1
|
Flannery JC, Tirrell PS, Baumgartner NE, Daniel JM. Neuroestrogens, the hippocampus, and female cognitive aging. Horm Behav 2025; 170:105710. [PMID: 40036999 DOI: 10.1016/j.yhbeh.2025.105710] [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: 08/30/2024] [Revised: 12/30/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Research conducted over the last several decades implicates ovarian estrogens as important modulators of hippocampal function. More recently however, the importance of estrogens synthesized in the brain de novo for hippocampal function has been recognized. These brain-derived neuroestrogens act in the hippocampus to regulate dendritic spine dynamics and synaptic plasticity as well as hippocampus-dependent memory. The current report provides an overview of research conducted in model systems elucidating the actions of neuroestrogens in the hippocampus and the subsequent consequences for cognition. We highlight the relationship between ovarian estrogens and brain-derived estrogens and discuss implications for female cognitive aging of the putative decline in hippocampal levels of neuroestrogens following loss of ovarian function. Finally, we propose a model of menopause in which a short-term period of midlife estradiol treatment changes the trajectory of hippocampal neuroestrogen production long-term, resulting in sustained interactions of neuroestrogens, insulin-like growth factor-1, and estrogen receptor signaling in the hippocampus, interactions that support successful brain and cognitive aging.
Collapse
Affiliation(s)
- Jill C Flannery
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America
| | - Parker S Tirrell
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America
| | - Nina E Baumgartner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States of America
| | - Jill M Daniel
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America; Department of Psychology, Tulane University, New Orleans, LA, 70118, United States of America.
| |
Collapse
|
2
|
Fan D, Wang X, Ling X, Li H, Zhang L, Zheng W, Wu Z, Ai L. Utilisation of the Innovative [18F]-Labelled Radiotracer [18F]-BIBD-071 Within HR+ Breast Cancer Xenograft Mouse Models. Pharmaceuticals (Basel) 2025; 18:66. [PMID: 39861129 PMCID: PMC11768299 DOI: 10.3390/ph18010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Aromatase plays a crucial role in the conversion of androgens to oestrogens and is often overexpressed in hormone-dependent tumours, particularly breast cancer. [18F]BIBD-071, which has excellent binding affinity for aromatase and good pharmacokinetics, has potential for the diagnosis and treatment of aromatase-related diseases. The MCF-7 cell line, which is hormone receptor-positive (HR+), was used in the assessment of the novel [18F]-labelled radiotracer [18F]BIBD-071 via positron emission tomography (PET) imaging of an HR+ breast cancer xenograft model. Methods: [18F]BIBD-071 was synthesised, radiolabelled, and then subjected to in vitro stability testing. MCF-7 cells were cultured and implanted into BALB/c nude mice to establish subcutaneous tumour models. MicroPET/CT imaging was conducted after injection of the tracer at 1 and 2 h, and a blocking study was also conducted using the aromatase inhibitor letrozole. A block experiment was used to prove the specificity of the probe. Biodistribution studies were performed at 0.5, 1, and 2 h post injection (p.i.). Immunofluorescence was used to assess aromatase expression in MCF-7 cells. Results: [18F]BIBD-071 showed excellent in vitro stability and specific uptake in an MCF-7 xenograft tumour model. MicroPET/CT imaging at 1 and 2 h p.i. revealed excellent tumour visualisation with a favourable tumour-to-background ratio. Biodistribution data revealed high tracer uptake in the liver, small intestine, and stomach, with significant washout from the bloodstream and tumour over time. The tumour uptakes at 0.5 h, 1 h, and 2 h were 3.84 ± 0.13, 2.5 ± 0.17, and 2.54 ± 0.32, respectively. The tumour uptake significantly decreased between 0.5 h and 1 h (p < 0.0001), whereas there was no significant difference between 1 and 2 h. The tumour/background ratios at 0.5 h, 1 h, and 2 h were 1.19 ± 0.03, 1.12 ± 0.17, and 1.42 ± 0.11, respectively. Immunofluorescence confirmed robust aromatase expression in MCF-7 cells, which was correlated with [18F]BIBD-071 tumour uptake. Conclusions: [18F]BIBD-071 is a promising PET tracer for diagnosing and monitoring HR+ breast cancer, warranting further research into hormone-dependent cancers.
Collapse
Affiliation(s)
- Di Fan
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China; (D.F.); (X.W.); (H.L.)
| | - Xin Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China; (D.F.); (X.W.); (H.L.)
| | - Xueyuan Ling
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China;
| | - Hongbin Li
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China; (D.F.); (X.W.); (H.L.)
| | - Lu Zhang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.)
| | - Wei Zheng
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.)
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.)
| | - Lin Ai
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China; (D.F.); (X.W.); (H.L.)
| |
Collapse
|
3
|
Bautista-Abad Á, García-Magro N, Pinto-Benito D, Cáceres-Pajuelo JE, Alises CV, Ganchala D, Lagunas N, Negredo P, García-Segura LM, Arevalo MA, Grassi D. Aging is associated with sex-specific alteration in the expression of genes encoding for neuroestradiol synthesis and signaling proteins in the mouse trigeminal somatosensory input. GeroScience 2024; 46:6459-6472. [PMID: 38954130 PMCID: PMC11493896 DOI: 10.1007/s11357-024-01268-z] [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: 10/26/2023] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
Pain perception is influenced by sex and aging, with previous studies indicating the involvement of aromatase, the estradiol synthase enzyme, in regulating pain perception. Previous research has established the presence of aromatase in dorsal root ganglia sensory neurons and its role in modulating pain perception. The present study aims to explore the implications of aging and sex on the expression of aromatase and estrogen receptors in the trigeminal ganglion. The study examined mRNA levels of aromatase, ERs, and the androgen receptor (AR) in the trigeminal ganglion of 3-month-old and 27-month-old male and female mice, as well as 3-month-old mice from the four-core genotype (FCG) transgenic model. The latter facilitates the assessment of gonadal hormone and sex chromosome implications for sex-specific traits. Aromatase localization in the ganglion was further assessed through immunohistochemistry. Aromatase immunoreactivity was observed for the first time in sensory neurons within the trigeminal ganglion. Trigeminal ganglion gene expressions were detected for aromatase, ERs, and AR in both sexes. Aromatase, ERβ, and GPER gene expressions were higher in young males versus young females. Analyses of the FCG model indicated that sex differences depended solely on gonadal sex. The aging process induced an enhancement in the expression of aromatase, ERs, and AR genes across both sexes, culminating in a reversal of the previously observed gender-based differences. the potential impact of estrogen synthesis and signaling in the trigeminal ganglion on age and sex differences warrants consideration, particularly in relation to trigeminal sensory functions and pain perception.
