1
|
Hannan CT, Jeremiah MK, Idenya PM. Histological features of the Purkinje neurons of the Albino rat ( Rattus norvegicus) following letrozole administration. Anat Cell Biol 2025; 58:76-85. [PMID: 39477464 PMCID: PMC11933817 DOI: 10.5115/acb.24.088] [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: 04/03/2024] [Revised: 08/17/2024] [Accepted: 08/29/2024] [Indexed: 03/22/2025] Open
Abstract
Aromatase inhibitors are increasingly being used as adjuvant therapy for hormone-responsive cancers. These drugs may reduce the endogenous estrogen production in the cerebellum. Prolonged use has been associated with symptoms such as ataxia, poorer balance performance and diminished verbal memory, suggesting impaired cerebellar function. Thus, this study sought to outline the structural basis for the cerebellar deficits observed. Twenty-seven male rats (3 baseline, 15 experimental, 9 control) aged three months were recruited with the intervention group receiving 0.5 mg/kg of letrozole daily for 50 days by oral gavage while the control group received normal saline. Their cerebella were harvested for histological processing on days 20, 35, and 50. Photomicrographs were taken and analysed using Fiji ImageJ software. The dendritic spine densities and Purkinje linear densities were coded and analyzed using IBM SPSS Statistics version 25.0. A P-value of ≤0.05 was considered significant. A temporal decline in the Purkinje linear density as well as pyknosis and cytoplasmic eosinophilia was noted in the intervention group (P=0.165). Further, the dendritic spine density of the Purkinje neurons in the intervention group was markedly reduced (P=0.01). The reduction in the linear cell density and the dendritic spine density of the Purkinje cells following letrozole administration may provide an anatomical basis for the functional cerebellar deficits seen in chronic aromatase inhibitor use.
Collapse
Affiliation(s)
- Chaudhry Talha Hannan
- Department of Human Anatomy and Medical Physiology, University of Nairobi, Nairobi, Kenya
| | | | - Pamela Mandela Idenya
- Department of Human Anatomy and Medical Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
2
|
Rothwell ES, Viechweg SS, Prokai L, Mong JA, Lacreuse A. Oral administration of ethinyl estradiol and the brain-selective estrogen prodrug DHED in a female common marmoset model of menopause: Effects on cognition, thermoregulation, and sleep. Horm Behav 2025; 167:105670. [PMID: 39721460 DOI: 10.1016/j.yhbeh.2024.105670] [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: 04/03/2024] [Revised: 10/15/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Menopausal symptoms of sleep disturbances, cognitive deficits, and hot flashes are understudied, in part due to the lack of animal models in which they co-occur. Common marmosets (Callithrix jacchus) are valuable nonhuman primates for studying these symptoms, and we examined changes in cognition (reversal learning), sleep (48 h/wk of sleep recorded by telemetry), and thermoregulation (nose temperature in response to mild external warming) in middle-aged, surgically-induced menopausal marmosets studied at baseline, during 3-week phases of ethinyl estradiol (EE2, 4 μg/kg/day, p.o.) treatment and after EE2 withdrawal. We also assessed a brain-selective hormonal therapy devoid of estrogenic effects in peripheral tissues on the same measures (cognition, sleep, thermoregulation) after treatment with the estrogen prodrug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED, 100 μg/kg/day, p.o) and DHED withdrawal. Reversal learning performance was improved with EE2 or DHED treatment relative to phases without hormone administration, as indicated by a faster reversal of the stimulus/reward contingencies. Both EE2 and DHED increased non-REM sleep and reduced nighttime awakenings relative to baseline, but to the detriment of REM sleep which was highest at baseline. Nasal temperature in response to mild external warming was highest, and overnight core body temperature lowest, in the DHED treatment phase compared to both the EE2 and baseline phases. These results suggest that low dose estradiol, delivered either peripherally or centrally via DHED, benefits selective aspects of cognition and sleep in a marmoset menopause model. DHED appears a promising therapeutic candidate for alleviating the cognitive and sleep disruptions associated with estrogen deficiency in primates.
Collapse
Affiliation(s)
- Emily S Rothwell
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Shaun S Viechweg
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Laszlo Prokai
- Department of Pharmacology and Neuroscience, The University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Jessica A Mong
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States of America
| |
Collapse
|
3
|
Rehel S, Duivon M, Doidy F, Champetier P, Clochon P, Grellard JM, Segura-Djezzar C, Geffrelot J, Emile G, Allouache D, Levy C, Viader F, Eustache F, Joly F, Giffard B, Perrier J. Sleep oscillations related to memory consolidation during aromatases inhibitors for breast cancer. Sleep Med 2024; 121:210-218. [PMID: 39004011 DOI: 10.1016/j.sleep.2024.07.002] [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: 05/21/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024]
Abstract
Aromatase inhibitors (AIs) are associated with sleep difficulties in breast cancer (BC) patients. Sleep is known to favor memory consolidation through the occurrence of specific oscillations, i.e., slow waves (SW) and sleep spindles, allowing a dialogue between prefrontal cortex and the hippocampus. Interestingly, neuroimaging studies in BC patients have consistently shown structural and functional modifications in these two brain regions. With the aim to evaluate sleep oscillations related to memory consolidation during AIs, we collected polysomnography data in BC patients treated (AI+, n = 17) or not (AI-, n = 17) with AIs compared to healthy controls (HC, n = 21). None of the patients had received chemotherapy and radiotherapy was finished since at least 6 months, that limit the confounding effects of other treatments than AIs. Fast and slow spindles were detected during sleep stage 2 at centro-parietal and frontal electrodes respectively. SW were detected at frontal electrodes during stage 3. Here, we show lower frontal SW densities in AI + patients compared to HC. These results concord with previous reports about frontal cortical alterations in cancer following AIs administration. Moreover, AI + patients tended to have lower spindle density at C4 electrode. Regression analyses showed that, in both patient groups, spindle density at C4 electrode explained a large variance of memory performances. Slow spindle characteristics did not differ between groups and sleep oscillations characteristics of AI- patients did not differ significantly from those of both AI + patients and HC. Overall, our results add to the compelling evidence of the systemic effects of AIs previously reported in animals, with deleterious effects on cortical activity during sleep and associated memory consolidation in the current study. There is thus a need to further investigate sleep modifications during AIs administration. Longitudinal studies are needed to confirm these findings and investigation in other cancers on this topic should be conducted.
Collapse
Affiliation(s)
- S Rehel
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France.
| | - M Duivon
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France
| | - F Doidy
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France
| | - P Champetier
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France
| | - P Clochon
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France
| | - J M Grellard
- Clinical Research Department, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France
| | - C Segura-Djezzar
- Institut Normand Du Sein, Centre François Baclesse, Caen, France; Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France
| | - J Geffrelot
- Institut Normand Du Sein, Centre François Baclesse, Caen, France; Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France
| | - G Emile
- Institut Normand Du Sein, Centre François Baclesse, Caen, France; Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France
| | - D Allouache
- Institut Normand Du Sein, Centre François Baclesse, Caen, France; Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France
| | - C Levy
- Institut Normand Du Sein, Centre François Baclesse, Caen, France; Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France
| | - F Viader
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France
| | - F Eustache
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France
| | - F Joly
- Clinical Research Department, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France; Institut Normand Du Sein, Centre François Baclesse, Caen, France; Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, France; INSERM, Normandie Univ, UNICAEN, U1086 ANTICIPE, Caen, France; Cancer and Cognition Platform, Ligue Nationale Contre le Cancer, 14076, Caen, France
| | - B Giffard
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France; Cancer and Cognition Platform, Ligue Nationale Contre le Cancer, 14076, Caen, France
| | - J Perrier
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de La Mémoire Humaine, 14000, Caen, France.
