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: 11/07/2023] [Accepted: 02/12/2024] [Indexed: 02/15/2024] Open
Abstract
Myelin plays a pivotal role in the efficient transmission of nerve impulses. Disruptions in myelin integrity are associated with numerous neurological disorders, including multiple sclerosis. In the central nervous system (CNS), myelin is formed by oligodendrocytes. Remyelination refers to the re-formation of the damaged myelin sheath by newly formed oligodendrocytes. Steroids have gained attention for their potential modulatory effects on myelin in both health and disease. Steroids are traditionally associated with endocrine functions, but their local synthesis within the nervous system has generated significant interest. The term "neuroactive steroids" refers to steroids that can act on cells of the nervous system. In the healthy state, neuroactive steroids promote myelin formation, maintenance, and repair by enhancing oligodendrocyte differentiation and maturation. In pathological conditions, such as demyelination injury, multiple neuroactive steroids have shown promise in promoting remyelination. Understanding the effects of neuroactive steroids on myelin could lead to novel therapeutic approaches for demyelinating diseases and neurodegenerative disorders. This review highlights the potential therapeutic significance of neuroactive steroids in myelin-related health and diseases. We review the synthesis of steroids by neurons and glial cells and discuss the roles of neuroactive steroids on myelin structure and function in health and disease. We emphasize the potential promyelinating effects of the varying levels of neuroactive steroids during different female physiological states such as the menstrual cycle, pregnancy, lactation, and postmenopause.
Collapse
Affiliation(s)
- Samah Kalakh
- Department of Physiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
- School of Engineering and Computing, American International University, Kuwait City, Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
13
|
Madison FN, Bingman VP, Smulders TV, Lattin CR. A bird's eye view of the hippocampus beyond space: Behavioral, neuroanatomical, and neuroendocrine perspectives. Horm Behav 2024; 157:105451. [PMID: 37977022 DOI: 10.1016/j.yhbeh.2023.105451] [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: 07/06/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Although the hippocampus is one of the most-studied brain regions in mammals, research on the avian hippocampus has been more limited in scope. It is generally agreed that the hippocampus is an ancient feature of the amniote brain, and therefore homologous between the two lineages. Because birds and mammals are evolutionarily not very closely related, any shared anatomy is likely to be crucial for shared functions of their hippocampi. These functions, in turn, are likely to be essential if they have been conserved for over 300 million years. Therefore, research on the avian hippocampus can help us understand how this brain region evolved and how it has changed over evolutionary time. Further, there is a strong research foundation in birds on hippocampal-supported behaviors such as spatial navigation, food caching, and brood parasitism that scientists can build upon to better understand how hippocampal anatomy, network circuitry, endocrinology, and physiology can help control these behaviors. In this review, we summarize our current understanding of the avian hippocampus in spatial cognition as well as in regulating anxiety, approach-avoidance behavior, and stress responses. Although there are still some questions about the exact number of subdivisions in the avian hippocampus and how that might vary in different avian families, there is intriguing evidence that the avian hippocampus might have complementary functional profiles along the rostral-caudal axis similar to the dorsal-ventral axis of the rodent hippocampus, where the rostral/dorsal hippocampus is more involved in cognitive processes like spatial learning and the caudal/ventral hippocampus regulates emotional states, anxiety, and the stress response. Future research should focus on elucidating the cellular and molecular mechanisms - including endocrinological - in the avian hippocampus that underlie behaviors such as spatial navigation, spatial memory, and anxiety-related behaviors, and in so doing, resolve outstanding questions about avian hippocampal function and organization.
Collapse
Affiliation(s)
- Farrah N Madison
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Verner P Bingman
- Department of Psychology, J. P. Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, OH 43403, USA
| | - Tom V Smulders
- Centre for Behaviour and Evolution, School of Psychology, Newcastle University, Newcastle upon Tyne NE2 4DR, UK
| | - Christine R Lattin
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70808, USA.
| |
Collapse
|
14
|
Boyd HM, Frick KM, Kwapis JL. Connecting the Dots: Potential Interactions Between Sex Hormones and the Circadian System During Memory Consolidation. J Biol Rhythms 2023; 38:537-555. [PMID: 37464775 PMCID: PMC10615791 DOI: 10.1177/07487304231184761] [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: 07/20/2023]
Abstract
Both the circadian clock and sex hormone signaling can strongly influence brain function, yet little is known about how these 2 powerful modulatory systems might interact during complex neural processes like memory consolidation. Individually, the molecular components and action of each of these systems have been fairly well-characterized, but there is a fundamental lack of information about how these systems cooperate. In the circadian system, clock genes function as timekeeping molecules that convey time-of-day information on a well-stereotyped cycle that is governed by the suprachiasmatic nucleus. Keeping time is particularly important to synchronize various physiological processes across the brain and body, including those that regulate memory consolidation. Similarly, sex hormones are powerful modulators of memory, with androgens, estrogens, and progestins, all influencing memory consolidation within memory-relevant brain regions like the hippocampus. Despite clear evidence that each system can influence memory individually, exactly how the circadian and hormonal systems might interact to impact memory consolidation remains unclear. Research investigating either sex hormone action or circadian gene function within memory-relevant brain regions has unveiled several notable places in which the two systems could interact to control memory. Here, we bring attention to known interactions between the circadian clock and sex hormone signaling. We then review sex hormone-mediated control of memory consolidation, highlighting potential nodes through which the circadian system might interact during memory formation. We suggest that the bidirectional relationship between these two systems is essential for proper control of memory formation based on an animal's hormonal and circadian state.
Collapse
Affiliation(s)
- Hannah M. Boyd
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin
| | - Janine L. Kwapis
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
15
|
Miguel-Hidalgo JJ. Neuroprotective astroglial response to neural damage and its relevance to affective disorders. EXPLORATION OF NEUROPROTECTIVE THERAPY 2023; 3:328-345. [PMID: 37920189 PMCID: PMC10622120 DOI: 10.37349/ent.2023.00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/03/2023] [Indexed: 11/04/2023]
Abstract
Astrocytes not only support neuronal function with essential roles in synaptic neurotransmission, action potential propagation, metabolic support, or neuroplastic and developmental adaptations. They also respond to damage or dysfunction in surrounding neurons and oligodendrocytes by releasing neurotrophic factors and other molecules that increase the survival of the supported cells or contribute to mechanisms of structural and molecular restoration. The neuroprotective responsiveness of astrocytes is based on their ability to sense signals of degeneration, metabolic jeopardy and structural damage, and on their aptitude to locally deliver specific molecules to remedy threats to the molecular and structural features of their cellular partners. To the extent that neuronal and other glial cell disturbances are known to occur in affective disorders, astrocyte responsiveness to those disturbances may help to better understand the roles astrocytes play in affective disorders. The astrocytic sensing apparatus supporting those responses involves receptors for neurotransmitters, purines, cell adhesion molecules and growth factors. Astrocytes also share with the immune system the capacity of responding to cytokines released upon neuronal damage. In addition, in responses to specific signals astrocytes release unique factors such as clusterin or humanin that have been shown to exert potent neuroprotective effects. Astrocytes integrate the signals above to further deliver structural lipids, removing toxic metabolites, stabilizing the osmotic environment, normalizing neurotransmitters, providing anti-oxidant protection, facilitating synaptogenesis and acting as barriers to contain varied deleterious signals, some of which have been described in brain regions relevant to affective disorders and related animal models. Since various of the injurious signals that activate astrocytes have been implicated in different aspects of the etiopathology of affective disorders, particularly in relation to the diagnosis of depression, potentiating the corresponding astrocyte neuroprotective responses may provide additional opportunities to improve or complement available pharmacological and behavioral therapies for affective disorders.
Collapse
|
16
|
Belluti S, Imbriano C, Casarini L. Nuclear Estrogen Receptors in Prostate Cancer: From Genes to Function. Cancers (Basel) 2023; 15:4653. [PMID: 37760622 PMCID: PMC10526871 DOI: 10.3390/cancers15184653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogens are almost ubiquitous steroid hormones that are essential for development, metabolism, and reproduction. They exert both genomic and non-genomic action through two nuclear receptors (ERα and ERβ), which are transcription factors with disregulated functions and/or expression in pathological processes. In the 1990s, the discovery of an additional membrane estrogen G-protein-coupled receptor augmented the complexity of this picture. Increasing evidence elucidating the specific molecular mechanisms of action and opposing effects of ERα and Erβ was reported in the context of prostate cancer treatment, where these issues are increasingly investigated. Although new approaches improved the efficacy of clinical therapies thanks to the development of new molecules targeting specifically estrogen receptors and used in combination with immunotherapy, more efforts are needed to overcome the main drawbacks, and resistance events will be a challenge in the coming years. This review summarizes the state-of-the-art on ERα and ERβ mechanisms of action in prostate cancer and promising future therapies.