Collapse
MESH Headings
- Animals
- Female
- Aromatase/genetics
- Aromatase/metabolism
- Male
- Trigeminal Ganglion/metabolism
- Aging/genetics
- Aging/metabolism
- Aging/physiology
- Mice
- Estradiol/metabolism
- Mice, Transgenic
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Sex Factors
- Pain Perception/physiology
- Signal Transduction/genetics
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Sensory Receptor Cells/metabolism
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Mice, Inbred C57BL
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
Collapse
Affiliation(s)
- Álvaro Bautista-Abad
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid, Spain
| | - Nuria García-Magro
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda Km 1,800, Pozuelo de Alarcón, Madrid, Spain
| | - Daniel Pinto-Benito
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Julio Eduardo Cáceres-Pajuelo
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid, Spain
| | - Carlos Vicente Alises
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid, Spain
| | - Danny Ganchala
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Natalia Lagunas
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Madrid, Spain
| | - Pilar Negredo
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid, Spain
| | - Luis Miguel García-Segura
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria-Angeles Arevalo
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| | - Daniela Grassi
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid, Spain.
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Aspesi D, Cornil CA. Role of neuroestrogens in the regulation of social behaviors - From social recognition to mating. Neurosci Biobehav Rev 2024; 161:105679. [PMID: 38642866 DOI: 10.1016/j.neubiorev.2024.105679] [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: 12/07/2023] [Revised: 03/12/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
In this mini-review, we summarize the brain distribution of aromatase, the enzyme catalyzing the synthesis of estrogens from androgens, and the mechanisms responsible for regulating estrogen production within the brain. Understanding this local synthesis of estrogens by neurons is pivotal as it profoundly influences various facets of social behavior. Neuroestrogen action spans from the initial processing of socially pertinent sensory cues to integrating this information with an individual's internal state, ultimately resulting in the manifestation of either pro-affiliative or - aggressive behaviors. We focus here in particular on aggressive and sexual behavior as the result of correct individual recognition of intruders and potential mates. The data summarized in this review clearly point out the crucial role of locally synthesized estrogens in facilitating rapid adaptation to the social environment in rodents and birds of both sexes. These observations not only shed light on the evolutionary significance but also indicate the potential implications of these findings in the realm of human health, suggesting a compelling avenue for further investigation.
Collapse
Affiliation(s)
- Dario Aspesi
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | | |
Collapse
|
5
|
Zhao J, He C, Fan X, Wang L, Zhao L, Liu H, Shen W, Jiang S, Pei K, Gao J, Qi Y, Liu Y, Zhao J, Zhang R, Lu C, Tong J, Huai J. Tripeptidyl peptidase II coordinates the homeostasis of calcium and lipids in the central nervous system and its depletion causes presenile dementia in female mice through calcium/lipid dyshomeostasis-induced autophagic degradation of CYP19A1. Theranostics 2024; 14:1390-1429. [PMID: 38389851 PMCID: PMC10879859 DOI: 10.7150/thno.92571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Tripeptidyl peptidase II (TPP2) has been proven to be related to human immune and neurological diseases. It is generally considered as a cytosolic protein which forms the largest known protease complex in eukaryotic cells to operate mostly downstream of proteasomes for degradation of longer peptides. However, this canonical function of TPP2 cannot explain its role in a wide variety of biological and pathogenic processes. The mechanistic interrelationships and hierarchical order of these processes have yet to be clarified. Methods: Animals, cells, plasmids, and viruses established and/or used in this study include: TPP2 knockout mouse line, TPP2 conditional knockout mouse lines (different neural cell type oriented), TRE-TPP2 knockin mouse line on the C57BL/6 background; 293T cells with depletion of TPP2, ATF6, IRE1, PERK, SYVN1, UCHL1, ATG5, CEPT1, or CCTα, respectively; 293T cells stably expressing TPP2, TPP2 S449A, TPP2 S449T, or CCTα-KDEL proteins on the TPP2-depleted background; Plasmids for eukaryotic transient expression of rat CYP19A1-Flag, CYP19A1 S118A-Flag, CYP19A1 S118D-Flag, Sac I ML GFP Strand 11 Long, OMMGFP 1-10, G-CEPIA1er, GCAMP2, CEPIA3mt, ACC-GFP, or SERCA1-GFP; AAV2 carrying the expression cassette of mouse CYP19A1-3 X Flag-T2A-ZsGreen. Techniques used in this study include: Flow cytometry, Immunofluorescence (IF) staining, Immunohistochemical (IHC) staining, Luxol fast blue (LFB) staining, β-galactosidase staining, Lipid droplet (LD) staining, Calcium (Ca2+) staining, Stimulated emission depletion (STED) imaging, Transmission electron microscopic imaging, Two-photon imaging, Terminal deoxynucleotidyl transferase (TdT) dUTP nick-end Labeling (TUNEL) assay, Bromodeoxyuridine (BrdU) assay, Enzymatic activity assay, Proximity ligation assay (PLA), In vivo electrophysiological recording, Long-term potentiation (LTP) recording, Split-GFP-based mitochondria-associated membrane (MAM) detection, Immunoprecipitation (IP), Cellular fractionation, In situ hybridization, Semi-quantitative RT-PCR, Immunoblot, Mass spectrometry-based lipidomics, metabolomics, proteomics, Primary hippocampal neuron culture and Morris water maze (MWM) test. Results: We found that TPP2, independent of its enzymatic activity, plays a crucial role in maintaining the homeostasis of intracellular Ca2+ and phosphatidylcholine (PC) in the central nervous system (CNS) of mice. In consistence with the critical importance of Ca2+ and PC in the CNS, TPP2 gene ablation causes presenile dementia in female mice, which is closely associated with Ca2+/PC dysregulation-induced endoplasmic reticulum (ER) stress, abnormal autophagic degradation of CYP19A1 (aromatase), and estrogen depletion. This work therefore uncovers a new role of TPP2 in lipogenesis and neurosteroidogenesis which is tightly related to cognitive function of adult female mice. Conclusion: Our study reveals a crucial role of TPP2 in controlling homeostasis of Ca2+ and lipids in CNS, and its deficiency causes sexual dimorphism in dementia. Thus, this study is not only of great significance for elucidating the pathogenesis of dementia and its futural treatment, but also for interpreting the role of TPP2 in other systems and their related disorders.