| |
Collapse
|
4
|
Pasha MH, Gondal HY, Munir S, Alhussain SA, Zaki MEA. New enantioenriched β-indolyl ketones as aromatase inhibitors: Unraveling heme-ligand interactions by MD simulation and MMPBSA analysis. Arch Pharm (Weinheim) 2024; 357:e2400010. [PMID: 38578079 DOI: 10.1002/ardp.202400010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
A series of enantioenriched β-indolyl ketones as aromatase inhibitors (AI) is synthesized through the Michael-type Friedel-Crafts alkylation of indole. A highly efficient bifunctionalized amino catalyst is developed to access structurally diverse β-indolyl ketones in high yields (up to 91%) and excellent enantioselectivity (enantiomeric ratio up to 98:2). All the synthesized compounds demonstrated promising aromatase inhibitory potential, where ortho-substituted analogs (3c and 3e) were found most active with IC50 values of 0.68 and 0.90 µM, respectively. Both of these compounds exhibited significant cytotoxicity (IC50 = 0.34 and 0.37 µM) against the MCF-7 breast cancer cell line in the (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay. Molecular docking studies of the synthesized compounds demonstrate favorable binding interactions with the estrogens controlling CYP19A1 (3EQM) and metabolizing CYP3A4 (5VCC) enzymes. Molecular dynamic (MD) simulation analysis revealed the essentiality of heme-ligand interactions to build a stable protein-ligand complex. An average root mean square deviation of 0.35 nm observed during a 100-ns MD simulation and binding free energy in the range of -190 to -227 kJ/mol calculated by g_mmpbsa analysis authenticated the stability of the 3c-3EQM complex. ADMET and drug-likeness parameters supported the suitability of these indole derivatives as the drug lead to develop potent inhibitors for estrogen-dependent breast cancer.
Collapse
Affiliation(s)
- Maira Hasnain Pasha
- Institute of Chemistry, Faculty of Science, University of Sargodha, Sargodha, Pakistan
| | | | - Shanza Munir
- Institute of Chemistry, Faculty of Science, University of Sargodha, Sargodha, Pakistan
| | - Sami A Alhussain
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Mamczarz J, Lane M, Merchenthaler I. Letrozole delays acquisition of water maze task in female BALB/c mice: Possible involvement of anxiety. Horm Behav 2024; 162:105524. [PMID: 38513526 PMCID: PMC11155665 DOI: 10.1016/j.yhbeh.2024.105524] [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: 09/27/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Letrozole, an aromatase inhibitor preventing estrogen synthesis from testosterone, is used as an adjuvant therapy in estrogen receptor-positive breast cancer patients. However, like other aromatase inhibitors, it induces many side effects, including impaired cognition. Despite its negative effect in humans, results from animal models are inconsistent and suggest that letrozole can either impair or improve cognition. Here, we studied the effects of chronic letrozole treatment on cognitive behavior of adult female BALB/c mice, a relevant animal model for breast cancer studies, to develop an appropriate animal model aimed at testing therapies to mitigate side effects of letrozole. In Morris water maze, letrozole 0.1 mg/kg impaired reference learning and memory. Interestingly, most of the letrozole 0.1 mg/kg-treated mice were able to learn the new platform position in reversal training and performed similar to control mice in a reversal probe test. Results of the reversal test suggest that letrozole did not completely disrupt spatial navigation, but rather delayed acquisition of spatial information. The delay might be related to increased anxiety as suggested by increased thigmotactic behavior during the reference memory training. The learning impairment was water maze-specific since we did not observe impairment in other spatial tasks such as in Y-maze or object location test. In contrast, the dose of 0.3 mg/kg did not have effect on water maze learning and facilitated locomotor habituation and recognition in novel object recognition test. The current study shows that letrozole dose-dependently modulates behavioral response and that its effects are task-dependent.
Collapse
Affiliation(s)
- Jacek Mamczarz
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America.
| | - Malcolm Lane
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Istvan Merchenthaler
- Division of Translational Toxicology, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| |
Collapse
|
6
|
Hartman SJ, Zablocki RW, Tam RM, Palmer BW, Parker BA, Sears DD, Ahles TA, Natarajan L. Relationship of physical activity and cognitive functioning among breast cancer survivors: a cross-sectional analysis. FRONTIERS IN COGNITION 2024; 3:1332960. [PMID: 39483324 PMCID: PMC11526472 DOI: 10.3389/fcogn.2024.1332960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Introduction Cancer related cognitive decline is a common long-term side effect of cancer and its treatments among breast cancer survivors. Physical activity is a modifiable risk factor related to cognitive decline. However, existing research lacks consensus regarding the relationship between cognition and exercise as well as the impact of cancer treatments on this relationship. Baseline data from an ongoing randomized clinical trial was utilized to examine the relationship between self-reported and objectively measured cognition with physical activity. Exploratory analyses examined cancer treatments as potential moderators. Methods Breast cancer survivors (N = 253) completed a battery of neurocognitive tests, the PROMIS Cognitive abilities questionnaire, medical charts abstracted for treatment information, and wore an ActiGraph accelerometer at the waist for 7 days. Data were analyzed using multiple linear regression models. Results Participants were on average 58.5 (SD = 8.88) years old, diagnosed 3 years prior to enrollment (SD = 1.27) with 57% treated with chemotherapy and 80% receiving hormone therapy at baseline. Better self-reported cognitive ability was significantly associated with greater min of moderate to vigorous physical activity (MVPA; β = 0.070, se = 0.028, p = 0.012). There were no significant associations with any objectively measured cognitive domains. Time since diagnosis (years) was a significant moderator of MVPA and Processing Speed (β = -0.103, se = 0.043, p = 0.017). Treatment with chemotherapy and/or hormones did not significantly moderate the relationship between MVPA and any of the cognitive measures or domains. Conclusion Findings suggest that physical activity is related to self-reported cognition but not objectively measured cognition. Greater physical activity was associated with faster processing speed in participants closer in time to their cancer diagnosis. These results emphasize the need for more research to understand when cancer survivors may benefit from physical activity and what aspects of cognition may be improved.
Collapse
Affiliation(s)
- Sheri J. Hartman
- Herbert Wertheim School of Public Health, UC San Diego, La Jolla, CA, United States
- UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, United States
| | - Rong W. Zablocki
- Herbert Wertheim School of Public Health, UC San Diego, La Jolla, CA, United States
| | - Rowena M. Tam
- Herbert Wertheim School of Public Health, UC San Diego, La Jolla, CA, United States
| | - Barton W. Palmer
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
| | - Barbara A. Parker
- UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, United States
- Department of Medicine, UC San Diego, La Jolla, CA, United States
| | - Dorothy D. Sears
- UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, United States
- Department of Medicine, UC San Diego, La Jolla, CA, United States
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
- Department of Family Medicine, UC San Diego, La Jolla, CA, United States
| | - Tim A. Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Loki Natarajan
- Herbert Wertheim School of Public Health, UC San Diego, La Jolla, CA, United States
- UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, United States
| |
Collapse
|
7
|
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
|
8
|
Cea Salazar VI, Perez MD, Robison AJ, Trainor BC. Impacts of sex differences on optogenetic, chemogenetic, and calcium-imaging tools. Curr Opin Neurobiol 2024; 84:102817. [PMID: 38042130 PMCID: PMC11374099 DOI: 10.1016/j.conb.2023.102817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 12/04/2023]
Abstract
Technical innovation in neuroscience introduced powerful tools for measuring and manipulating neuronal activity via optical, chemogenetic, and calcium-imaging tools. These tools were initially tested primarily in male animals but are now increasingly being used in females as well. In this review, we consider how these tools may work differently in males and females. For example, we review sex differences in the metabolism of chemogenetic ligands and their downstream signaling effects. Optical tools more directly alter depolarization or hyperpolarization of neurons, but biological sex and gonadal hormones modulate synaptic inputs and intrinsic excitability. We review studies demonstrating that optogenetic manipulations are sometimes consistent across the rodent estrous cycle but within certain circuits; manipulations can vary across the ovarian cycle. Finally, calcium-imaging methods utilize genetically encoded calcium indicators to measure neuronal activity. Testosterone and estradiol can directly modulate calcium influx, and we consider these implications for interpreting the results of calcium-imaging studies. Together, our findings suggest that these neuroscientific tools may sometimes work differently in males and females and that users should be aware of these differences when applying these methods.