Collapse
Affiliation(s)
- Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| |
Collapse
|
17
|
Reed MB, Handschuh PA, Klöbl M, Konadu ME, Kaufmann U, Hahn A, Kranz GS, Spies M, Lanzenberger R. The influence of sex steroid treatment on insular connectivity in gender dysphoria. Psychoneuroendocrinology 2023; 155:106336. [PMID: 37499299 DOI: 10.1016/j.psyneuen.2023.106336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Sex-specific differences in brain connectivity were found in various neuroimaging studies, though little is known about sex steroid effects on insular functioning. Based on well-characterized sex differences in emotion regulation, interoception and higher-level cognition, gender-dysphoric individuals receiving gender-affirming hormone therapy represent an interesting cohort to investigate how sex hormones might influence insular connectivity and related brain functions. METHODS To analyze the potential effect of sex steroids on insular connectivity at rest, 11 transgender women, 14 transgender men, 20 cisgender women, and 11 cisgender men were recruited. All participants underwent two magnetic resonance imaging sessions involving resting-state acquisitions separated by a median time period of 4.5 months and also completed the Bermond-Vorst alexithymia questionnaire at the initial and final examination. Between scans, transgender subjects received gender-affirming hormone therapy. RESULTS A seed based functional connectivity analysis revealed a significant 2-way interaction effect of group-by-time between right insula, cingulum, left middle frontal gyrus and left angular gyrus. Post-hoc tests demonstrated an increase in connectivity for transgender women when compared to cisgender men. Furthermore, spectral dynamic causal modelling showed reduced effective connectivity from the posterior cingulum and left angular gyrus to the left middle frontal gyrus as well as from the right insula to the left middle frontal gyrus. Alexithymia changes were found after gender-affirming hormone therapy for transgender women in both fantasizing and identifying. CONCLUSION These findings suggest a considerable influence of estrogen administration and androgen suppression on brain networks implicated in interoception, own-body perception and higher-level cognition.
Collapse
Affiliation(s)
- Murray B Reed
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Patricia A Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Manfred Klöbl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Melisande E Konadu
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Ulrike Kaufmann
- Department of Obstetrics and Gynecology, Medical University of Vienna, Austria
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria; Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
| | - Marie Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria.
| |
Collapse
|
18
|
Torgerson C, Ahmadi H, Choupan J, Fan CC, Blosnich JR, Herting MM. Sex, gender diversity, and brain structure in children ages 9 to 11 years old. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551036. [PMID: 37546960 PMCID: PMC10402171 DOI: 10.1101/2023.07.28.551036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
There remains little consensus about the relationship between sex and brain structure, particularly in childhood. Moreover, few pediatric neuroimaging studies have analyzed both sex and gender as variables of interest - many of which included small sample sizes and relied on binary definitions of gender. The current study examined gender diversity with a continuous felt-gender score and categorized sex based on X and Y allele frequency in a large sample of children ages 9-11 years-old (N=7693). Then, a statistical model-building approach was employed to determine whether gender diversity and sex independently or jointly relate to brain morphology, including subcortical volume, cortical thickness, gyrification, and white matter microstructure. The model with sex, but not gender diversity, was the best-fitting model in 75% of gray matter regions and 79% of white matter regions examined. The addition of gender to the sex model explained significantly more variance than sex alone with regard to bilateral cerebellum volume, left precentral cortical thickness, as well as gyrification in the right superior frontal gyrus, right parahippocampal gyrus, and several regions in the left parietal lobe. For mean diffusivity in the left uncinate fasciculus, the model with sex, gender, and their interaction captured the most variance. Nonetheless, the magnitude of variance accounted for by sex was small in all cases and felt-gender score was not a significant predictor on its own for any white or gray matter regions examined. Overall, these findings demonstrate that at ages 9-11 years-old, sex accounts for a small proportion of variance in brain structure, while gender diversity is not directly associated with neurostructural diversity.
Collapse
Affiliation(s)
- Carinna Torgerson
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Hedyeh Ahmadi
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jeiran Choupan
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Chun Chieh Fan
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, OK, USA
- Department of Radiology, School of Medicine, University of California, San Diego
| | - John R. Blosnich
- Suzanne Dworak-Peck School of Social Work, University of Southern California, Los Angeles, CA, USA
| | - Megan M. Herting
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Wang J, Pratap UP, Lu Y, Sareddy GR, Tekmal RR, Vadlamudi RK, Brann DW. Development and Characterization of Inducible Astrocyte-Specific Aromatase Knockout Mice. BIOLOGY 2023; 12:621. [PMID: 37106821 PMCID: PMC10135694 DOI: 10.3390/biology12040621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
17β-estradiol (E2) is produced in the brain as a neurosteroid, in addition to being an endocrine signal in the periphery. The current animal models for studying brain-derived E2 include global and conditional non-inducible knockout mouse models. The aim of this study was to develop a tamoxifen (TMX)-inducible astrocyte-specific aromatase knockout mouse line (GFAP-ARO-iKO mice) to specifically deplete the E2 synthesis enzymes and aromatase in astrocytes after their development in adult mice. The characterization of the GFAP-ARO-iKO mice revealed a specific and robust depletion in the aromatase expressions of their astrocytes and a significant decrease in their hippocampal E2 levels after a GCI. The GFAP-ARO-iKO animals were alive and fertile and had a normal general brain anatomy, with a normal astrocyte shape, intensity, and distribution. In the hippocampus, after a GCI, the GFAP-ARO-iKO animals showed a major deficiency in their reactive astrogliosis, a dramatically increased neuronal loss, and increased microglial activation. These findings indicate that astrocyte-derived E2 (ADE2) regulates the ischemic induction of reactive astrogliosis and microglial activation and is neuroprotective in the ischemic brain. The GFAP-ARO-iKO mouse models thus provide an important new model to help elucidate the roles and functions of ADE2 in the brain.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Rajeshwar R. Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA
| | - Darrell W. Brann
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
20
|
Jensen A, Thériault K, Yilmaz E, Pon E, Davidson PSR. Mental rotation, episodic memory, and executive control: Possible effects of biological sex and oral contraceptive use. Neurobiol Learn Mem 2023; 198:107720. [PMID: 36621560 DOI: 10.1016/j.nlm.2023.107720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/02/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023]
Abstract
Oral contraceptives (OCs) are one of the most common forms of hormonal birth control. A small literature suggests that OC use may affect visuospatial ability, episodic memory, and executive control. However, previous studies have been criticized for small sample sizes and the use of different, single cognitive tests. We investigated the degree to which biological sex and OC use might affect individual mental rotation, episodic memory, and executive control in a large sample of healthy, young adults (N = 155, including 52 OC users, 53 naturally cycling females, and 50 males) tested individually over videoconference. To measure cognition, we used a set of neuropsychological tasks inspired by Glisky and colleagues' two-factor episodic memory and executive control battery, from which two composite scores (based on principal component analysis) were derived for each participant. Our pre-registered analysis revealed a clear female advantage in episodic memory, independent of OC use. In an exploratory analysis, gist memory was elevated in OC users. Interestingly, we found no significant sex-related differences nor effects of OC use on mental rotation or executive control. Duration of OC use was also not related to any of our cognitive measures. These results suggest that the use of combined, monophasic OCs does not lead to many significant changes in cognition in young adults, although young females overall may have better episodic memory than young males. Additional studies, including longitudinal designs and looking in more detail at the menstrual cycle and OC use history, will further clarify the effects of different types of OCs and their duration of use on different aspects of cognition.