Collapse
Affiliation(s)
- Jin Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Chengtong He
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Xueyu Fan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Lin Wang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Liao Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Hui Liu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Wujun Shen
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Sanwei Jiang
- Henan International Key Laboratory for Noninvasive Neuromodulation, Department of Physiology & Pathology, Xinxiang Medical University, Xinxiang, PR China
| | - Kaixuan Pei
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Jingjing Gao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Yawei Qi
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Yang Liu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Junqiang Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
| | - Ruiling Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
| | - Chengbiao Lu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
- Henan International Key Laboratory for Noninvasive Neuromodulation, Department of Physiology & Pathology, Xinxiang Medical University, Xinxiang, PR China
- Senior author for electrophysiological experiments and related analysis
| | - Jia Tong
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Jisen Huai
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, PR China
| |
Collapse
|
6
|
Wasilczuk AZ, Rinehart C, Aggarwal A, Stone ME, Mashour GA, Avidan MS, Kelz MB, Proekt A. Hormonal basis of sex differences in anesthetic sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2312913120. [PMID: 38190526 PMCID: PMC10801881 DOI: 10.1073/pnas.2312913120] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/20/2023] [Indexed: 01/10/2024] Open
Abstract
General anesthesia-a pharmacologically induced reversible state of unconsciousness-enables millions of life-saving procedures. Anesthetics induce unconsciousness in part by impinging upon sexually dimorphic and hormonally sensitive hypothalamic circuits regulating sleep and wakefulness. Thus, we hypothesized that anesthetic sensitivity should be sex-dependent and modulated by sex hormones. Using distinct behavioral measures, we show that at identical brain anesthetic concentrations, female mice are more resistant to volatile anesthetics than males. Anesthetic sensitivity is bidirectionally modulated by testosterone. Castration increases anesthetic resistance. Conversely, testosterone administration acutely increases anesthetic sensitivity. Conversion of testosterone to estradiol by aromatase is partially responsible for this effect. In contrast, oophorectomy has no effect. To identify the neuronal circuits underlying sex differences, we performed whole brain c-Fos activity mapping under anesthesia in male and female mice. Consistent with a key role of the hypothalamus, we found fewer active neurons in the ventral hypothalamic sleep-promoting regions in females than in males. In humans, we demonstrate that females regain consciousness and recover cognition faster than males after identical anesthetic exposures. Remarkably, while behavioral and neurocognitive measures in mice and humans point to increased anesthetic resistance in females, cortical activity fails to show sex differences under anesthesia in either species. Cumulatively, we demonstrate that sex differences in anesthetic sensitivity are evolutionarily conserved and not reflected in conventional electroencephalographic-based measures of anesthetic depth. This covert resistance to anesthesia may explain the higher incidence of unintended awareness under general anesthesia in females.
Collapse
Affiliation(s)
- Andrzej Z. Wasilczuk
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Cole Rinehart
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
| | - Adeeti Aggarwal
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
| | - Martha E. Stone
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
| | - George A. Mashour
- Center for Consciousness Science, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI48105
| | - Michael S. Avidan
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO63110
| | - Max B. Kelz
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Alex Proekt
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - ReCCognition Study Group
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Center for Consciousness Science, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI48105
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
7
|
Arjmand S, Bender D, Jakobsen S, Wegener G, Landau AM. Peering into the Brain's Estrogen Receptors: PET Tracers for Visualization of Nuclear and Extranuclear Estrogen Receptors in Brain Disorders. Biomolecules 2023; 13:1405. [PMID: 37759805 PMCID: PMC10526964 DOI: 10.3390/biom13091405] [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: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogen receptors (ERs) play a multitude of roles in brain function and are implicated in various brain disorders. The use of positron emission tomography (PET) tracers for the visualization of ERs' intricate landscape has shown promise in oncology but remains limited in the context of brain disorders. Despite recent progress in the identification and development of more selective ligands for various ERs subtypes, further optimization is necessary to enable the reliable and efficient imaging of these receptors. In this perspective, we briefly touch upon the significance of estrogen signaling in the brain and raise the setbacks associated with the development of PET tracers for identification of specific ERs subtypes in the brain. We then propose avenues for developing efficient PET tracers to non-invasively study the dynamics of ERs in the brain, as well as neuropsychiatric diseases associated with their malfunction in a longitudinal manner. This perspective puts several potential candidates on the table and highlights the unmet needs and areas requiring further research to unlock the full potential of PET tracers for ERs imaging, ultimately aiding in deepening our understanding of ERs and forging new avenues for potential therapeutic strategies.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Dirk Bender
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
| | - Anne M. Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| |
Collapse
|
8
|
Li Q, Qi S, Liang J, Tian Y, He S, Liao Q, Xing S, Han L, Chen X. Review of triazole scaffolds for treatment and diagnosis of Alzheimer's disease. Chem Biol Interact 2023; 382:110623. [PMID: 37451665 DOI: 10.1016/j.cbi.2023.110623] [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: 05/09/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Triazole scaffolds, a series of 5-membered heterocycles, are well known for their high efficacy, low toxicity, and superior pharmacokinetics. Alzheimer's disease (AD) is the first neurodegenerative disorder with complex pathological mechanisms. Triazole, as an aromatic group with three nitrogen atoms, forms polar and non-polar interactions with diverse key residues in the receptor-ligand binding procedure, and has been widely used in the molecular design in the development of anti-AD agents. Moreover, considering the simple synthesis approaches, triazole scaffolds are commonly used to link two pharmacodynamic groups in one chemical molecule, forming multi-target directed ligands (MTDLs). Furthermore, the click reaction between azide- and cyano-modified enzyme and ligand provides feasibility for the new modulator discovery, compound tissue distribution evaluation, enzyme localization, and pharmacological mechanism study, promoting the diagnosis of AD course.