Collapse
Affiliation(s)
| | - Melvin D Perez
- Department of Physiology, University of California, Davis, CA 95616, USA
| | - A J Robison
- Department of Psychology, University of California, Davis, CA 95616, USA
| | - Brian C Trainor
- Neuroscience Graduate Group, University of California, Davis, CA 95616, USA; Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
9
|
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
|
10
|
Fleming B, Edison P, Kenny L. Cognitive impairment after cancer treatment: mechanisms, clinical characterization, and management. BMJ 2023; 380:e071726. [PMID: 36921926 DOI: 10.1136/bmj-2022-071726] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is a debilitating side effect experienced by patients with cancer treated with systemically administered anticancer therapies. With around 19.3 million new cases of cancer worldwide in 2020 and the five year survival rate growing from 50% in 1970 to 67% in 2013, an urgent need exists to understand enduring side effects with severe implications for quality of life. Whereas cognitive impairment associated with chemotherapy is recognized in patients with breast cancer, researchers have started to identify cognitive impairment associated with other treatments such as immune, endocrine, and targeted therapies only recently. The underlying mechanisms are diverse and therapy specific, so further evaluation is needed to develop effective therapeutic interventions. Drug and non-drug management strategies are emerging that target mechanistic pathways or the cognitive deficits themselves, but they need to be rigorously evaluated. Clinically, consistent use of objective diagnostic tools is necessary for accurate diagnosis and clinical characterization of cognitive impairment in patients treated with anticancer therapies. This should be supplemented with clinical guidelines that could be implemented in daily practice. This review summarizes the recent advances in the mechanisms, clinical characterization, and novel management strategies of cognitive impairment associated with treatment of non-central nervous system cancers.
Collapse
Affiliation(s)
- Ben Fleming
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Paul Edison
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Laura Kenny
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
11
|
Edwards M, Lam S, Ranjan R, Pereira M, Babbitt C, Lacreuse A. Letrozole treatment alters hippocampal gene expression in common marmosets (Callithrix jacchus). Horm Behav 2023; 147:105281. [PMID: 36434852 PMCID: PMC9839488 DOI: 10.1016/j.yhbeh.2022.105281] [Citation(s) in RCA: 2] [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: 04/30/2022] [Revised: 10/28/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
Aromatase inhibitors (AIs) are a class of drugs commonly given to patients with estrogen receptor (ER)-dependent breast cancers to reduce estrogenic stimulation. However, AIs like Letrozole are associated with negative side effects such as cognitive deficits, sleep disturbances and hot flashes. We have previously shown that these negative effects can be recapitulated in common marmosets (Callithrix jacchus) treated with Letrozole (20 μg daily) for 4 weeks and that marmosets treated with Letrozole show increased levels of estradiol in the hippocampus (Gervais et al., 2019). In order to better understand the mechanisms through which AIs affect cognitive function and increase steroid levels in the hippocampus, we used bulk, paired-end RNA-sequencing to examine differentially expressed genes among Letrozole-treated (LET; n = 8) and vehicle-treated (VEH; n = 8) male and female animals. Gene ontology results show significant reduction across hundreds of categories, some of the most significant being inflammatory response, stress response, MHC Class II protein complex binding, T-cell activation, carbohydrate binding and signaling receptor binding in LET animals. GSEA results indicate that LET females, but not LET males, show enrichment for hormonal gene sets. Based on the transcriptional changes observed, we conclude that AIs may differentially affect the sexes in part due to processes mediated by the CYP-450 superfamily. Ongoing studies will further investigate the longitudinal effects of AIs on behavior and whether AIs increase the risk of stress-induced neurodegeneration.
Collapse
Affiliation(s)
- Mélise Edwards
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA.
| | - Sam Lam
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA
| | - Ravi Ranjan
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Genomics Resource Laboratory, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Mariana Pereira
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Courtney Babbitt
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; University of Massachusetts Amherst, Department of Biology, Amherst, MA 01003, USA
| | - Agnès Lacreuse
- University of Massachusetts Amherst, Department of Psychological & Brain Sciences, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
12
|
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
|
13
|
Fainanta T, Jaroenporn S, Wititsuwankul P, Malaivijitnond S. Comparison of neuroprotective effects of dihydrotestosterone, 17β-estradiol, and Pueraria mirifica herb extract on cognitive impairment in androgen deficient male rats. Horm Behav 2022; 143:105198. [PMID: 35609404 DOI: 10.1016/j.yhbeh.2022.105198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 01/30/2023]
Abstract
This study investigated the neuroprotective effects of dihydrotestosterone (DHT), 17β-estradiol (E2), and Pueraria mirifica herb extract (PME; an alternative source of natural estrogens) on the (i) learning and memory in androgen-deficient male rats, and on the hippocampus expression levels of (ii) mRNA of genes associated with synaptic transmission and structure, neurofibrillary tangles, and amyloid plaques, and (iii) total and phosphorylated tau proteins. The four-month-old male rats were sham-operated or orchidectomized (ODX). The ODX rats were divided into four groups, and orally treated for 2 months with either 1 mL/d of distilled water or 100 mg/kg/d of PME; or subcutaneously injected with 1 mg/kg/d of DHT or 80 μg/kg/d of E2. The impairment of spatial learning behavior and memory capacity in the ODX rats was prevented by DHT, E2, and PME. Recovery of the orchidectomy-induced deterioration of the synaptic plasticity in the hippocampus of rats was ranked as E2 ≥ PME > DHT. Both DHT and PME mitigated the increased Tau3 and Tau4 mRNA levels, and Tau-5 and P-Tau Ser396 protein levels more than E2 (DHT ≥ PME > E2). Only DHT tended to decrease App mRNA expression level. In conclusion, DHT showed a stronger efficacy for mitigation of the impaired spatial learning behavior and memory capacity in androgen-deficient male rats compared to E2 and PME, and their mechanisms of action are slightly different.
Collapse
Affiliation(s)
- Taratorn Fainanta
- Biological Sciences Program, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sukanya Jaroenporn
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Patteera Wititsuwankul
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Suchinda Malaivijitnond
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
14
|
Kraynak M, Willging MM, Kuehlmann AL, Kapoor AA, Flowers MT, Colman RJ, Levine JE, Abbott DH. Aromatase Inhibition Eliminates Sexual Receptivity Without Enhancing Weight Gain in Ovariectomized Marmoset Monkeys. J Endocr Soc 2022; 6:bvac063. [PMID: 35592515 PMCID: PMC9113444 DOI: 10.1210/jendso/bvac063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Indexed: 11/19/2022] Open
Abstract
Context Ovarian estradiol supports female sexual behavior and metabolic function. While ovariectomy (OVX) in rodents abolishes sexual behavior and enables obesity, OVX in nonhuman primates decreases, but does not abolish, sexual behavior, and inconsistently alters weight gain. Objective We hypothesize that extra-ovarian estradiol provides key support for both functions, and to test this idea, we employed aromatase inhibition to eliminate extra-ovarian estradiol biosynthesis and diet-induced obesity to enhance weight gain. Methods Thirteen adult female marmosets were OVX and received (1) estradiol-containing capsules and daily oral treatments of vehicle (E2; n = 5); empty capsules and daily oral treatments of either (2) vehicle (VEH, 1 mL/kg, n = 4), or (3) letrozole (LET, 1 mg/kg, n = 4). Results After 7 months, we observed robust sexual receptivity in E2, intermediate frequencies in VEH, and virtually none in LET females (P = .04). By contrast, few rejections of male mounts were observed in E2, intermediate frequencies in VEH, and high frequencies in LET females (P = .04). Receptive head turns were consistently observed in E2, but not in VEH and LET females. LET females, alone, exhibited robust aggressive rejection of males. VEH and LET females demonstrated increased % body weight gain (P = .01). Relative estradiol levels in peripheral serum were E2 >>> VEH > LET, while those in hypothalamus ranked E2 = VEH > LET, confirming inhibition of local hypothalamic estradiol synthesis by letrozole. Conclusion Our findings provide the first evidence for extra-ovarian estradiol contributing to female sexual behavior in a nonhuman primate, and prompt speculation that extra-ovarian estradiol, and in particular neuroestrogens, may similarly regulate sexual motivation in other primates, including humans.