Collapse
Affiliation(s)
- Adelaide Jensen
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Canada.
| | - Kim Thériault
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Canada
| | - Ece Yilmaz
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Canada
| | - Ethan Pon
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Canada
| | | |
Collapse
|
21
|
Potential Regulation of miRNA-29 and miRNA-9 by Estrogens in Neurodegenerative Disorders: An Insightful Perspective. Brain Sci 2023; 13:brainsci13020243. [PMID: 36831786 PMCID: PMC9954655 DOI: 10.3390/brainsci13020243] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 02/04/2023] Open
Abstract
Finding a link between a hormone and microRNAs (miRNAs) is of great importance since it enables the adjustment of genetic composition or cellular functions without needing gene-level interventions. The dicer-mediated cleavage of precursor miRNAs is an interface link between miRNA and its regulators; any disruption in this process can affect neurogenesis. Besides, the hormonal regulation of miRNAs can occur at the molecular and cellular levels, both directly, through binding to the promoter elements of miRNAs, and indirectly, via regulation of the signaling effects of the post-transcriptional processing proteins. Estrogenic hormones have many roles in regulating miRNAs in the brain. This review discusses miRNAs, their detailed biogenesis, activities, and both the general and estrogen-dependent regulations. Additionally, we highlight the relationship between miR-29, miR-9, and estrogens in the nervous system. Such a relationship could be a possible etiological route for developing various neurodegenerative disorders.
Collapse
|
22
|
Turek J, Gąsior Ł. Estrogen fluctuations during the menopausal transition are a risk factor for depressive disorders. Pharmacol Rep 2023; 75:32-43. [PMID: 36639604 PMCID: PMC9889489 DOI: 10.1007/s43440-022-00444-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023]
Abstract
Women are significantly more likely to develop depression than men. Fluctuations in the ovarian estrogen hormone levels are closely linked with women's well-being. This narrative review discusses the available knowledge on the role of estrogen in modulating brain function and the correlation between changes in estrogen levels and the development of depression. Equally discussed are the possible mechanisms underlying these effects, including the role of estrogen in modulating brain-derived neurotrophic factor activity, serotonin neurotransmission, as well as the induction of inflammatory response and changes in metabolic activity, are discussed.
Collapse
Affiliation(s)
- Justyna Turek
- Department of Neurobiology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland
| | - Łukasz Gąsior
- Department of Neurobiology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland
| |
Collapse
|
23
|
Altered visual cortex excitability in premenstrual dysphoric disorder: Evidence from magnetoencephalographic gamma oscillations and perceptual suppression. PLoS One 2022; 17:e0279868. [PMID: 36584199 PMCID: PMC9803314 DOI: 10.1371/journal.pone.0279868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
Premenstrual dysphoric disorder (PMDD) is a psychiatric condition characterized by extreme mood shifts during the luteal phase of the menstrual cycle (MC) due to abnormal sensitivity to neurosteroids and unbalanced neural excitation/inhibition (E/I) ratio. We hypothesized that in women with PMDD in the luteal phase, these factors would alter the frequency of magnetoencephalographic visual gamma oscillations, affect modulation of their power by excitatory drive, and decrease perceptual spatial suppression. Women with PMDD and control women were examined twice-during the follicular and luteal phases of their MC. We recorded visual gamma response (GR) while modulating the excitatory drive by increasing the drift rate of the high-contrast grating (static, 'slow', 'medium', and 'fast'). Contrary to our expectations, GR frequency was not affected in women with PMDD in either phase of the MC. GR power suppression, which is normally associated with a switch from the 'optimal' for GR slow drift rate to the medium drift rate, was reduced in women with PMDD and was the only GR parameter that distinguished them from control participants specifically in the luteal phase and predicted severity of their premenstrual symptoms. Over and above the atypical luteal GR suppression, in both phases of the MC women with PMDD had abnormally strong GR facilitation caused by a switch from the 'suboptimal' static to the 'optimal' slow drift rate. Perceptual spatial suppression did not differ between the groups but decreased from the follicular to the luteal phase only in PMDD women. The atypical modulation of GR power suggests that neuronal excitability in the visual cortex is constitutively elevated in PMDD and that this E/I imbalance is further exacerbated during the luteal phase. However, the unaltered GR frequency does not support the hypothesis of inhibitory neuron dysfunction in PMDD.
Collapse
|
24
|
Brain-Derived Estrogen and Neurological Disorders. BIOLOGY 2022; 11:biology11121698. [PMID: 36552208 PMCID: PMC9774965 DOI: 10.3390/biology11121698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Astrocytes and neurons in the male and female brains produce the neurosteroid brain-derived 17β-estradiol (BDE2) from androgen precursors. In this review, we discuss evidence that suggest BDE2 has a role in a number of neurological conditions, such as focal and global cerebral ischemia, traumatic brain injury, excitotoxicity, epilepsy, Alzheimer's disease, and Parkinson's disease. Much of what we have learned about BDE2 in neurological disorders has come from use of aromatase inhibitors and global aromatase knockout mice. Recently, our group developed astrocyte- and neuron-specific aromatase knockout mice, which have helped to clarify the precise functions of astrocyte-derived 17β-estradiol (ADE2) and neuron-derived 17β-estradiol (NDE2) in the brain. The available evidence to date suggests a primarily beneficial role of BDE2 in facilitating neuroprotection, synaptic and cognitive preservation, regulation of reactive astrocyte and microglia activation, and anti-inflammatory effects. Most of these beneficial effects appear to be due to ADE2, which is induced in most neurological disorders, but there is also recent evidence that NDE2 exerts similar beneficial effects. Furthermore, in certain situations, BDE2 may also have deleterious effects, as recent evidence suggests its overproduction in epilepsy contributes to seizure induction. In this review, we examine the current state of this quickly developing topic, as well as possible future studies that may be required to provide continuing growth in the field.
Collapse
|
25
|
Flück CE, Kuiri-Hänninen T, Silvennoinen S, Sankilampi U, Groessl M. The Androgen Metabolome of Preterm Infants Reflects Fetal Adrenal Gland Involution. J Clin Endocrinol Metab 2022; 107:3111-3119. [PMID: 35994776 DOI: 10.1210/clinem/dgac482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT The human adrenal cortex changes with fetal-neonatal transition from the fetal to the adult organ, accompanied by changes in the steroid metabolome. OBJECTIVE As it is unclear how the observed developmental changes differ between preterm and full-term neonates, we investigated whether the involution of the fetal adrenals is following a fixed time course related to postmenstrual age or whether it is triggered by birth. Furthermore, the fetal and postnatal androgen metabolome of preterm infants was characterized in comparison to term babies. METHODS This was a prospective, longitudinal, 2-center study collecting spot urines of preterm and term infants during the first 12 to 18 months of life. Steroid metabolites were measured from spot urines by gas chromatography-mass spectrometry. Data relating were modeled according to established pre- and postnatal pathways. RESULTS Fetal adrenal involution occurs around term-equivalent age in preterm infants and is not triggered by premature birth. Testosterone levels are higher in preterm infants at birth and decline slower until term compared to full-term babies. Dihydrotestosterone levels and the activity of the classic androgen biosynthesis pathway are lower in premature infants as is 5α-reductase activity. No difference was found in the activity of the alternate backdoor pathway for androgen synthesis. CONCLUSION Human adrenal involution follows a strict timing that is not affected by premature birth. By contrast, prematurity is associated with an altered androgen metabolome after birth. Whether this reflects altered androgen biosynthesis in utero remains to be investigated.