Collapse
Affiliation(s)
- Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| | - Shulei Qi
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Jinxin Liang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Yuqing Tian
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Siyu He
- Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Qinghong Liao
- Shandong Junrong Technology Transfer Co., Ltd, Qingdao, 266071, Shandong, PR China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Lingfei Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Xuehong Chen
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| |
Collapse
|
9
|
Dubol M, Immenschuh J, Jonasson M, Takahashi K, Niwa T, Hosoya T, Roslin S, Wikström J, Antoni G, Watanabe Y, Lubberink M, Biegon A, Sundström-Poromaa I, Comasco E. Acute nicotine exposure blocks aromatase in the limbic brain of healthy women: A [ 11C]cetrozole PET study. Compr Psychiatry 2023; 123:152381. [PMID: 36905856 DOI: 10.1016/j.comppsych.2023.152381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/07/2023] [Accepted: 02/28/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Of interest to women's mental health, a wealth of studies suggests sex differences in nicotine addiction and treatment response, but their psychoneuroendocrine underpinnings remain largely unknown. A pathway involving sex steroids could indeed be involved in the behavioural effects of nicotine, as it was found to inhibit aromatase in vitro and in vivo in rodents and non-human primates, respectively. Aromatase regulates the synthesis of oestrogens and, of relevance to addiction, is highly expressed in the limbic brain. METHODS The present study sought to investigate in vivo aromatase availability in relation to exposure to nicotine in healthy women. Structural magnetic resonance imaging and two [11C]cetrozole positron emission tomography (PET) scans were performed to assess the availability of aromatase before and after administration of nicotine. Gonadal hormones and cotinine levels were measured. Given the region-specific expression of aromatase, a ROI-based approach was employed to assess changes in [11C]cetrozole non-displaceable binding potential. RESULTS The highest availability of aromatase was found in the right and left thalamus. Upon nicotine exposure, [11C]cetrozole binding in the thalamus was acutely decreased bilaterally (Cohen's d = -0.99). In line, cotinine levels were negatively associated with aromatase availability in the thalamus, although as non-significant trend. CONCLUSIONS These findings indicate acute blocking of aromatase availability by nicotine in the thalamic area. This suggests a new putative mechanism mediating the effects of nicotine on human behaviour, particularly relevant to sex differences in nicotine addiction.
Collapse
Affiliation(s)
- Manon Dubol
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jana Immenschuh
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - My Jonasson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Kayo Takahashi
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takashi Niwa
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takamitsu Hosoya
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sara Roslin
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Johan Wikström
- Department of Surgical Sciences, Neuroradiology, Uppsala University, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Mark Lubberink
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anat Biegon
- Departments of Radiology and Neurology, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | | | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Court L, Balthazart J, Ball GF, Cornil CA. Role of aromatase in distinct brain nuclei of the social behaviour network in the expression of sexual behaviour in male Japanese quail. J Neuroendocrinol 2022; 34:e13127. [PMID: 35394094 PMCID: PMC9250618 DOI: 10.1111/jne.13127] [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: 11/14/2021] [Revised: 02/15/2022] [Accepted: 03/09/2022] [Indexed: 11/29/2022]
Abstract
In male Japanese quail, brain aromatase is crucial for the hormonal activation of sexual behaviour, but the sites producing neuro-oestrogens that are critical for these behaviours have not been completely identified. This study examined the function of aromatase expressed in several nuclei of the social behaviour network on a measure of sexual motivation known as the frequency of rhythmic cloacal sphincter movements (RCSM) and on copulatory behaviour. Sexually experienced castrated males chronically treated with testosterone were stereotaxically implanted with the aromatase inhibitor vorozole (VOR), or cholesterol as control, and tested for sexual behaviour. In experiment 1, males were implanted in the medial preoptic nucleus (POM) with VOR, a manipulation known to reduce the expression of copulatory behaviour. This experiment served as positive control, but also showed that VOR implanted in the dorsomedial or lateral portions of the POM similarly inhibits male copulatory behaviour compared to control implants. In experiments 2 to 4, males received stereotaxic implants of VOR in the periaqueductal gray (PAG), the nucleus taeniae of the amygdala (TnA) and the ventromedial nucleus of the hypothalamus (VMN), respectively. Sexual behaviour was affected only in individuals where VOR was implanted in the PAG: these males displayed significantly lower frequencies of cloacal contact movements, the last step of the copulatory sequence. Inhibition of aromatase in the TnA and VMN did not alter copulatory ability. Overall, RCSM frequency remained unaffected by VOR regardless of implantation site. Together, these results suggest that neuro-oestrogens produced in the POM contribute the most to the control of male copulatory behaviour, while aromatase expressed in the PAG might also participate to premotor aspects of male copulatory behaviour.
Collapse
Affiliation(s)
- Lucas Court
- GIGA Neurosciences, University of Liège, B-4000 Liège, Belgium
| | | | - Gregory F. Ball
- Department of Psychology, University of Maryland, College Park, MD, USA
| | | |
Collapse
|
11
|
Zheng W, Cheng X, Chen H, Jiang Z, Sun Y, Yu Z, Yang T, Zhang L, Liu Y, Ji X, Wu Z. Novel 18F-Labeled PET Tracers Specific to Aromatase: Design, Synthesis, and Biological Evaluation. Mol Pharm 2022; 19:2456-2470. [PMID: 35621695 DOI: 10.1021/acs.molpharmaceut.2c00176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The abnormal expression of aromatase is associated with the occurrence and development of a variety of neurological diseases and tumors. A series of 18F-labeled and 68Ga-labeled potential aromatase-binding candidate compounds were designed and synthesized based on the structures of aromatase inhibitors. Competitive inhibition experiments in vitro and molecular docking showed that BIBD-069 and BIBD-071 have high affinity for aromatase. The radiolabeling conditions of [18F]BIBD-069 and [18F]BIBD-071 were simple, and the yields were high. Biodistribution and in vivo inhibition experiments confirmed that [18F]BIBD-069 and [18F]BIBD-071 specifically bind to aromatase. [18F]BIBD-069 and [18F]BIBD-071 selectively imaged the amygdala and nucleus of the stria terminalis, which is similar to the imaging result of [11C]vorozole. Radiometabolites of [18F]BIBD-069 and [18F]BIBD-071 did not bind to aromatase and interfered with brain imaging. MicroPET-CT imaging further confirmed that [18F]BIBD-069 and [18F]BIBD-071 can specifically bind to aromatase and were not defluorinated in vivo. Given that [18F]BIBD-069 and [18F]BIBD-071 exhibit excellent aromatase binding affinities, mild radiolabeling conditions, and good pharmacokinetics, they can be important tools for the diagnosis and treatment of aromatase-related diseases.