Collapse
Affiliation(s)
- Marissa Kraynak
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Molly M Willging
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin-Madison, Madison, WI 53715, USA
- Center for Women’s Health, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Alex L Kuehlmann
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Amita A Kapoor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Matthew T Flowers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Ricki J Colman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - David H Abbott
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
- Endocrinology-Reproductive Physiology Training Program, University of Wisconsin-Madison, Madison, WI 53715, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, USA
| |
Collapse
|
15
|
Dukes NJ, Ash H, de Faria Oliveira G, Sosa ME, Goy RW, Colman RJ, Ziegler TE. Motivational increase of androgens and behavior by infant distress calls in highly responsive common marmoset fathers, Callithrix jacchus. Horm Behav 2022; 142:105162. [PMID: 35366411 PMCID: PMC9177807 DOI: 10.1016/j.yhbeh.2022.105162] [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: 11/10/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/25/2022]
Abstract
Common marmoset fathers are highly involved in care of their infants. However, variability exists in their response to infant behavior even in paternally experienced fathers. Using infant distress cries as a motivation test, we investigated: 1. the differences in paternally experienced fathers' motivation to search for the infant vocalization stimuli; 2. the relationship between a father's motivation to search for the source of the infant cries and testosterone levels; and 3. if there is a rapid steroidogenesis pathway leading to increased testosterone and estradiol in the peripheral circulation. Only 44% of the paternally experienced fathers showed a high frequency of searching for the source of the infant distress cries. Through the use of multisteroid analysis, we found high responsive fathers had significantly higher levels of progesterone and testosterone in response to infant distress cries compared to a control stimulus with progesterone and androstenedione correlating with testosterone, while no differences were seen in low responders. The frequency to search for the infant stimuli was positively correlated with higher testosterone compared to control vocal levels. These results suggest that searching for the source of infant cries represents a motivation behavior for fathers that is activated by testosterone and reflects rapid circulating testosterone.
Collapse
Affiliation(s)
- Natalie J Dukes
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America
| | - Hayley Ash
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America
| | - Gabriela de Faria Oliveira
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America
| | - Megan E Sosa
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America
| | - Robinson W Goy
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America
| | - Ricki J Colman
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America; Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States of America
| | - Toni E Ziegler
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States of America.
| |
Collapse
|
16
|
Khosrow-Khavar F, Azoulay L, Montastruc JL, Montastruc F, Renoux C. Aromatase inhibitors and the incidence of Parkinson disease: A population-based cohort study. Cancer 2022; 128:2339-2347. [PMID: 35363379 DOI: 10.1002/cncr.34208] [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: 12/06/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND Current guidelines recommend the treatment of hormone receptor-positive breast cancer with aromatase inhibitors (AIs) and tamoxifen in the adjuvant setting. Some observational studies have raised concerns that tamoxifen may be associated with an increased risk of Parkinson disease (PD). However, no studies have directly compared the risk of PD between AIs and tamoxifen in women diagnosed with breast cancer. METHODS Using the UK Clinical Practice Research Datalink, the authors assembled a cohort of women newly diagnosed with breast cancer and newly treated with either AIs or tamoxifen between January 1, 1995, and December 31, 2017. Patients were followed 1 year after treatment initiation (ie, a 1-year lag) until an incident diagnosis of PD or were censored at death from any cause, the date of transfer out of the practice, or the end of the study period (December 31, 2018). Cox proportional hazards models with inverse probability of treatment weights were used to estimate weighted hazard ratios (HRs) and 95% confidence intervals (CIs) for PD comparing AIs with tamoxifen and accounting for more than 30 confounders. RESULTS In all, 30,140 women with nonmetastatic breast cancer were identified: 13,838 initiated AIs, and 16,302 initiated tamoxifen. Compared with tamoxifen, AIs were not associated with an increased risk of PD (HR, 0.94; 95% CI, 0.60-1.47). Consistent results were observed across all secondary and sensitivity analyses. CONCLUSIONS In this large observational study, the use of AIs, in comparison with tamoxifen, was not associated with an increased risk of PD in women diagnosed with nonmetastatic breast cancer in a real-world setting. LAY SUMMARY Previous studies have indicated that tamoxifen may increase the risk of Parkinson disease in the treatment of breast cancer. However, no studies have directly compared the risk of Parkinson disease between aromatase inhibitors and tamoxifen. This study included 30,140 women diagnosed with breast cancer and treated with aromatase inhibitors or tamoxifen. Overall, compared with tamoxifen, aromatase inhibitors were not associated with an increased risk of Parkinson disease in women diagnosed with breast cancer. This study provides an important addition to the comparative safety profile of aromatase inhibitors and tamoxifen in the treatment of breast cancer.
Collapse
Affiliation(s)
- Farzin Khosrow-Khavar
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Québec, Canada
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada
| | - Laurent Azoulay
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Québec, Canada
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Québec, Canada
| | - Jean-Louis Montastruc
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Centre d'Investigation Clinique 1436, Team PEPSS (Pharmacologie en Population Cohortes et Biobanques), Toulouse University Hospital, Toulouse, France
| | - François Montastruc
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Centre d'Investigation Clinique 1436, Team PEPSS (Pharmacologie en Population Cohortes et Biobanques), Toulouse University Hospital, Toulouse, France
| | - Christel Renoux
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Québec, Canada
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| |
Collapse
|
17
|
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: 13] [Impact Index Per Article: 4.3] [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
|
18
|
Abstract
The role of central estrogen in cognitive, metabolic, and reproductive health has long fascinated the lay public and scientists alike. In the last two decades, insight into estrogen signaling in the brain and its impact on female physiology is beginning to catch up with the vast information already established for its actions on peripheral tissues. Using newer methods to manipulate estrogen signaling in hormone-sensitive brain regions, neuroscientists are now identifying the molecular pathways and neuronal subtypes required for controlling sex-dependent energy allocation. However, the immense cellular complexity of these hormone-sensitive brain regions makes it clear that more research is needed to fully appreciate how estrogen modulates neural circuits to regulate physiological and behavioral end points. Such insight is essential for understanding how natural or drug-induced hormone fluctuations across lifespan affect women's health.