Collapse
Affiliation(s)
- Christa E Flück
- Department of Pediatrics, Division of Endocrinology, Diabetology and Metabolism, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Tanja Kuiri-Hänninen
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, 70029 Kuopio, Finland
| | - Sanna Silvennoinen
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, 70029 Kuopio, Finland
| | - Ulla Sankilampi
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, 70029 Kuopio, Finland
| | - Michael Groessl
- Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
26
|
Ou R, Wei Q, Hou Y, Zhang L, Liu K, Lin J, Yang T, Yang J, Jiang Z, Song W, Cao B, Shang H. Reproductive Lifespan and Motor Progression of Parkinson’s Disease. J Clin Med 2022; 11:jcm11206163. [PMID: 36294482 PMCID: PMC9605617 DOI: 10.3390/jcm11206163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Objectives: Estrogen not only plays a key role in the decreased risk of Parkinson’s disease (PD) but also influences its severity. We aimed to explore the effect of the reproductive lifespan on the motor progression of PD female patients in a large prospective cohort study. Methods: A competing risk analysis with a Fine and Gray model on 491 female and 609 male patients with PD was conducted. We regarded the chance of faster motor progression (as measured by the Unified Parkinson’s Disease Rating Scale (UPDRS) III increasing by ≥16 points during follow-up) and the chance of death as competing risks. The reproductive lifespan was regarded as the variable of interest, while faster motor progression was set as the primary outcome. Results: In the multivariable competing risk analysis, the male sex was not significantly associated with faster motor progression (subdistribution hazard ratio (SHR) 0.888, 95% CI 0.652–1.209, p = 0.450), while a shorter reproductive lifespan was associated with faster motor progression in women (SHR 0.964, 95% CI 0.936–0.994, p = 0.019). Sensitivity analysis indicated that a shorter reproductive lifespan was also significantly associated with faster motor progression in the 48 female patients who reported menopause after the onset of PD (SHR 0.156, 95% CI 0.045–0.542, p = 0.003). A linear mixed model also revealed the significant main effects of a short reproductive lifespan on the higher UPDRS III score in PD female patients at the last visit (p = 0.026). Conclusions: Our study indicates that a short reproductive lifespan contributes to faster motor progression in PD female patients, which has important implications for understanding the role of endogenous estrogen exposure in female PD and is beneficial to select appropriate patients in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Huifang Shang
- Correspondence: ; Tel.: +86-18980602127; Fax: +86-028-85423550
| |
Collapse
|
27
|
Strehle LD, Russart KLG, Burch VA, Grant CV, Pyter LM. Ovarian status modulates endocrine and neuroinflammatory responses to a murine mammary tumor. Am J Physiol Regul Integr Comp Physiol 2022; 323:R432-R444. [PMID: 35993563 PMCID: PMC9512114 DOI: 10.1152/ajpregu.00124.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Patients with breast cancer have increased circulating inflammatory markers and mammary tumors increase neuroinflammation in rodent models. Menopausal status is not only important in the context of breast cancer as circulating estrogen influences tumor progression, but also because estrogen is anti-inflammatory and an essential modulator of endocrine function in the brain and body. Here, we manipulated "menopause" status (ovary-intact and ovariectomized) in an estrogen receptor (ER)+ mouse mammary tumor model to determine the extent to which ovarian status modulates: 1) tumor effects on estrogen concentrations and signaling in the brain, 2) tumor effects on estrogen-associated neurobiology and inflammation, and 3) the ability for tumor resection to resolve the effects of a tumor. We hypothesized that reduced circulating estradiol (E2) after an ovariectomy exacerbates tumor-induced peripheral and central inflammation. Notably, we observed ovarian-dependent modulation on tumor-induced peripheral outcomes, including E2-dependent processes and, to a lesser degree, circulating inflammatory markers. In the brain, ovariectomy exacerbated neuroinflammatory markers in select brain regions and modulated E2-related neurobiology due to a tumor and/or resection. Overall, our data suggest that ovarian status has moderate implications for tumor-induced alterations in neuroendocrinology and neuroinflammation and mild effects on peripheral inflammatory outcomes in this murine mammary tumor model.
Collapse
Affiliation(s)
- Lindsay D Strehle
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Kathryn L G Russart
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Valerie A Burch
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Corena V Grant
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, Ohio
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| |
Collapse
|
28
|
Almutlaq RN, Newell-Fugate AE, Evans LC, Fatima H, Gohar EY. Aromatase inhibition increases blood pressure and markers of renal injury in female rats. Am J Physiol Renal Physiol 2022; 323:F349-F360. [PMID: 35900340 PMCID: PMC9423724 DOI: 10.1152/ajprenal.00055.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Aromatase is a monooxygenase that catalyzes the rate-limiting step of estrogen biosynthesis from androgens. Aromatase inhibitors are widely used for the treatment of patients with hormone receptor-positive breast cancer. However, the effects of aromatase inhibitors on cardiovascular and renal health in females are understudied. Given that estrogen is protective against cardiovascular and kidney diseases, we hypothesized that aromatase inhibition elevates blood pressure and induces kidney injury in female Sprague-Dawley rats. Twelve-week-old female rats were implanted with radiotelemetry transmitters to continuously monitor blood pressure. After baseline blood pressure recording, rats were randomly assigned to treatment with the aromatase inhibitor anastrozole (ASZ) or vehicle (Veh) in drinking water. Twenty days after treatment initiation, rats were shifted from a normal-salt (NS) diet to a high-salt (HS) diet for an additional 40 days. Rats were euthanized 60 days after treatment initiation. Body weight increased in both groups over the study period, but the increase was greater in the ASZ-treated group than in the Veh-treated group. Mean arterial pressure increased in ASZ-treated rats during the NS and HS diet phases but remained unchanged in Veh-treated rats. In addition, urinary excretion of albumin and kidney injury marker-1 and plasma urea were increased in response to aromatase inhibition. Furthermore, histological assessment revealed that ASZ treatment increased morphological evidence of renal tubular injury and proximal tubular brush border loss. In conclusion, chronic aromatase inhibition in vivo with ASZ increases blood pressure and markers of renal proximal tubular injury in female Sprague-Dawley rats, suggesting an important role for aromatization in the maintenance cardiovascular and renal health in females.NEW & NOTEWORTHY Aromatase enzyme catalyzes the rate-limiting step in estrogen biosynthesis. Aromatase inhibitors are clinically used for the treatment of patients with breast cancer; however, the impact of inhibiting aromatization on blood pressure and renal function is incompletely understood. The present findings demonstrate that systemic anastrozole treatment increases blood pressure and renal tubular injury markers in female rats fed a high-salt diet, suggesting an important role for aromatization in preserving cardiovascular and renal health in females.
Collapse
Affiliation(s)
- Rawan N Almutlaq
- Cardiorenal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Annie E Newell-Fugate
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Louise C Evans
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Huma Fatima
- Division of Anatomic and Clinical Pathology, Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
29
|
Immunofluorescent Evidence for Nuclear Localization of Aromatase in Astrocytes in the Rat Central Nervous System. Int J Mol Sci 2022; 23:ijms23168946. [PMID: 36012212 PMCID: PMC9408820 DOI: 10.3390/ijms23168946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/22/2022] Open
Abstract
Estrogens regulate a variety of neuroendocrine, reproductive and also non-reproductive brain functions. Estradiol biosynthesis in the central nervous system (CNS) is catalyzed by the enzyme aromatase, which is expressed in several brain regions by neurons, astrocytes and microglia. In this study, we performed a complex fluorescent immunocytochemical analysis which revealed that aromatase is colocalized with the nuclear stain in glial fibrillary acidic protein (GFAP) positive astrocytes in cell cultures. Confocal immunofluorescent Z-stack scanning analysis confirmed the colocalization of aromatase with the nuclear DAPI signal. Nuclear aromatase was also detectable in the S100β positive astrocyte subpopulation. When the nuclear aromatase signal was present, estrogen receptor alpha was also abundant in the nucleus. Immunostaining of frozen brain tissue sections showed that the nuclear colocalization of the enzyme in GFAP-positive astrocytes is also detectable in the adult rat brain. CD11b/c labelled microglial cells express aromatase, but the immunopositive signal was distributed only in the cytoplasm both in the ramified and amoeboid microglial forms. Immunostaining of rat ovarian tissue sections and human granulosa cells revealed that aromatase was present only in the cytoplasm. This novel observation suggests a new unique mechanism in astrocytes that may regulate certain CNS functions via estradiol production.
Collapse
|
30
|
Scarpa GB, Starrett JR, Li GL, Brooks C, Morohashi Y, Yazaki-Sugiyama Y, Remage-Healey L. Estrogens rapidly shape synaptic and intrinsic properties to regulate the temporal precision of songbird auditory neurons. Cereb Cortex 2022; 33:3401-3420. [PMID: 35849820 PMCID: PMC10068288 DOI: 10.1093/cercor/bhac280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 01/14/2023] Open
Abstract
Sensory neurons parse millisecond-variant sound streams like birdsong and speech with exquisite precision. The auditory pallial cortex of vocal learners like humans and songbirds contains an unconventional neuromodulatory system: neuronal expression of the estrogen synthesis enzyme aromatase. Local forebrain neuroestrogens fluctuate when songbirds hear a song, and subsequently modulate bursting, gain, and temporal coding properties of auditory neurons. However, the way neuroestrogens shape intrinsic and synaptic properties of sensory neurons remains unknown. Here, using a combination of whole-cell patch clamp electrophysiology and calcium imaging, we investigate estrogenic neuromodulation of auditory neurons in a region resembling mammalian auditory association cortex. We found that estradiol rapidly enhances the temporal precision of neuronal firing via a membrane-bound G-protein coupled receptor and that estradiol rapidly suppresses inhibitory synaptic currents while sparing excitation. Notably, the rapid suppression of intrinsic excitability by estradiol was predicted by membrane input resistance and was observed in both males and females. These findings were corroborated by analysis of in vivo electrophysiology recordings, in which local estrogen synthesis blockade caused acute disruption of the temporal correlation of song-evoked firing patterns. Therefore, on a modulatory timescale, neuroestrogens alter intrinsic cellular properties and inhibitory neurotransmitter release to regulate the temporal precision of higher-order sensory neurons.