Collapse
Affiliation(s)
- Wei Zheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xuebo Cheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zeng Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yuli Sun
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ziyue Yu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Tingyu Yang
- School of Pharmaceutical Science, Capital Medical University, Beijing 100069, China
| | - Lu Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yajing Liu
- School of Pharmaceutical Science, Capital Medical University, Beijing 100069, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.,Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
12
|
Maioli S, Leander K, Nilsson P, Nalvarte I. Estrogen receptors and the aging brain. Essays Biochem 2021; 65:913-925. [PMID: 34623401 PMCID: PMC8628183 DOI: 10.1042/ebc20200162] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022]
Abstract
The female sex hormone estrogen has been ascribed potent neuroprotective properties. It signals by binding and activating estrogen receptors that, depending on receptor subtype and upstream or downstream effectors, can mediate gene transcription and rapid non-genomic actions. In this way, estrogen receptors in the brain participate in modulating neural differentiation, proliferation, neuroinflammation, cholesterol metabolism, synaptic plasticity, and behavior. Circulating sex hormones decrease in the course of aging, more rapidly at menopause in women, and slower in men. This review will discuss what this drop entails in terms of modulating neuroprotection and resilience in the aging brain downstream of spatiotemporal estrogen receptor alpha (ERα) and beta (ERβ) signaling, as well as in terms of the sex differences observed in Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, controversies related to ER expression in the brain will be discussed. Understanding the spatiotemporal signaling of sex hormones in the brain can lead to more personalized prevention strategies or therapies combating neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Maioli
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Karin Leander
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57 Huddinge, Sweden
| |
Collapse
|
13
|
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: 16] [Impact Index Per Article: 4.0] [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
|
14
|
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: 6] [Impact Index Per Article: 1.5] [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
|
15
|
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: 67] [Impact Index Per Article: 16.8] [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
|
16
|
Baron JA, Nichols HB, Anderson C, Safe S. Cigarette Smoking and Estrogen-Related Cancer. Cancer Epidemiol Biomarkers Prev 2021; 30:1462-1471. [PMID: 33990391 DOI: 10.1158/1055-9965.epi-20-1803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/13/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Cigarette smoking is a known cause of many cancers, yet epidemiologic studies have found protective associations with the risk of four "estrogen-related" malignancies: endometrial cancer, endometrioid and clear cell ovarian cancers, and thyroid cancer. This review considers epidemiologic and biological aspects of these associations, focusing particularly on estrogen signaling, and contrasts them with those for breast cancer, another estrogen-related malignancy. The observational findings regarding the inverse associations are consistent and remain after adjustment for possible confounding factors. In general, women who smoke do not have lower circulating estrogen levels than nonsmokers, eliminating one possible explanation for reduced risks of these malignancies. For endometrial and endometrioid ovarian cancer, the negative associations could plausibly be explained by interference with signaling through the estrogen receptor α. However, this is unlikely to explain the lower risks of thyroid and clear cell ovarian cancers. For thyroid cancer, an anti-inflammatory effect of nicotine and reduced TSH levels from smoking have been proposed explanations for the inverse association, but both lack convincing evidence. While the overall impact of cigarette smoking is overwhelmingly negative, protective associations such as those discussed here can provide potential clues to disease etiology, treatment, and prevention.
Collapse
Affiliation(s)
- John A Baron
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina. .,Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina.,Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Hazel B Nichols
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Chelsea Anderson
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|
17
|
Court L, Balthazart J, Ball GF, Cornil CA. Effect of chronic intracerebroventricular administration of an aromatase inhibitor on the expression of socio-sexual behaviors in male Japanese quail. Behav Brain Res 2021; 410:113315. [PMID: 33901434 DOI: 10.1016/j.bbr.2021.113315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022]
Abstract
Aromatase converts androgens into estrogens in the brain of vertebrates including humans. This enzyme is also expressed in other tissues where its action may result in negative effects on human health (e.g., promotion of tumor growth). To prevent these effects, aromatase inhibitors were developed and are currently used to block human estrogen-dependent tumors. In vertebrates including quail, aromatase is expressed in a highly conserved set of interconnected brain nuclei known as the social behavior network. This network is directly implicated in the expression of a large range of social behaviors. The primary goal of this study was to characterize in Japanese quail the potential impact of brain aromatase on sexual behavior, aggressiveness and social motivation (i.e., tendency to approach and stay close to conspecifics). An additional goal was to test the feasibility and effectiveness of long-term delivery of an aromatase inhibitor directly into the third ventricle via Alzet™ osmotic minipumps using male sexual behavior as the aromatase dependent measure. We demonstrate that this mode of administration results in the strongest inhibition of both copulatory behavior and sexual motivation ever observed in this species, while other social behaviors were variably affected. Sexual motivation and the tendency to approach a group of conspecifics including females clearly seem to depend on brain aromatase, but the effects of central estrogen production on aggressive behavior and on the motivation to approach males remain less clear.