Collapse
Affiliation(s)
- Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| |
Collapse
|
19
|
PET imaging of brain aromatase in humans and rhesus monkeys by 11C-labeled cetrozole analogs. Sci Rep 2021; 11:23623. [PMID: 34880350 PMCID: PMC8654920 DOI: 10.1038/s41598-021-03063-8] [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: 07/26/2021] [Accepted: 11/22/2021] [Indexed: 11/08/2022] Open
Abstract
Aromatase is an estrogen synthetic enzyme that plays important roles in brain functions. To quantify aromatase expression in the brain by positron emission tomography (PET), we had previously developed [11C]cetrozole, which showed high specificity and affinity. To develop more efficient PET tracer(s) for aromatase imaging, we synthesized three analogs of cetrozole. We synthesized meta-cetrozole, nitro-cetrozole, and iso-cetrozole, and prepared the corresponding 11C-labeled tracers. The inhibitory activities of these three analogs toward aromatase were evaluated using marmoset placenta, and PET imaging of brain aromatase was performed using the 11C-labeled tracers in monkeys. The most promising analog in the monkey study, iso-cetrozole, was evaluated in the human PET study. The highest to lowest inhibitory activity of the analogs toward aromatase in the microsomal fraction from marmoset placenta was in the following order: iso-cetrozole, nitro-cetrozole, cetrozole, and meta-cetrozole. This order showed good agreement with the order of the binding potential (BP) of each 11C-labeled analog to aromatase in the rhesus monkey brain. A human PET study using [11C]iso-analog showed a similar distribution pattern of binding as that of [11C]cetrozole. The time-activity curves showed that elimination of [11C]iso-cetrozole from brain tissue was faster than that of 11C-cetrozole, indicating more rapid metabolism of [11C]iso-cetrozole. [11C]Cetrozole has preferable metabolic stability for brain aromatase imaging in humans, although [11C]iso-cetrozole might also be useful to measure aromatase level in living human brain because of its high binding potential.
Collapse
|
20
|
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
|
21
|
Shaw GA. Mitochondria as the target for disease related hormonal dysregulation. Brain Behav Immun Health 2021; 18:100350. [PMID: 34746877 PMCID: PMC8554460 DOI: 10.1016/j.bbih.2021.100350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/12/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondria play an important role in the synthesis of steroid hormones, including the sex hormone estrogen. Sex-specific regulation of these hormones is important for phenotypic development and downstream, sex-specific activational effects in both brain and behavior. First, mitochondrial contribution to the synthesis of estrogen, followed by a discussion of the signaling interactions between estrogen and the mitochondria will be reviewed. Next, disorders with an established sex difference related to aging, mood, and cognition will be examined. Finally, review of mitochondria as a biomarker of disease and data supporting efforts in targeting mitochondria as a therapeutic target for the amelioration of these disorders will be discussed. Taken together, this review aims to assess the influence of E2 on mitochondrial function within the brain via exploration of E2-ER interactions within neural mitochondria and how they may act to influence the development and presentation of neurodegenerative and neurocognitive diseases with known sex differences.
Collapse
Affiliation(s)
- Gladys A. Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
22
|
Popp I, Rau A, Kellner E, Reisert M, Fennell JT, Rothe T, Nieder C, Urbach H, Egger K, Grosu AL, Kaller CP. Hippocampus-Avoidance Whole-Brain Radiation Therapy Is Efficient in the Long-Term Preservation of Hippocampal Volume. Front Oncol 2021; 11:714709. [PMID: 34490112 PMCID: PMC8417356 DOI: 10.3389/fonc.2021.714709] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose With improved life expectancy, preventing neurocognitive decline after cerebral radiotherapy is gaining more importance. Hippocampal damage has been considered the main culprit for cognitive deficits following conventional whole-brain radiation therapy (WBRT). Here, we aimed to determine to which extent hippocampus-avoidance WBRT (HA-WBRT) can prevent hippocampal atrophy compared to conventional WBRT. Methods and Materials Thirty-five HA-WBRT and 48 WBRT patients were retrospectively selected, comprising a total of 544 contrast-enhanced T1-weighted magnetic resonance imaging studies, longitudinally acquired within 24 months before and 48 months after radiotherapy. HA-WBRT patients were treated analogously to the ongoing HIPPORAD-trial (DRKS00004598) protocol with 30 Gy in 12 fractions and dose to 98% of the hippocampus ≤ 9 Gy and to 2% ≤ 17 Gy. WBRT was mainly performed with 35 Gy in 14 fractions or 30 Gy in 10 fractions. Anatomical images were segmented and the hippocampal volume was quantified using the Computational Anatomy Toolbox (CAT), including neuroradiological expert review of the segmentations. Results After statistically controlling for confounding variables such as age, gender, and total intracranial volume, hippocampal atrophy was found after both WBRT and HA-WBRT (p < 10-6). However, hippocampal decline across time following HA-WBRT was approximately three times lower than following conventional WBRT (p < 10-6), with an average atrophy of 3.1% versus 8.5% in the first 2 years after radiation therapy, respectively. Conclusion HA-WBRT is a therapeutic option for patients with multiple brain metastases, which can effectively and durably minimize hippocampal atrophy compared to conventional WBRT.
Collapse
Affiliation(s)
- Ilinca Popp
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Rau
- Department of Neuroradiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elias Kellner
- Medical Physics, Department of Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Reisert
- Medical Physics, Department of Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jamina Tara Fennell
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Rothe
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carsten Nieder
- Department of Oncology and Palliative Medicine, Nordland Hospital, Bodø, Norway.,Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Horst Urbach
- Department of Neuroradiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Karl Egger
- Department of Neuroradiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca Ligia Grosu
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph P Kaller
- Department of Neuroradiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
23
|
Park HG, Kim JH, Dancer AN, Kothapalli KS, Brenna JT. The aromatase inhibitor letrozole restores FADS2 function in ER+ MCF7 human breast cancer cells. Prostaglandins Leukot Essent Fatty Acids 2021; 171:102312. [PMID: 34303883 DOI: 10.1016/j.plefa.2021.102312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022]
Abstract
PURPOSE Plasticity in fatty acid metabolism is increasingly recognized as a major feature influencing cancer progression and efficacy of treatments. Estrogen receptor positive MCF7 human breast cancer cells have long been known to have no FADS2-mediated Δ6-desaturase activity. Our objective was to examine the effect of estrogen and the "antiestrogen" aromatase inhibitor letrozole, on Δ5- and Δ6-desaturase synthesized fatty acids in vitro. METHODS Eicosa-11,14-dienoic acid (20:2n-6), a known substrate for both FADS1 and FADS2, was used as a sentinel of relative FADS2 and FADS1 activity. MCF7 cells and four additional estrogen responsive wild type cell lines (HepG2, SK-N-SH, Y79 and Caco2) were studied. FAME were quantified by GC-FID and structures identified by GCCACI-MS/MS. RESULTS In all five cell lines, estrogen caused a dose dependent decrease in sciadonic acid (5,11,14-20:3, ScA) via apparent inhibition of FADS1 activity, and had no effect on FADS2 catalyzed synthesis of dihomo-gamma linolenic acid (8,11,14-20:3; DGLA). In MCF7 cells, letrozole caused a dose dependent increase in FADS2-catalyzed DGLA synthesis, which plateaued in SK-N-SH cells. CONCLUSION Letrozole restores Δ6-desaturase mediated synthesis of the anti-inflammatory PGE1-precursor DGLA in vitro and is the first endocrine-active agent to have opposing effects on FADS1 and FADS2 catalyzed activities.