Collapse
Affiliation(s)
- Garrett B Scarpa
- Neuroscience and Behavior, Center for Neuroendocrine Studies, University of Massachusetts, 639 N. Pleasant St., Amherst, MA 01003, United States
| | - Joseph R Starrett
- Neuroscience and Behavior, Center for Neuroendocrine Studies, University of Massachusetts, 639 N. Pleasant St., Amherst, MA 01003, United States
| | - Geng-Lin Li
- Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, 83 Fenyang Rd, Xuhui District, Shanghai 200031, China
| | - Colin Brooks
- Neuroscience and Behavior, Center for Neuroendocrine Studies, University of Massachusetts, 639 N. Pleasant St., Amherst, MA 01003, United States
| | - Yuichi Morohashi
- Okinawa Institute of Science and Technology (OIST) Graduate University, 1919-1 Tancha, Onna, Kunigami District, Okinawa, Japan
| | - Yoko Yazaki-Sugiyama
- Okinawa Institute of Science and Technology (OIST) Graduate University, 1919-1 Tancha, Onna, Kunigami District, Okinawa, Japan
| | - Luke Remage-Healey
- Neuroscience and Behavior, Center for Neuroendocrine Studies, University of Massachusetts, 639 N. Pleasant St., Amherst, MA 01003, United States
| |
Collapse
|
31
|
McDonald BC, Van Dyk K, Deardorff RL, Bailey JN, Zhai W, Carroll JE, Root JC, Ahles TA, Mandelblatt JS, Saykin AJ. Multimodal MRI examination of structural and functional brain changes in older women with breast cancer in the first year of antiestrogen hormonal therapy. Breast Cancer Res Treat 2022; 194:113-126. [PMID: 35476252 PMCID: PMC9255382 DOI: 10.1007/s10549-022-06597-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Cancer patients are concerned about treatment-related cognitive problems. We examined effects of antiestrogen hormonal therapy on brain imaging metrics in older women with breast cancer. METHODS Women aged 60 + treated with hormonal therapy only and matched non-cancer controls (n = 29/group) completed MRI and objective and self-reported cognitive assessment at pre-treatment/enrollment and 12 months later. Gray matter was examined using voxel-based morphometry (VBM), FreeSurfer, and brain age calculations. Functional MRI (fMRI) assessed working memory-related activation. Analyses examined cross-sectional and longitudinal differences and tested associations between brain metrics, cognition, and days on hormonal therapy. RESULTS The cancer group showed regional reductions over 12 months in frontal, temporal, and parietal gray matter on VBM, reduced FreeSurfer cortical thickness in prefrontal, parietal, and insular regions, and increased working memory-related fMRI activation in frontal, cingulate, and visual association cortex. Controls showed only reductions in fusiform gyrus on VBM and FreeSurfer temporal and parietal cortex thickness. Women with breast cancer showed higher estimated brain age and lower regional gray matter volume than controls at both time points. The cancer group showed a trend toward lower performance in attention, processing speed, and executive function at follow-up. There were no significant associations between brain imaging metrics and cognition or days on hormonal therapy. CONCLUSION Older women with breast cancer showed brain changes in the first year of hormonal therapy. Increased brain activation during working memory processing may be a sign of functional compensation for treatment-related structural changes. This hypothesis should be tested in larger samples over longer time periods. CLINICALTRIALS GOV IDENTIFIER NCT03451383.
Collapse
Affiliation(s)
- Brenna C McDonald
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine and Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
| | - Kathleen Van Dyk
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine and UCLA Jonnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA
| | - Rachael L Deardorff
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine and Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Jessica N Bailey
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine and Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Wanting Zhai
- Georgetown University and Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Judith E Carroll
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine and UCLA Jonnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA, USA
| | - James C Root
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tim A Ahles
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeanne S Mandelblatt
- Georgetown University and Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine and Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
32
|
Eroğlu İ, Eroğlu BÇ. Potential role of tryptophan catabolism in cancer-related cognitive impairment. Nutrition 2022; 103-104:111765. [PMID: 35908496 DOI: 10.1016/j.nut.2022.111765] [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: 12/01/2021] [Accepted: 05/31/2022] [Indexed: 12/24/2022]
Abstract
Oncology may be the most rapidly expanding field in medicine, with several innovative diagnostic and therapeutic procedures appearing daily. Advances in oncology have improved the survival rate for patients with cancer and promoting quality of life is now one of the goals in the care of these patients. Patients face a variety of disease- and treatment-related side effects, including anorexia, nausea, vomiting, recurring infections, and sleep difficulties. Cancer-related cognitive impairment (CRCI) is an overlooked clinical condition found in oncologic practice, particularly in patients with breast cancer. Although several potential mechanisms for CRCI have been hypothesized, to our knowledge, the exact mechanism is still unknown. Alterations in the tryptophan kynurenine pathway have been shown to impair cognitive skills in several mental illnesses. However, its possible function in CRCI has yet to be investigated. The aim of this was to examine the possible interactions between tryptophan catabolism and CRCI.
Collapse
Affiliation(s)
- İmdat Eroğlu
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey.
| | - Burcu Çelik Eroğlu
- Hacettepe University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey
| |
Collapse
|
33
|
Gray SL, Soma KK, Duncan KA. Steroid profiling in brain and plasma of adult zebra finches following traumatic brain injury. J Neuroendocrinol 2022; 34:e13151. [PMID: 35608024 DOI: 10.1111/jne.13151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/20/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022]
Abstract
Traumatic brain injury (TBI) is a serious health concern and a leading cause of death. Emerging evidence strongly suggests that steroid hormones (estrogens, androgens, and progesterone) modulate TBI outcomes by regulating inflammation, oxidative stress, free radical production, and extracellular calcium levels. Despite this growing body of evidence on steroid-mediated neuroprotection, very little is known about the local synthesis of these steroids following injury. Here, we examine the effect of TBI on local neurosteroid levels around the site of injury and in plasma in adult male and female zebra finches. Using ultrasensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS), we examined estrogens, androgens, and progesterone in the entopallium and plasma of injured and uninjured animals. Three days after injury, elevated levels of 17β-estradiol (E2 ), estrone (E1 ), and testosterone (T) were detected near injured brain tissue with a corresponding increase in E2 also detected in plasma. Taken together, these results provide further evidence that TBI alters neurosteroid levels and are consistent with studies showing that neurosteroids provide neuroprotection following injury.
Collapse
Affiliation(s)
- Sofia L Gray
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kelli A Duncan
- Department of Biology, Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, New York, USA
| |
Collapse
|
34
|
Identification of Peripheral Blood miRNA Biomarkers in First-Episode Drug-Free Schizophrenia Patients Using Bioinformatics Strategy. Mol Neurobiol 2022; 59:4730-4746. [DOI: 10.1007/s12035-022-02878-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022]
|
35
|
Merii MH, Fardoun MM, El-Asmar K, Khalil MI, Eid A, Dhaini HR. Effect of BPA on CYP450s expression, and nicotine modulation, in fetal rat brain. Neurotoxicol Teratol 2022; 92:107095. [DOI: 10.1016/j.ntt.2022.107095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 10/18/2022]
|
36
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
37
|
Spool JA, Bergan JF, Remage-Healey L. A neural circuit perspective on brain aromatase. Front Neuroendocrinol 2022; 65:100973. [PMID: 34942232 PMCID: PMC9667830 DOI: 10.1016/j.yfrne.2021.100973] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022]
Abstract
This review explores the role of aromatase in the brain as illuminated by a set of conserved network-level connections identified in several vertebrate taxa. Aromatase-expressing neurons are neurochemically heterogeneous but the brain regions in which they are found are highly-conserved across the vertebrate lineage. During development, aromatase neurons have a prominent role in sexual differentiation of the brain and resultant sex differences in behavior and human brain diseases. Drawing on literature primarily from birds and rodents, we delineate brain regions that express aromatase and that are strongly interconnected, and suggest that, in many species, aromatase expression essentially defines the Social Behavior Network. Moreover, in several cases the inputs to and outputs from this core Social Behavior Network also express aromatase. Recent advances in molecular and genetic tools for neuroscience now enable in-depth and taxonomically diverse studies of the function of aromatase at the neural circuit level.