Collapse
Affiliation(s)
- Lucas Court
- GIGA Neurosciences, University of Liège, B-4000, Liège, Belgium
| | | | - Gregory F Ball
- Department of Psychology, University of Maryland, College Park, MD, USA
| | | |
Collapse
|
18
|
Handelsman DJ, Gibson E, Davis S, Golebiowski B, Walters KA, Desai R. Ultrasensitive Serum Estradiol Measurement by Liquid Chromatography-Mass Spectrometry in Postmenopausal Women and Mice. J Endocr Soc 2020; 4:bvaa086. [PMID: 33154982 DOI: 10.1210/jendso/bvaa086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 07/09/2020] [Indexed: 11/19/2022] Open
Abstract
Accurate measurement of very low circulating estradiol (E2) (<5 pg/ml) in postmenopausal women and in mice is essential to investigating sex steroid action in target tissues. However, direct immunoassays are too inaccurate and conventional mass spectrometry-based measurement too insensitive at these serum E2 levels. We report application of an ultrasensitive method using a novel estrogen-selective derivatization in liquid chromatography-mass spectrometry to measure serum E2, with a detection limit of 0.25 pg/ml in small (0.2 ml) serum volumes that can quantify serum E2 in 98% and serum E1 in 100% of healthy postmenopausal women. Aromatase inhibitor (AI) treatment of postmenopausal women with breast cancer further reduces serum E2 by 85% and serum estrone (E1) by 80%. The wide scatter of circulating E2 in AI-treated women suggests that the degree of sustained E2 depletion, now quantifiable, may be an efficacy or safety biomarker of adjuvant AI treatment. This ultrasensitive method can also measure serum E2 in most (65%) female but not in any male mice. Further studies are warranted using this and comparable ultrasensitive liquid chromatography-mass spectrometry estrogen measurements to investigate the relationship of circulating E2 (and E1) in male, postmenopausal female, and childhood health where accurate quantification of serum estrogens was not previously feasible. This will focus on the direct impact of estrogens as well as the indirect effects of androgen aromatization on reproductive, bone, and brain tissues and, notably, the efficacy and safety of AIs in adjuvant breast cancer treatment.
Collapse
Affiliation(s)
- David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Emma Gibson
- Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK.,School of Optometry and Vision Science, University of New South Wales, NSW, Australia
| | - Susan Davis
- Women's Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Blanka Golebiowski
- School of Optometry and Vision Science, University of New South Wales, NSW, Australia
| | - Kirsty A Walters
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, Australia.,Fertility and Research Centre, School of Women's & Children's Health, University of New South Wales Sydney, NSW, Australia
| | - Reena Desai
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
19
|
Relationship of estrogen synthesis capacity in the brain with obesity and self-control in men and women. Proc Natl Acad Sci U S A 2020; 117:22962-22966. [PMID: 32868418 DOI: 10.1073/pnas.2006117117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gonadal hormones are linked to mechanisms that govern appetitive behavior and its suppression. Estrogens are synthesized from androgens by the enzyme aromatase, highly expressed in the ovaries of reproductive-aged women and in the brains of men and women of all ages. We measured aromatase availability in the amygdala using positron emission tomography (PET) with the aromatase inhibitor [11C]vorozole in a sample of 43 adult, normal-weight, overweight, or obese men and women. A subsample of 27 also completed personality measures to examine the relationship between aromatase and personality traits related to self-regulation and inhibitory control. Results indicated that aromatase availability in the amygdala was negatively associated with body mass index (BMI) (in kilograms per square meter) and positively correlated with scores of the personality trait constraint independent of sex or age. Individual variations in the brain's capacity to synthesize estrogen may influence the risk of obesity and self-control in men and women.
Collapse
|
20
|
Jonasson M, Nordeman P, Eriksson J, Wilking H, Wikström J, Takahashi K, Niwa T, Hosoya T, Watanabe Y, Antoni G, Sundström Poromaa I, Lubberink M, Comasco E. Quantification of aromatase binding in the female human brain using [
11
C]cetrozole positron emission tomography. J Neurosci Res 2020; 98:2208-2218. [DOI: 10.1002/jnr.24707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022]
Affiliation(s)
- My Jonasson
- Department of Surgical Sciences Uppsala University Uppsala Sweden
- Department of Medical Physics Uppsala University Hospital Uppsala Sweden
| | - Patrik Nordeman
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | | | - Johan Wikström
- Department of Surgical Sciences Uppsala University Uppsala Sweden
| | - Kayo Takahashi
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
| | - Takashi Niwa
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
| | - Takamitsu Hosoya
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
- Institute of Biomaterials and Bioengineering Tokyo Medical and Dental University Tokyo Japan
| | | | - Gunnar Antoni
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | | | - Mark Lubberink
- Department of Surgical Sciences Uppsala University Uppsala Sweden
- Department of Medical Physics Uppsala University Hospital Uppsala Sweden
| | - Erika Comasco
- Department of Neuroscience Science for Life Laboratory Uppsala University Uppsala Sweden
| |
Collapse
|
21
|
The effects of testosterone on the physiological response to social and somatic stressors. Psychoneuroendocrinology 2020; 117:104693. [PMID: 32413673 DOI: 10.1016/j.psyneuen.2020.104693] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/20/2020] [Accepted: 04/11/2020] [Indexed: 01/10/2023]
Abstract
Higher testosterone levels in males have previously been linked to decreased stress reactivity, but in other cases, testosterone has been reported to increase the stress response. We addressed these inconsistencies in a placebo-controlled single-dose testosterone administration study, in which 120 male participants were randomly assigned to undergo a cold-pressor test (CPT, a non-social somatic stressor), a socially evaluated cold-pressor test (SECPT, a social-somatic stressor), or a lukewarm water test (LWT, a non-stressful control condition). Throughout the experiment, blood pressure and interbeat intervals were measured continuously, and saliva samples for hormonal analyses were taken repeatedly at predefined time points. When comparing the groups treated with placebo, the SECPT elicited a larger increase in the systolic blood pressure than CPT, in agreement with previous studies. However, testosterone administration altered this pattern. Compared to placebo, testosterone increased systolic blood pressure during the CPT, whereas the opposite effect was found during the SECPT. Cortisol reactivity was not affected by testosterone administration. The CAG repeat polymorphism of the androgen receptor gene was unrelated to the effects of testosterone on the stress response, but it was correlated with blood pressure across the whole sample. Our findings demonstrate that testosterone's effects on the stress response are dependent on the social context. Testosterone's ability to flexibly influence the response to stressors may be an important mechanism through which the hormone promotes adaptive behavior. Our results are also in line with research showing that testosterone decreases social anxiety and suggest it may help to modulate the effects of stress in socially challenging situations.