Collapse
Affiliation(s)
- Hui Gyu Park
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Jae Hun Kim
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Andrew N Dancer
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Kumar S Kothapalli
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - J Thomas Brenna
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA.
| |
Collapse
|
24
|
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
|
25
|
Hartman SJ, Weiner LS, Natarajan L, Sears DD, Palmer BW, Parker B, Ahles T, Irwin ML, Au K. A randomized trial of physical activity for cognitive functioning in breast cancer survivors: Rationale and study design of I Can! Improving Cognition After Cancer. Contemp Clin Trials 2021; 102:106289. [PMID: 33503496 PMCID: PMC8009833 DOI: 10.1016/j.cct.2021.106289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/11/2020] [Accepted: 01/19/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Difficulties with cognition are extremely common among breast cancer survivors and can significantly impact quality of life, daily functioning, and ability to return to work. One promising intervention is increasing physical activity, as it has been effective in improving cognition in non-cancer populations. Few physical activity intervention trials with cognition outcomes have included cancer survivors. This project builds upon our previous work indicating that increased physical activity can improve objectively measured processing speed and self-reported cognition among breast cancer survivors. METHODS The I Can! study will examine whether a physical activity intervention improves cognition among 250 post-treatment breast cancer survivors (Stages I-III, <5 years post-treatment) who are reporting cognitive difficulties. This 2-arm randomized controlled trial comparing a 6-month physical activity intervention (Exercise Group) to a health & wellness attention-comparison condition (Health & Wellness Group) will examine intervention effects on cognition (at 3 and 6 months) and maintenance of effects at 12 months. The primary aim is to investigate the impact of exercise on objectively measured processing speed and self-reported cognition. Secondary aims are to investigate maintenance of cognitive changes and examine candidate biological mechanisms and psychological mediators. CONCLUSION The I Can! study will contribute to the scientific, public health, and survivorship intervention literature by providing new information on the impact of physical activity for cognitive impairment in breast cancer survivors. Findings from this study will inform guidelines for physical activity to improve the lives of millions of breast cancer survivors.
Collapse
Affiliation(s)
- Sheri J Hartman
- Herbert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA, USA; UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, USA.
| | - Lauren S Weiner
- Herbert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA, USA; UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Loki Natarajan
- Herbert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA, USA; UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Dorothy D Sears
- Herbert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA, USA; UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, USA; Department of Medicine, UC San Diego, La Jolla, CA, USA; College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Barton W Palmer
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Barbara Parker
- UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, USA; Department of Medicine, UC San Diego, La Jolla, CA, USA
| | - Tim Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melinda L Irwin
- Department of Chronic Disease Epidemiology, Yale School of Public Health, USA
| | - Kaylene Au
- UC San Diego Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Dijkshoorn ABC, van Stralen HE, Sloots M, Schagen SB, Visser-Meily JMA, Schepers VPM. Prevalence of cognitive impairment and change in patients with breast cancer: A systematic review of longitudinal studies. Psychooncology 2021; 30:635-648. [PMID: 33533166 PMCID: PMC8248098 DOI: 10.1002/pon.5623] [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: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Patients with breast cancer face cognitive impairment that affects their quality of life; partially attributable to treatment. Our aim was to detail the prevalence and change of cognitive impairment during the course of treatment. We also investigated the effect of therapy (chemotherapy [CT]) vs. radiotherapy and/or endocrine therapy vs. healthy controls). METHODS This article reviews longitudinal cohort studies published to date in Medline and Embase that (i) assess cognition before and after therapy, (ii) report prevalence cognitive impairment or change, and (iii) use standardized and valid neuropsychological tests. We used the original authors' criteria for cognitive impairment. RESULTS The title and abstract of 891 articles were screened, resulting in the identification of 90 potentially relevant articles while applying the eligibility criteria. After full-text examination, 17 studies were included. Prevalence of cognitive impairment range from 25% before therapy, through 24% after therapy to 21% at maximal 1-year follow-up (FU). Compared to their pretreatment cognitive functioning, 24% of patients decline after treatment and 24% at 1-year FU. Some studies also reported cognitive improvement showing that 15% and 31% of patients improve, respectively. In general, patients undergoing CT have a higher chance of cognitive impairment and decline than no-CT patients and healthy controls. CONCLUSIONS This study shows that one out of four breast cancer patients shows cognitive impairment prior to treatment administration CT and a significant number of patients decline during the course of disease, suggesting that cognitive impairment is not exclusively related to CT and/or no-CT therapies. This study shows that assessment of cognitive functioning, ideally over time, is crucial and may help the implementation of personalized rehabilitation pathways.
Collapse
Affiliation(s)
- Aicha B C Dijkshoorn
- Department of Rehabilitation, Physical Therapy Science & Sports, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Haike E van Stralen
- Department of Rehabilitation, Physical Therapy Science & Sports, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maurits Sloots
- Department of Rehabilitation, Physical Therapy Science & Sports, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sanne B Schagen
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Johanna M A Visser-Meily
- Department of Rehabilitation, Physical Therapy Science & Sports, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands.,Center of Excellence for Rehabilitation Medicine, Brain Center Rudolf Magnus, University Medical Center Utrecht and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
| | - Vera P M Schepers
- Department of Rehabilitation, Physical Therapy Science & Sports, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands.,Center of Excellence for Rehabilitation Medicine, Brain Center Rudolf Magnus, University Medical Center Utrecht and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
| |
Collapse
|
27
|
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: 8] [Impact Index Per Article: 2.0] [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
|
28
|
Taylor CM, Pritschet L, Jacobs EG. The scientific body of knowledge - Whose body does it serve? A spotlight on oral contraceptives and women's health factors in neuroimaging. Front Neuroendocrinol 2021; 60:100874. [PMID: 33002517 PMCID: PMC7882021 DOI: 10.1016/j.yfrne.2020.100874] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Women constitute half of the world's population, yet neuroscience research does not serve the sexes equally. Fifty years of preclinical animal evidence documents the tightly-coupled relationship between our endocrine and nervous systems, yet human neuroimaging studies rarely consider how endocrine factors shape the structural and functional architecture of the human brain. Here, we quantify several blind spots in neuroimaging research, which overlooks aspects of the human condition that impact women's health (e.g. the menstrual cycle, hormonal contraceptives, pregnancy, menopause). Next, we illuminate potential consequences of this oversight: today over 100 million women use oral hormonal contraceptives, yet relatively few investigations have systematically examined whether disrupting endogenous hormone production impacts the brain. We close by presenting a roadmap for progress, highlighting the University of California Women's Brain Initiative which is addressing unmet needs in women's health research.
Collapse
Affiliation(s)
- Caitlin M Taylor
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, United States.
| | - Laura Pritschet
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, United States
| | - Emily G Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, United States; Neuroscience Research Institute, University of California, Santa Barbara, United States.
| |
Collapse
|
29
|
Taxier LR, Gross KS, Frick KM. Oestradiol as a neuromodulator of learning and memory. Nat Rev Neurosci 2020; 21:535-550. [PMID: 32879508 PMCID: PMC8302223 DOI: 10.1038/s41583-020-0362-7] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 12/24/2022]
Abstract
Although hormones such as glucocorticoids have been broadly accepted in recent decades as general neuromodulators of memory processes, sex steroid hormones such as the potent oestrogen 17β-oestradiol have been less well recognized by the scientific community in this capacity. The predominance of females in studies of oestradiol and memory and the general (but erroneous) perception that oestrogens are 'female' hormones have probably prevented oestradiol from being more widely considered as a key memory modulator in both sexes. Indeed, although considerable evidence supports a crucial role for oestradiol in regulating learning and memory in females, a growing body of literature indicates a similar role in males. This Review discusses the mechanisms of oestradiol signalling and provides an overview of the effects of oestradiol on spatial, object recognition, social and fear memories. Although the primary focus is on data collected in females, effects of oestradiol on memory in males will be discussed, as will sex differences in the molecular mechanisms that regulate oestrogenic modulation of memory, which may have important implications for the development of future cognitive therapeutics.