Collapse
Affiliation(s)
- Jeremy A Spool
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Joseph F Bergan
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Luke Remage-Healey
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
38
|
Non-sensory Influences on Auditory Learning and Plasticity. J Assoc Res Otolaryngol 2022; 23:151-166. [PMID: 35235100 PMCID: PMC8964851 DOI: 10.1007/s10162-022-00837-3] [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: 08/10/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022] Open
Abstract
Distinguishing between regular and irregular heartbeats, conversing with speakers of different accents, and tuning a guitar-all rely on some form of auditory learning. What drives these experience-dependent changes? A growing body of evidence suggests an important role for non-sensory influences, including reward, task engagement, and social or linguistic context. This review is a collection of contributions that highlight how these non-sensory factors shape auditory plasticity and learning at the molecular, physiological, and behavioral level. We begin by presenting evidence that reward signals from the dopaminergic midbrain act on cortico-subcortical networks to shape sound-evoked responses of auditory cortical neurons, facilitate auditory category learning, and modulate the long-term storage of new words and their meanings. We then discuss the role of task engagement in auditory perceptual learning and suggest that plasticity in top-down cortical networks mediates learning-related improvements in auditory cortical and perceptual sensitivity. Finally, we present data that illustrates how social experience impacts sound-evoked activity in the auditory midbrain and forebrain and how the linguistic environment rapidly shapes speech perception. These findings, which are derived from both human and animal models, suggest that non-sensory influences are important regulators of auditory learning and plasticity and are often implemented by shared neural substrates. Application of these principles could improve clinical training strategies and inform the development of treatments that enhance auditory learning in individuals with communication disorders.
Collapse
|
39
|
Brann DW, Lu Y, Wang J, Sareddy GR, Pratap UP, Zhang Q, Tekmal RR, Vadlamudi RK. Neuron-Derived Estrogen-A Key Neuromodulator in Synaptic Function and Memory. Int J Mol Sci 2021; 22:ijms222413242. [PMID: 34948039 PMCID: PMC8706511 DOI: 10.3390/ijms222413242] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 01/31/2023] Open
Abstract
In addition to being a steroid hormone, 17β-estradiol (E2) is also a neurosteroid produced in neurons in various regions of the brain of many species, including humans. Neuron-derived E2 (NDE2) is synthesized from androgen precursors via the action of the biosynthetic enzyme aromatase, which is located at synapses and in presynaptic terminals in neurons in both the male and female brain. In this review, we discuss evidence supporting a key role for NDE2 as a neuromodulator that regulates synaptic plasticity and memory. Evidence supporting an important neuromodulatory role of NDE2 in the brain has come from studies using aromatase inhibitors, aromatase overexpression in neurons, global aromatase knockout mice, and the recent development of conditional forebrain neuron-specific knockout mice. Collectively, these studies demonstrate a key role of NDE2 in the regulation of synapse and spine density, efficacy of excitatory synaptic transmission and long-term potentiation, and regulation of hippocampal-dependent recognition memory, spatial reference memory, and contextual fear memory. NDE2 is suggested to achieve these effects through estrogen receptor-mediated regulation of rapid kinase signaling and CREB-BDNF signaling pathways, which regulate actin remodeling, as well as transcription, translation, and transport of synaptic proteins critical for synaptic plasticity and function.
Collapse
Affiliation(s)
- Darrell W. Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Correspondence:
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health, Shreveport, LA 71103, USA;
| | - Rajeshwar R. Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (G.R.S.); (U.P.P.); (R.R.T.); (R.K.V.)
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| |
Collapse
|
40
|
Azcoitia I, Mendez P, Garcia-Segura LM. Aromatase in the Human Brain. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:189-202. [PMID: 35024691 PMCID: PMC8744447 DOI: 10.1089/andro.2021.0007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 11/30/2022]
Abstract
The aromatase cytochrome P450 (P450arom) enzyme, or estrogen synthase, which is coded by the CYP19A1 gene, is widely expressed in a subpopulation of excitatory and inhibitory neurons, astrocytes, and other cell types in the human brain. Experimental studies in laboratory animals indicate a prominent role of brain aromatization of androgens to estrogens in regulating different brain functions. However, the consequences of aromatase expression in the human brain remain poorly understood. Here, we summarize the current knowledge about aromatase expression in the human brain, abundant in the thalamus, amygdala, hypothalamus, cortex, and hippocampus and discuss its role in the regulation of sensory integration, body homeostasis, social behavior, cognition, language, and integrative functions. Since brain aromatase is affected by neurodegenerative conditions and may participate in sex-specific manifestations of autism spectrum disorders, major depressive disorder, multiple sclerosis, stroke, and Alzheimer's disease, we discuss future avenues for research and potential clinical and therapeutic implications of the expression of aromatase in the human brain.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Mendez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Luis M. Garcia-Segura
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
41
|
Brann DW, Lu Y, Wang J, Zhang Q, Thakkar R, Sareddy GR, Pratap UP, Tekmal RR, Vadlamudi RK. Brain-derived estrogen and neural function. Neurosci Biobehav Rev 2021; 132:793-817. [PMID: 34823913 PMCID: PMC8816863 DOI: 10.1016/j.neubiorev.2021.11.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/26/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023]
Abstract
Although classically known as an endocrine signal produced by the ovary, 17β-estradiol (E2) is also a neurosteroid produced in neurons and astrocytes in the brain of many different species. In this review, we provide a comprehensive overview of the localization, regulation, sex differences, and physiological/pathological roles of brain-derived E2 (BDE2). Much of what we know regarding the functional roles of BDE2 has come from studies using specific inhibitors of the E2 synthesis enzyme, aromatase, as well as the recent development of conditional forebrain neuron-specific and astrocyte-specific aromatase knockout mouse models. The evidence from these studies support a critical role for neuron-derived E2 (NDE2) in the regulation of synaptic plasticity, memory, socio-sexual behavior, sexual differentiation, reproduction, injury-induced reactive gliosis, and neuroprotection. Furthermore, we review evidence that astrocyte-derived E2 (ADE2) is induced following brain injury/ischemia, and plays a key role in reactive gliosis, neuroprotection, and cognitive preservation. Finally, we conclude by discussing the key controversies and challenges in this area, as well as potential future directions for the field.
Collapse
Affiliation(s)
- Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Roshni Thakkar
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA; Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
42
|
Ibrahim MMH, Bheemanapally K, Sylvester PW, Briski KP. Norepinephrine Regulation of Adrenergic Receptor Expression, 5' AMP-Activated Protein Kinase Activity, and Glycogen Metabolism and Mass in Male Versus Female Hypothalamic Primary Astrocyte Cultures. ASN Neuro 2021; 12:1759091420974134. [PMID: 33176438 PMCID: PMC7672765 DOI: 10.1177/1759091420974134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Norepinephrine (NE) control of hypothalamic gluco-regulation involves astrocyte-derived energy fuel supply. In male rats, exogenous NE regulates astrocyte glycogen metabolic enzyme expression in vivo through 5’-AMP-activated protein kinase (AMPK)-dependent mechanisms. Current research utilized a rat hypothalamic astrocyte primary culture model to investigate the premise that NE imposes sex-specific direct control of AMPK activity and glycogen mass and metabolism in these glia. In male rats, NE down-regulation of pAMPK correlates with decreased CaMMKB and increased PP1 expression, whereas noradrenergic augmentation of female astrocyte pAMPK may not involve these upstream regulators. NE concentration is a critical determinant of control of hypothalamic astrocyte glycogen enzyme expression, but efficacy varies between sexes. Data show sex variations in glycogen synthase expression and glycogen phosphorylase-brain and –muscle type dose-responsiveness to NE. Narrow dose-dependent NE augmentation of astrocyte glycogen content during energy homeostasis infers that NE maintains, over a broad exposure range, constancy of glycogen content despite possible changes in turnover. In male rats, beta1- and beta2-adrenergic receptor (AR) profiles displayed bi-directional responses to increasing NE doses; female astrocytes exhibited diminished beta1-AR content at low dose exposure, but enhanced beta2-AR expression at high NE dosages. Thus, graded variations in noradrenergic stimulation may modulate astrocyte receptivity to NE in vivo. Sex dimorphic NE regulation of hypothalamic astrocyte AMPK activation and glycogen metabolism may be mediated, in part, by one or more ARs characterized here by divergent sensitivity to this transmitter.