Collapse
|
22
|
Perrino C, Ferdinandy P, Bøtker HE, Brundel BJJM, Collins P, Davidson SM, den Ruijter HM, Engel FB, Gerdts E, Girao H, Gyöngyösi M, Hausenloy DJ, Lecour S, Madonna R, Marber M, Murphy E, Pesce M, Regitz-Zagrosek V, Sluijter JPG, Steffens S, Gollmann-Tepeköylü C, Van Laake LW, Van Linthout S, Schulz R, Ytrehus K. Improving translational research in sex-specific effects of comorbidities and risk factors in ischaemic heart disease and cardioprotection: position paper and recommendations of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2020; 117:367-385. [PMID: 32484892 DOI: 10.1093/cvr/cvaa155] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/29/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary.,Pharmahungary Group, Hajnoczy str. 6., H-6722 Szeged, Hungary
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, 1108 HV, the Netherlands
| | - Peter Collins
- Imperial College, Faculty of Medicine, National Heart & Lung Institute, South Kensington Campus, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), Schwabachanlage 12, 91054 Erlangen, Germany
| | - Eva Gerdts
- Department for Clinical Science, University of Bergen, PO Box 7804, 5020 Bergen, Norway
| | - Henrique Girao
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, and Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, 169609, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Chris Barnard Building, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, 6410 Fannin St #1014, Houston, TX 77030, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS Via Parea, 4, I-20138 Milan, Italy
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender in Medicine, Center for Cardiovascular Research, DZHK, partner site Berlin, Geschäftsstelle Potsdamer Str. 58, 10785 Berlin, Germany.,University of Zürich, Rämistrasse 71, 8006 Zürich, Germany
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands.,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstr.35, A - 6020 Innsbruck, Austria
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Ludwigstraße 23, 35390 Giessen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9037 Tromsø, Norway
| |
Collapse
|
23
|
Estrogenic Regulation of Neuroprotective and Neuroinflammatory Mechanisms: Implications for Depression and Cognition. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/978-3-030-11355-1_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Brocca ME, Garcia-Segura LM. Non-reproductive Functions of Aromatase in the Central Nervous System Under Physiological and Pathological Conditions. Cell Mol Neurobiol 2019; 39:473-481. [PMID: 30084008 PMCID: PMC11469900 DOI: 10.1007/s10571-018-0607-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023]
Abstract
The modulation of brain function and behavior by steroid hormones was classically associated with their secretion by peripheral endocrine glands. The discovery that the brain expresses the enzyme aromatase, which produces estradiol from testosterone, expanded this traditional concept. One of the best-studied roles of brain estradiol synthesis is the control of reproductive behavior. In addition, there is increasing evidence that estradiol from neural origin is also involved in a variety of non-reproductive functions. These include the regulation of neurogenesis, neuronal development, synaptic transmission, and plasticity in brain regions not directly related with the control of reproduction. Central aromatase is also involved in the modulation of cognition, mood, and non-reproductive behaviors. Furthermore, under pathological conditions aromatase is upregulated in the central nervous system. This upregulation represents a neuroprotective and likely also a reparative response by increasing local estradiol levels in order to maintain the homeostasis of the neural tissue. In this paper, we review the non-reproductive functions of neural aromatase and neural-derived estradiol under physiological and pathological conditions. We also consider the existence of sex differences in the role of the enzyme in both contexts.
Collapse
Affiliation(s)
- Maria Elvira Brocca
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
25
|
Bereiter DA, Thompson R, Rahman M. Sex Differences in Estradiol Secretion by Trigeminal Brainstem Neurons. Front Integr Neurosci 2019; 13:3. [PMID: 30809134 PMCID: PMC6379465 DOI: 10.3389/fnint.2019.00003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/22/2019] [Indexed: 12/01/2022] Open
Abstract
Estrogen status is a significant risk factor in the development of temporomandibular joint disorders (TMD). Classically, estrogen status is thought to derive mainly from ovarian sources; however, it is well known that estradiol (E2) also is synthesized by neurons in the brain. This study tested the hypothesis that E2 is produced by neurons in trigeminal subnucleus caudalis (Vc), the principal site of termination for sensory afferents that supply the temporomandibular joint (TMJ), to modify evoked responses in a model of TMJ nociception in male and female rats. Intra-TMJ injection of the small fiber excitant, allyl isothiocyanate (AIC), increased the levels of E2 collected from microdialysis probes sites at Vc of ovariectomized (OvX) female rats, ipsilateral to the stimulus, whereas males displayed no change. Dialysate levels of E2 collected from probe sites in the contralateral Vc or cerebellum in OvX rats were not affected by TMJ stimulation. Reverse dialysis of anastrozole, an aromatase (ARO) inhibitor, via the probe reduced perfusate levels of E2 in Vc. Systemic administration of letrozole, a non-steroid ARO inhibitor, for 4 days prevented TMJ-evoked increases in masseter muscle electromyography (MMemg) activity. ARO-positive neurons were distributed mainly in superficial laminae (I-III) at Vc and cell counts revealed no significant difference between OvX and male rats. Intra-TMJ injection of AIC revealed similar numbers of ARO/Fos dual-labeled neurons in OvX and male rats. By contrast, the percentage of ARO neurons co-labeled for glutamic acid decarboxylase (GAD), the biosynthetic enzyme for GABA, was greater in OvX (35%) than male rats (14%). Few ARO-positive neurons were co-labeled for estrogen receptor alpha. These data indicate that E2 is secreted continuously by Vc neurons and that acute stimulation of TMJ nociceptors evokes further secretion in a sex-dependent manner. Reduced TMJ-evoked MMemg activity after ARO inhibition suggests that locally produced E2 by Vc neurons acts via paracrine mechanisms to modify TMJ nociception in female rats.