Collapse
Affiliation(s)
- Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Kellie S Gross
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
30
|
Brown AMC, Gervais NJ. Role of Ovarian Hormones in the Modulation of Sleep in Females Across the Adult Lifespan. Endocrinology 2020; 161:5879359. [PMID: 32735650 PMCID: PMC7450669 DOI: 10.1210/endocr/bqaa128] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022]
Abstract
Ovarian hormones, including 17β-estradiol, are implicated in numerous physiological processes, including sleep. Beginning at puberty, girls report more sleep complaints than boys, which is maintained throughout the reproductive life stage. Sleep problems are exacerbated during the menopausal transition, evidenced by greater risk for sleep disorders. There is emerging evidence that menopause-associated hormone loss contributes to this elevated risk, but age is also an important factor. The extent to which menopause-associated sleep disturbance persists into postmenopause above and beyond the effects of age remains unknown. Untreated sleep disturbances have important implications for cognitive health, as they are emerging as risk factors for dementia. Given that sleep loss impairs memory, an important knowledge gap concerns the role played by menopause-associated hormone loss in exacerbating sleep disturbance and, ultimately, cognitive function in aging women. In this review, we take a translational approach to illustrate the contribution of ovarian hormones in maintaining the sleep-wake cycle in younger and middle-aged females, with evidence implicating 17β-estradiol in supporting the memory-promoting effects of sleep. Sleep physiology is briefly reviewed before turning to behavioral and neural evidence from young females linking 17β-estradiol to sleep-wake cycle maintenance. Implications of menopause-associated 17β-estradiol loss is also reviewed before discussing how ovarian hormones may support the memory-promoting effects of sleep, and why menopause may exacerbate pathological aging via effects on sleep. While still in its infancy, this research area offers a new sex-based perspective on aging research, with a focus on a modifiable risk factor for pathological aging.
Collapse
Affiliation(s)
- Alana M C Brown
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Nicole J Gervais
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Correspondence: Nicole J. Gervais, University of Toronto, Department of Psychology, 100 St. George Street, Toronto, ON, Canada M5S 3G3. E-mail:
| |
Collapse
|
31
|
Saldanha CJ. Estrogen as a Neuroprotectant in Both Sexes: Stories From the Bird Brain. Front Neurol 2020; 11:497. [PMID: 32655477 PMCID: PMC7324752 DOI: 10.3389/fneur.2020.00497] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Estrogens such as estradiol (E2) are potent effectors of neural structure and function via peripheral and central synthesis. In the zebra finch (Taeniopygia guttata), neural E2 synthesis is among the highest reported in homeotherms due to the abundant constitutive expression of aromatase (E-synthase) in discrete neuronal pools across the forebrain. Following penetrating or concussive trauma, E2 synthesis increases even further via the induced expression of aromatase in reactive astrocytes around the site of damage. Injury-associated astrocytic aromatization occurs in the brains of both sexes regardless of the site of injury and can remain elevated for weeks following trauma. Interestingly, penetrating injury induces astrocytic aromatase more rapidly in females compared to males, but this sex difference is not detectable 24 h posttrauma. Indeed, unilateral penetrating injury can increase E2 content 4-fold relative to the contralateral uninjured hemisphere, suggesting that glial aromatization may be a powerful source of neural E2 available to circuits. Glial aromatization is neuroprotective as inhibition of injury-induced aromatase increases neuroinflammation, gliosis, necrosis, apoptosis, and infarct size. These effects are ameliorated upon replacement with E2, suggesting that the songbird may have evolved a rapidly responsive neurosteroidogenic system to protect vulnerable brain circuits. The precise signals that induce aromatase expression in astrocytes include elements of the inflammatory cascade and underscore the sentinel role of the innate immune system as a crucial effector of trauma-associated E2 provision in the vertebrate brain. This review will describe the inductive signals of astroglial aromatase and the neuroprotective role for glial E2 synthesis in the adult songbird brains of both sexes.
Collapse
Affiliation(s)
- Colin J Saldanha
- Departments of Neuroscience, Biology, Psychology & The Center for Behavioral Neuroscience, American University, Washington, DC, United States
| |
Collapse
|
32
|
WANG C, LIANG L. [Reconsideration of the third-generation non-steroidal aromatase inhibitors in pediatrics]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020; 49:275-282. [PMID: 32762160 PMCID: PMC8800707 DOI: 10.3785/j.issn.1008-9292.2020.04.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/21/2020] [Indexed: 11/15/2022]
Abstract
Aromatase is the rate-limiting enzyme in estrogen biosynthesis. The third generation aromatase inhibitors (AIs), represented by letrozoleand and anastrozole, can combine with aromatase, effectively reducing the estrogen level in the body. Because of its high efficiency, selectivity and reversibility, it has been used in the treatment of McCune-Albright syndrome, familial male-limited precocious puberty, gynecomastia, and adolescent boy with short stature. The good efficacy and safety of AIs have been observed. However, so far the drug instructions of AIs usually do not show indications for children; there are risks of adverse reactions involving liver and kidney function, lipid metabolism, hyperandrogenemia and bone metabolism; especially the long-term effects on reproductive system and bone metabolism are still not clear. Therefore, it is necessary to prescribe it carefully and follow up closely. It was not recommended that AIs be routinely used to improve adult height of adolescent boy with short stature. And more clinical evidences are needed for the safety and effectiveness of AIs prescribed in pediatrics.
Collapse
Affiliation(s)
| | - Li LIANG
- 梁黎(1956-), 女, 硕士, 教授, 博士生导师, 主要从事儿科内分泌研究; E-mail:
;
https://orcid.org/0000-0001-7048-8898
| |
Collapse
|
33
|
Mamlouk GM, Dorris DM, Barrett LR, Meitzen J. Sex bias and omission in neuroscience research is influenced by research model and journal, but not reported NIH funding. Front Neuroendocrinol 2020; 57:100835. [PMID: 32070715 PMCID: PMC7225067 DOI: 10.1016/j.yfrne.2020.100835] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/31/2022]
Abstract
Neuroscience research has historically demonstrated sex bias that favors male over female research subjects, as well as sex omission, which is the lack of reporting sex. Here we analyzed the status of sex bias and omission in neuroscience research published across six different journals in 2017. Regarding sex omission, 16% of articles did not report sex. Regarding sex bias, 52% of neuroscience articles reported using both males and females, albeit only 15% of articles using both males and females reported assessing sex as an experimental variable. Overrepresentation of the sole use of males compared to females persisted (26% versus 5%, respectively). Sex bias and omission differed across research models, but not by reported NIH funding status. Sex omission differed across journals. These findings represent the latest information regarding the complex status of sex in neuroscience research and illustrate the continued need for thoughtful and informed action to enhance scientific discovery.
Collapse
Affiliation(s)
- Gabriella M Mamlouk
- Dept. of Biological Sciences, NC State University, Raleigh, NC, United States
| | - David M Dorris
- Dept. of Biological Sciences, NC State University, Raleigh, NC, United States
| | - Lily R Barrett
- Dept. of Psychology, Florida State University, Tallahassee, FL, United States
| | - John Meitzen
- Dept. of Biological Sciences, NC State University, Raleigh, NC, United States; Center for Human Health and the Environment, NC State University, Raleigh, NC, United States.