Collapse
Affiliation(s)
- Mostafa M H Ibrahim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| | - Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| | - Paul W Sylvester
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, United States
| |
Collapse
|
43
|
Ruhnau J, Hübner S, Sunny D, Ittermann T, Hartmann MF, De Lafollie J, Wudy SA, Heckmann M. Impact of Gestational and Postmenstrual Age on Excretion of Fetal Zone Steroids in Preterm Infants Determined by Gas Chromatography-Mass Spectrometry. J Clin Endocrinol Metab 2021; 106:e3725-e3738. [PMID: 33822093 DOI: 10.1210/clinem/dgab194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Fetal zone steroids (FZSs) are excreted in high concentrations in preterm infants. Experimental data suggest protective effects of FZSs in models of neonatal disease. OBJECTIVE We aimed to characterize the postnatal FZS metabolome of well preterm and term infants. METHODS Twenty-four-hour urinary FZS excretion rates were determined in early preterm (<30 weeks' gestation), preterm (30-36 weeks), and term (>37 weeks) infants. Pregnenolone and 17-OH-pregnenolone metabolites (n = 5), and dehydroepiandrosterone sulfate and metabolites (n = 12) were measured by gas chromatography mass spectrometry. Postnatal concentrations of FZSs were compared with already published prenatal concentrations in amniotic fluid. RESULTS Excretion rates of total FZSs and most of the single metabolites were highest in early preterm infants. In this group, excretion rates approach those of term infants at term equivalent postmenstrual age. Preterm infants of 30-36 weeks had more than half lower median excretion rates of FZSs than early preterm infants at the same time of postmenstrual age. Postnatal concentrations of FZSs were partly more than 100-fold higher in all gestational age groups than prenatal concentrations in amniotic fluid at midgestation. CONCLUSION The excretion rates of FZSs as a proxy of the involution of the fetal zone of the most immature preterm infants approached those of term infants at term equivalent. In contrast, the fetal zone in more mature preterm infants undergoes more rapid involution. These data in exclusively well neonates can serve as a basis to investigate the effects of illness on the FZS metabolome in future studies.
Collapse
Affiliation(s)
- Johanna Ruhnau
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Stephanie Hübner
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| | - Donna Sunny
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Michaela F Hartmann
- Paediatric Endocrinology & Diabetology, Laboratory for Translational Hormone Analytics, Steroid Research & Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Jan De Lafollie
- Department of General Pediatrics and Neonatology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Paediatric Endocrinology & Diabetology, Laboratory for Translational Hormone Analytics, Steroid Research & Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
- Department of General Pediatrics and Neonatology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Matthias Heckmann
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| |
Collapse
|
44
|
Endocrine Therapy With or Without CDK4/6 Inhibitors in Women With Hormone-receptor Positive Breast Cancer: What do we Know About the Effects on Cognition? Clin Breast Cancer 2021; 22:191-199. [PMID: 34556423 DOI: 10.1016/j.clbc.2021.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/01/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
Adjuvant endocrine therapy (ET) is the cornerstone of treatment for hormone-receptor positive breast cancer. Recently, ET is increasingly combined with "cyclin-dependent kinases 4 and 6'' (CDK4/6) inhibitors. Given the importance of estrogens in neural processes and the role of cyclin D in hippocampal cell proliferation, it is plausible that these therapies affect cognition, but studies on these potential cognitive effects are sparse. In this review, we summarize existing knowledge on the cognitive effects of ET and CDK4/6 inhibitors in pre-, peri- and postmenopausal patients with breast cancer. We show that several clinical studies support adverse cognitive effects, especially on verbal memory, after ET-induced decrease of estrogen-levels or inactivation of estrogen-receptors. Clinical studies on the cognitive effects of CDK4/6 inhibitors are virtually non-existent and no conclusions can yet be drawn. Longitudinal studies on the cognitive effects of the combined ET-CDK4/6 inhibitors are highly needed to properly inform patients about potential short-term and long-term cognitive side effects. These studies should preferably include cognitive assessments (including a measurement prior to ET), and be designed in such a way that they can account for variables such as type and duration of ET, CDK4/6 inhibition, menopausal status, and other disease- and treatment-related symptoms that can impact cognition, such as fatigue and distress.
Collapse
|
45
|
Astrocyte-Derived Thrombospondin Induces Cortical Synaptogenesis in a Sex-Specific Manner. eNeuro 2021; 8:ENEURO.0014-21.2021. [PMID: 34266964 PMCID: PMC8328272 DOI: 10.1523/eneuro.0014-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/22/2021] [Accepted: 07/03/2021] [Indexed: 12/29/2022] Open
Abstract
The regulation of synaptic connectivity in the brain is vital to proper functioning and development of the CNS. Formation of neural networks in the CNS has been shown to be heavily influenced by astrocytes, which secrete factors, including thrombospondin (TSP) family proteins, that promote synaptogenesis. However, whether this process is different between males and females has not been thoroughly investigated. In this study, we found that cortical neurons purified from newborn male rats showed a significantly more robust synaptogenic response compared with female-derived cells when exposed to factors secreted from astrocytes. This difference was driven largely by the neuronal response to TSP2, which increased synapses in male neurons while showing no effect on female neurons. Blockade of endogenous 17β-estradiol (E2) production with letrozole normalized the TSP response between male and female cells, indicating a level of regulation by estrogen signaling. Our results suggest that male and female neurons show a divergent response to TSP synaptogenic signaling, contributing to sex differences in astrocyte-mediated synaptic connectivity.
Collapse
|
46
|
Lan Z, Meng Z, Lian B, Liu M, Sun T, Sun H, Liu Z, Hu Z, Guo Q, Zhang J. Hippocampal Aromatase Knockdown Aggravates Ovariectomy-Induced Spatial Memory Impairment, Aβ Accumulation and Neural Plasticity Deficiency in Adult Female Mice. Neurochem Res 2021; 46:1188-1202. [PMID: 33559105 DOI: 10.1007/s11064-021-03258-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 12/23/2022]
Abstract
Ovarian estrogens (mainly 17β estradiol, E2) have been involved in the regulation of the structure of hippocampus, the center of spatial memory. In recent years, high levels of aromatase (AROM), the estrogen synthase, has been localized in hippocampus; and this hippocampus-derived E2 seems to be functional in synaptic plasticity and spatial memory as ovarian E2 does. However, the contribution of ovarian E2 and hippocampal E2 to spatial memory and neural plasticity remains unclear. In this study, AROM-specific RNA interference AAVs (shAROM) were constructed and injected into the hippocampus of control or ovariectomized (OVX) mice. Four weeks later the spatial learning and memory behavior was examined with Morris water maze, the expression of hippocampal Aβ related proteins, selected synaptic proteins and CA1 synapse density, actin polymerization related proteins and CA1 spine density were also examined. The results showed that while OVX and hippocampal shAROM contributed similarly to most of the parameters examined, shAROM induced more increase in BACE1 (amyloidogenic β-secretase), more decrease in neprilysin (Aβ remover) and Profilin-1 (actin polymerization inducer). More importantly, combined OVX and shAROM treatment displayed most significant impairment of spatial learning and memory as well as decrease in synaptic plasticity compared to OVX or shAROM alone. In conclusion, the above results clearly demonstrated the crucial role of hippocampal E2 in the regulation of the structure and function of hippocampus besides ovarian E2, indicating that hippocampal E2 content should also be taken into consideration during estrogenic replacement.