Collapse
Affiliation(s)
- David A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN, United States
| | - Randall Thompson
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN, United States
| | - Mostafeezur Rahman
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN, United States
| |
Collapse
|
26
|
Petersen N, London ED. Addiction and Dopamine: Sex Differences and Insights from Studies of Smoking. Curr Opin Behav Sci 2018; 23:150-159. [PMID: 30746429 PMCID: PMC6368096 DOI: 10.1016/j.cobeha.2018.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesolimbic dopaminergic function influences addiction through effects on reinforcement learning, decision-making, and impulsivity. This review covers sex differences in dopaminergic neurochemistry, their hormonal and genetic determinants, and how differences in dopaminergic tone interact with sex and/or ovarian hormone status to affect cognitive functions. Findings from research on cigarette smoking reveal sex differences in striatal and midbrain dopamine D2-type receptor availability and striatal dopamine release that suggest mechanisms of nicotine dependence, and stronger subjective responses to nicotine and efficacy of nicotine replacement therapies in male smokers than in their female counterparts. Opportunities exist to extend such efforts in studies of how sex and hormone status influence other addictions.
Collapse
Affiliation(s)
- Nicole Petersen
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA, 90024
| | - Edythe D. London
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA, 90024,Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA, 90024,Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA, 90024,Corresponding author: Dr. Edythe D. London, Department of Psychiatry and Biobehavioral Sciences, Semel Institute, University of California Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90025, USA, Tel: 310 825 0606, Fax: 310 825-0812,
| |
Collapse
|
27
|
Relation among Aromatase P450 and Tumoral Growth in Human Prolactinomas. Int J Mol Sci 2017; 18:ijms18112299. [PMID: 29104246 PMCID: PMC5713269 DOI: 10.3390/ijms18112299] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 02/01/2023] Open
Abstract
The pituitary gland is part of hypothalamic-pituitary–gonadal axis, which controls development, reproduction, and aging in humans and animals. In addition, the pituitary gland is regulated mainly by hormones and neurotransmitters released from the hypothalamus and by systemic hormones secreted by target glands. Aromatase P450, the enzyme responsible for the catabolization of aromatizable androgens to estrogens, is expressed in different parts of body, including the pituitary gland. Moreover, aromatase P450 is involved in sexual dimorphism where alteration in the level of aromatase can initiate a number of diseases in both genders. On the other hand, the direct actions of estrogens, mainly estradiol, are well known for stimulating prolactin release. Numerous studies have shown that changes in the levels of estrogens, among other factors, have been implicated in the genesis and development of prolactinoma. The pituitary gland can produce estradiol locally in several types of endocrine cells, and it is possible that aromatase could be responsible for the maintenance of the population of lactotroph cells and the modulation of the action of central or peripheral regulators. Aromatase overexpression due to inappropriate gene regulation has clinical effects such as the pathogenesis of prolactinomas. The present study reports on the synthesis of pituitary aromatase, its regulation by gonadal steroids, and the physiological roles of aromatase on pituitary endocrine cells. The involvement of aromatase in the pathogenesis of pituitary tumors, mainly prolactinomas, through the auto-paracrine production of estradiol is reviewed.
Collapse
|
28
|
Massa MG, David C, Jörg S, Berg J, Gisevius B, Hirschberg S, Linker RA, Gold R, Haghikia A. Testosterone Differentially Affects T Cells and Neurons in Murine and Human Models of Neuroinflammation and Neurodegeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2017. [PMID: 28634006 DOI: 10.1016/j.ajpath.2017.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The high female-to-male sex ratio of multiple sclerosis (MS) prevalence has continuously confounded researchers, especially in light of male patients' accelerated disease course at later stages of MS. Although multiple studies have concentrated on estrogenic mechanisms of disease modulation, fairly little attention has been paid to androgenic effects in a female system, and even fewer studies have attempted to dissociate hormonal effects on the neurodegenerative and neuroinflammatory processes of MS. Herein, we demonstrate the differential effects of hormone treatment on the acute inflammatory and chronic neurodegenerative phases of murine experimental autoimmune encephalomyelitis. Although s.c. treatment with testosterone and aromatase inhibitor applied beginning on the day of immunization ameliorated initial course of disease, similar treatment administered therapeutically exacerbated chronic disease course. Spinal cord analyses of axonal densities reflected the clinical scores of the chronic phase. In vitro, testosterone treatment not only decreased Th1 and Th17 differentiation in an aromatase-independent fashion, but also exacerbated cell death in induced pluripotent stem cell-derived primary human neurons under oxidative stress conditions in an aromatase inhibitor-dependent manner. Thus, through the alleviation of inflammatory processes and the exacerbation of neurodegenerative processes, androgens may contribute to the epidemiologic sex differentials observed in MS prevalence and course.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Christina David
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Stefanie Jörg
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Berg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Barbara Gisevius
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Sarah Hirschberg
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Ralf A Linker
- Department of Neurology, Friedrich-Alexander University-Erlangen-Nuremberg, Erlangen, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Ruhr University-Bochum, Bochum, Germany.
| |
Collapse
|
29
|
Sex hormone levels are not associated with progression of renal disease in male patients with T2DM. DIABETES & METABOLISM 2017; 43:140-145. [DOI: 10.1016/j.diabet.2016.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/05/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023]
|
30
|
Competition, testosterone, and adult neurobehavioral plasticity. PROGRESS IN BRAIN RESEARCH 2016; 229:213-238. [PMID: 27926439 DOI: 10.1016/bs.pbr.2016.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Motivation in performance is often measured via competitions. Winning a competition has been found to increase the motivation to perform in subsequent competitions. One potential neurobiological mechanism that regulates the motivation to compete involves sex hormones, such as the steroids testosterone and estradiol. A wealth of studies in both nonhuman animals and humans have shown that a rise in testosterone levels before and after winning a competition enhances the motivation to compete. There is strong evidence for acute behavioral effects in response to steroid hormones. Intriguingly, a substantial testosterone surge following a win also appears to improve an individual's performance in later contests resulting in a higher probability of winning again. These effects may occur via androgen and estrogen pathways modulating dopaminergic regions, thereby behavior on longer timescales. Hormones thus not only regulate and control social behavior but are also key to adult neurobehavioral plasticity. Here, we present literature showing hormone-driven behavioral effects that persist for extended periods of time beyond acute effects of the hormone, highlighting a fundamental role of sex steroid hormones in adult neuroplasticity. We provide an overview of the relationship between testosterone, motivation measured from objective effort, and their influence in enhancing subsequent effort in competitions. Implications for an important role of testosterone in enabling neuroplasticity to improve performance will be discussed.
Collapse
|