| |
Collapse
|
34
|
Branigan GL, Soto M, Neumayer L, Rodgers K, Brinton RD. Association Between Hormone-Modulating Breast Cancer Therapies and Incidence of Neurodegenerative Outcomes for Women With Breast Cancer. JAMA Netw Open 2020; 3:e201541. [PMID: 32207833 PMCID: PMC7093781 DOI: 10.1001/jamanetworkopen.2020.1541] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/26/2020] [Indexed: 12/17/2022] Open
Abstract
Importance The association between exposure to hormone-modulating therapy (HMT) as breast cancer treatment and neurodegenerative disease (NDD) is unclear. Objective To determine whether HMT exposure is associated with the risk of NDD in women with breast cancer. Design, Setting, and Participants This retrospective cohort study used the Humana claims data set from January 1, 2007, to March 31, 2017. The Humana data set contains claims from private-payer and Medicare insurance data sets from across the United States with a population primarily residing in the Southeast. Patient claims records were surveyed for a diagnosis of NDD starting 1 year after breast cancer diagnosis for the duration of enrollment in the claims database. Participants were 57 843 women aged 45 years or older with a diagnosis of breast cancer. Patients were required to be actively enrolled in Humana claims records for 6 months prior to and at least 3 years after the diagnosis of breast cancer. The analyses were conducted between January 1 and 15, 2020. Exposure Hormone-modulating therapy (selective estrogen receptor modulators, estrogen receptor antagonists, and aromatase inhibitors). Main Outcomes and Measures Patients receiving HMT for breast cancer treatment were identified. Survival analysis was used to determine the association between HMT exposure and diagnosis of NDD. A propensity score approach was used to minimize measured and unmeasured selection bias. Results Of the 326 485 women with breast cancer in the Humana data set between 2007 and 2017, 57 843 met the study criteria. Of these, 18 126 (31.3%; mean [SD] age, 76.2 [7.0] years) received HMT, whereas 39 717 (68.7%; mean [SD] age, 76.8 [7.0] years) did not receive HMT. Mean (SD) follow-up was 5.5 (1.8) years. In the propensity score-matched population, exposure to HMT was associated with a decrease in the number of women who received a diagnosis of NDD (2229 of 17 878 [12.5%] vs 2559 of 17 878 [14.3%]; relative risk, 0.89; 95% CI, 0.84-0.93; P < .001), Alzheimer disease (877 of 17 878 [4.9%] vs 1068 of 17 878 [6.0%]; relative risk, 0.82; 95% CI, 0.75-0.90; P < .001), and dementia (1862 of 17 878 [10.4%] vs 2116 of 17 878 [11.8%]; relative risk, 0.88; 95% CI, 0.83-0.93; P < .001). The number needed to treat was 62.51 for all NDDs, 93.61 for Alzheimer disease, and 69.56 for dementia. Conclusions and Relevance Among patients with breast cancer, tamoxifen and steroidal aromatase inhibitors were associated with a decrease in the number who received a diagnosis of NDD, specifically Alzheimer disease and dementia.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
- MD-PhD Training Program, University of Arizona College of Medicine, Tucson
| | - Maira Soto
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
| | - Leigh Neumayer
- Department of Surgery, University of Arizona College of Medicine, Tucson
- Department of Obstetrics and Gynecology, University of Arizona College of Medicine, Tucson
| | - Kathleen Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson
- Department of Neurology, University of Arizona College of Medicine, Tucson
| |
Collapse
|
35
|
Merchenthaler I, Stennett CA, Haughey B, Puche A, Urbanski HF. Establishment of a non-human primate model for menopausal hot flushes. EC GYNAECOLOGY 2020; 9:https://www.ecronicon.com/ecgy/pdf/establishment-of-a-non-human-primate-model-for-menopausal-hot-flushes.pdf. [PMID: 32743563 PMCID: PMC7394307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Menopause affects the quality of life of millions of women. With modern lifespan the postmenopausal attenuation of circulating estrogen levels can negatively impact a women's life for 30-40 years. The major hypoestrogenic consequence is hot flushes but decline in cognitive function, sleep disorders, depression/anxiety, cardiovascular disease, and osteoporosis are also characteristic for the menopause. Current treatments of hot flushes include estrogen therapy alone or in combination with progestins, soy products, and serotonin and norepinephrine reuptake inhibitors. However, with the exception of estrogens, none of these have satisfactory efficacy. But estrogens come with the unwanted side effects in the periphery, including stimulation of the uterus and breast leading to elevated cancer risk. Therefore, a tremendous effort has been devoted to developing safer therapies and the research has utilized classic rodent models of hot flush with considerable limitations. As hot flushes are primate-specific symptoms, the development of a non-invasive primate hot flush model would have a tremendous impact on drug development. Therefore, our aim was to develop such a non-human primate (NHP) model a hot flush that both recapitulates flushes women experience and is minimally invasive. We investigated if recent developments in thermal imaging have made it possible to accurately monitor skin temperature via camera imaging. In this study, the skin temperature of an ovariectomized rhesus monkey was measured continuously with an infrared camera in a freely moving animal over long time period. Following mapping skin temperatures of several areas of the neck and face we found that the nose of the monkeys showed that largest changes in skin temperature. In the ovariectomized monkey the temperature of the skin on the nose shows up to 9 °C elevations representing hot flushes. In the untreated monkey, hot flushes occurred more frequently in late afternoon/early evening hours than in the morning and last for several minutes. We observed 58 flushes in the 64 evenings of observation. The average number of hot flushes was 0.51 per evening. Oral administration of biotin (niacin) for seven days exaggerated the number of hot flushes to 2.43 per evening. Oral treatment with estradiol benzoate prevented hot flushes and only 2 flushes were detected in the 12 evenings after treatment, averaging 0.17 per evening. The development of this NHP model of hot flush provides great hope for utilizing it for future drug development and mechanistic studied.
Collapse
Affiliation(s)
- Istvan Merchenthaler
- Department of Epidemiology and Public Health, University of Maryland Baltimore
- Department of Anatomy and Neurobiology, University of Maryland Baltimore
| | | | - Bethany Haughey
- Department of Epidemiology and Public Health, University of Maryland Baltimore
| | - Adam Puche
- Department of Anatomy and Neurobiology, University of Maryland Baltimore
| | - Henryk F. Urbanski
- Oregon National Primate Research Center, Oregon Health and Science University
| |
Collapse
|
36
|
Baumgartner NE, Grissom EM, Pollard KJ, McQuillen SM, Daniel JM. Neuroestrogen-Dependent Transcriptional Activity in the Brains of ERE-Luciferase Reporter Mice following Short- and Long-Term Ovariectomy. eNeuro 2019; 6:ENEURO.0275-19.2019. [PMID: 31575604 PMCID: PMC6795557 DOI: 10.1523/eneuro.0275-19.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/03/2019] [Accepted: 09/22/2019] [Indexed: 12/22/2022] Open
Abstract
Previous work has demonstrated that estrogen receptors are transcriptionally active in the absence of ovarian estrogens. The current work aims to determine whether brain-derived estrogens influence estrogen receptor-dependent transcription after short- or long-term loss of ovarian function. Experiments were conducted using estrogen response element (ERE)-Luciferase reporter mice, which express the gene for luciferase driven by consensus ERE, allowing for the quantification of ERE-dependent transcription. Brain regions examined were hippocampus, cortex, and hypothalamus. In Experiment 1, short-term (10 d) ovariectomy had no impact on ERE-dependent transcription across brain regions compared with sham surgery. In Experiment 2, chronic intracerebroventricular administration of the aromatase inhibitor letrozole significantly decreased transcriptional activity in 10-d-old ovariectomized mice across brain regions, indicating that the sustained transcription in short-term ovariectomized mice is mediated at least in part via actions of neuroestrogens. Additionally, intracerebroventricular administration of estrogen receptor antagonist ICI-182,780 blocked transcription in 10-d-old ovariectomized mice across brain regions, providing evidence that sustained transcription in ovariectomized mice is estrogen receptor dependent. In Experiment 3, long-term (70 d) ovariectomy significantly decreased ERE-dependent transcription across brain regions, though some residual activity remained. In Experiment 4, chronic intracerebroventricular letrozole administration had no impact on transcription in 70 d ovariectomized mice across brain regions, indicating that the residual ERE-dependent transcription in long-term ovariectomized mice is not mediated by neuroestrogens. Overall, the results indicate that ERE-dependent transcription in the brain continues after ovariectomy and that the actions of neuroestrogens contribute to the maintenance of ERE-dependent transcription in the brain following short-term, but not long-term, loss of ovarian function.
Collapse
Affiliation(s)
| | - Elin M Grissom
- Neuroscience Program
- Department of Psychology, Tulane University, New Orleans, Louisiana 70118
| | | | | | - Jill M Daniel
- Neuroscience Program
- Tulane Brain Institute
- Department of Psychology, Tulane University, New Orleans, Louisiana 70118
| |
Collapse
|