Collapse
Affiliation(s)
- Zhen Lan
- Department of Neurobiology, Army Medical University, Chongqing, China
| | - Zhaoyou Meng
- Department of Neurobiology, Army Medical University, Chongqing, China
| | - Biyao Lian
- Department of Neurobiology, Army Medical University, Chongqing, China
- Department of Pediatrics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mengying Liu
- Department of Neurobiology, Army Medical University, Chongqing, China
- The 305 Hospital of PLA, Beijing, China
| | - Tao Sun
- Department of Neurobiology, Army Medical University, Chongqing, China
- The 63650 Hospital of PLA, Malan, China
| | - Huan Sun
- Department of Neurobiology, Army Medical University, Chongqing, China
- Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhi Liu
- Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Zhenxin Hu
- Battalion One of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Qiang Guo
- Department of Basic Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing, China.
| |
Collapse
|
47
|
Iglesias-Osma MC, Blanco EJ, Carretero-Hernández M, Catalano-Iniesta L, García-Barrado MJ, Sánchez-Robledo V, Blázquez JL, Carretero J. The lack of Irs2 induces changes in the immunocytochemical expression of aromatase in the mouse retina. Ann Anat 2021; 239:151726. [PMID: 33798691 DOI: 10.1016/j.aanat.2021.151726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 02/01/2023]
Abstract
Insulin receptor substrate (Irs) belongs to a family of proteins that mediate the intracellular signaling of insulin and IGF-1. Insulin receptor substrate 2 (Irs2) is necessary for retinal function, since its failure in Irs2-deficient mice in hyperglycemic situation promotes photoreceptor degeneration and visual dysfunction, like in diabetic retinopathy. The expression of P450 aromatase, which catalyzes androgen aromatization to form 17ß-estradiol, increases in some neurodegenerative diseases thus promoting the local synthesis of neuroestrogens that exert relevant neuroprotective functions. Aromatase is also expressed in neurons and glial cells of the central nervous system (CNS), including the retina. To further understand the role of Irs2 at the retinal level, we performed an immunocytochemical study in adult normoglycemic Irs2-deficient mice. For this aim, the retinal immunoexpression of neuromodulators, such as aromatase, glutamine synthetase (GS), and tyrosine hydroxylase (TH) was analyzed, joint to a morphometric and planimetric study of the retinal layers. Comparing with wild-type (WT) control mice, the Irs2-knockout (Irs2-KO) animals showed a significant increase in the immunopositivity to aromatase in almost all of the retinal layers. Besides, Irs2-KO mice exhibited a decreased immunopositive reaction for GS and TH, in Müller and amacrine cells, respectively; morphological variations were also found in these retinal cell types. Furthermore, the retina of Irs2-KO mice displayed alterations in the structural organization, and a generalized decrease in the retinal thickness was observed in each of the layers, except for the inner nuclear layer. Our findings suggest that the absence of Irs2 induces retinal neurodegenerative changes in Müller and amacrine cells that are unrelated to hyperglycemia. Accordingly, in the Irs2-KO mice, the increased retinal immunocytochemical reactivity of aromatase could be associated with an attempt to repair such neural retina injuries by promoting local neuroprotective mediators.
Collapse
Affiliation(s)
- Maria Carmen Iglesias-Osma
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain.
| | - Enrique J Blanco
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain; Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain
| | - Marta Carretero-Hernández
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain; Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain
| | - Leonardo Catalano-Iniesta
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - Maria Jose García-Barrado
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - Virginia Sánchez-Robledo
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - Juan Luis Blázquez
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain; Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain
| | - Jose Carretero
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain; Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain.
| |
Collapse
|
48
|
Baumgartner NE, Daniel JM. Estrogen receptor α: a critical role in successful female cognitive aging. Climacteric 2021; 24:333-339. [PMID: 33522313 DOI: 10.1080/13697137.2021.1875426] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Due to potential health risks, current recommendations are that individuals who wish to use hormone therapy to treat menopausal symptoms do so for the shortest period of time possible. In our investigation into how short-term use of estrogens in midlife following loss of ovarian function exerts long-term effects on female cognitive aging in rodents, we discovered a link between the ability of previous exposure to estradiol to enhance memory in the long term and its ability to increase estrogen receptor α (ERα) levels in the hippocampus, a brain area important for memory. Follow-up studies in model systems implicate a role for ERα in enhanced cognitive function independent of ovarian or exogenously administered estrogens. Results are consistent with clinical studies in which brain ERα levels in older women and men are related to cognitive functioning and risk of cognitive decline is associated with polymorphisms in the gene that transcribes ERα. Research in preclinical models reveals mechanisms through which ERα can be activated and affect cognition in the absence of ovarian estrogens, including ligand-independent activation via insulin-like growth factor-1 signaling and activation by brain-derived neuroestrogens. This report reviews preclinical and clinical data that collectively point to the importance of ERα in cognition and highlights the need to differentiate the role of estrogen receptors from their classical ligands as we seek approaches to facilitate successful cognitive aging.
Collapse
Affiliation(s)
- N E Baumgartner
- Neuroscience Program, Tulane University, New Orleans, LA, USA.,Brain Institute, Tulane University, New Orleans, LA, USA
| | - J M Daniel
- Neuroscience Program, Tulane University, New Orleans, LA, USA.,Brain Institute, Tulane University, New Orleans, LA, USA.,Department of Psychology, Tulane University, New Orleans, LA, USA
| |
Collapse
|
49
|
Briski KP, Ibrahim MMH, Mahmood ASMH, Alshamrani AA. Norepinephrine Regulation of Ventromedial Hypothalamic Nucleus Astrocyte Glycogen Metabolism. Int J Mol Sci 2021; 22:ijms22020759. [PMID: 33451134 PMCID: PMC7828624 DOI: 10.3390/ijms22020759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/04/2021] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
The catecholamine norepinephrine (NE) links hindbrain metabolic-sensory neurons with key glucostatic control structures in the brain, including the ventromedial hypothalamic nucleus (VMN). In the brain, the glycogen reserve is maintained within the astrocyte cell compartment as an alternative energy source to blood-derived glucose. VMN astrocytes are direct targets for metabolic stimulus-driven noradrenergic signaling due to their adrenergic receptor expression (AR). The current review discusses recent affirmative evidence that neuro-metabolic stability in the VMN may be shaped by NE influence on astrocyte glycogen metabolism and glycogen-derived substrate fuel supply. Noradrenergic modulation of estrogen receptor (ER) control of VMN glycogen phosphorylase (GP) isoform expression supports the interaction of catecholamine and estradiol signals in shaping the physiological stimulus-specific control of astrocyte glycogen mobilization. Sex-dimorphic NE control of glycogen synthase and GP brain versus muscle type proteins may be due, in part, to the dissimilar noradrenergic governance of astrocyte AR and ER variant profiles in males versus females. Forthcoming advances in the understanding of the molecular mechanistic framework for catecholamine stimulus integration with other regulatory inputs to VMN astrocytes will undoubtedly reveal useful new molecular targets in each sex for glycogen mediated defense of neuronal metabolic equilibrium during neuro-glucopenia.
Collapse
|
50
|
Ambrase A, Lewis CA, Barth C, Derntl B. Influence of ovarian hormones on value-based decision-making systems: Contribution to sexual dimorphisms in mental disorders. Front Neuroendocrinol 2021; 60:100873. [PMID: 32987043 DOI: 10.1016/j.yfrne.2020.100873] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/28/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Women and men exhibit differences in behavior when making value-based decisions. Various hypotheses have been proposed to explain these findings, stressing differences in functional lateralization of the brain, functional activation, neurotransmitter involvement and more recently, sex hormones. While a significant interaction of neurotransmitter systems and sex hormones has been shown for both sexes, decision-making in women might be particularly affected by variations of ovarian hormones. In this review we have gathered information from animal and human studies on how ovarian hormones affect decision-making processes in females by interacting with neurotransmitter systems at functionally relevant brain locations and thus modify the computation of decision aspects. We also review previous findings on impaired decision-making in animals and clinical populations with substance use disorder and depression, emphasizing how little we know about the role of ovarian hormones in aberrant decision-making.
Collapse
Affiliation(s)
- Aiste Ambrase
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; International Max Planck Research School for Cognitive and Systems Neuroscience, University of Tübingen, Tuebingen, Germany
| | - Carolin A Lewis
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; Emotion Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; International Max Planck Research School on Neuroscience of Communication: Function, Structure, and Plasticity, Leipzig, Germany
| | - Claudia Barth
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tübingen, Germany; International Max Planck Research School for Cognitive and Systems Neuroscience, University of Tübingen, Tuebingen, Germany; TübingenNeuroCampus, University of Tübingen, Tübingen, Germany; LEAD Research School and Graduate Network, University of Tübingen, Tübingen, Germany.
| |
Collapse
|