1
|
Mashimo A, Oshida R, Oka Y, Kawabata S, Takasu C, Nihei K, Kojima T, Kanemura N, Murata K. Hormonal fluctuations in rodent models using 4-vinylcyclohexene diepoxide: A systematic review and meta-analysis. Horm Behav 2025; 168:105680. [PMID: 39826372 DOI: 10.1016/j.yhbeh.2025.105680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/01/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
An animal model of 4-vinylcyclohexene diepoxide (VCD)-induced premature ovarian failure was developed to mimic menopause; this model has been used in various field studies. However, detailed reports on the rodent model using VCD are lacking, and the animal species used, administration methods, and hormonal fluctuations in the creation of the VCD model have not been comprehensively elucidated. The aim of this study was to systematically review these aspects of the rodent model using VCD and elucidate its characteristics. Thirty-two studies were extracted; rats and mice (66 %/44 %) are the most commonly used animal species. In most of the studies involving mice, a dose of 160 mg/kg was administered, whereas in most rat studies, doses of 80 mg/kg and 160 mg/kg were administered. On most mice studies (70 %), the most frequently applied dosage duration was 15 days. In most rat studies (63 %), the most frequently applied duration was 25 days, followed by 14 and 15 days in 30 % of the studies. Meta-analysis indicated that the mouse model using VCD simulates significant hormonal changes, such as estradiol (E2), anti-Müllerian hormone (AMH), and follicle stimulating hormone (FSH) changes. In conclusion, although the VCD model has demonstrated significant promise in replicating menopausal hormonal conditions, further systematic studies are required to fully understand its potential applications and refine its methodologies. This comprehensive review of existing literature highlights the need for continued research to expand the use of the VCD model in diverse medical fields.
Collapse
Affiliation(s)
- Aoi Mashimo
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Ryuga Oshida
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Yuichiro Oka
- Graduate School of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Sora Kawabata
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Chiharu Takasu
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Kota Nihei
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Takuma Kojima
- Department of Health and Social Services, Health and Social Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Naohiko Kanemura
- Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, Saitama, Japan
| | - Kenji Murata
- Department of Physical Therapy, School of Health and Social Services, Saitama Prefectural University, Saitama, Japan.
| |
Collapse
|
2
|
Yu Y, Zhang T, Li X, Yu T, Meng F, Luan Y, Cong H, Wu X. Apigenin Improves Ovarian Dysfunction Induced by 4-Vinylcyclohexene Diepoxide via the AKT/FOXO3a Pathway. Cell Biochem Funct 2024; 42:e70015. [PMID: 39511926 DOI: 10.1002/cbf.70015] [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: 08/13/2024] [Revised: 10/10/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Perimenopausal syndrome is a significant issue that disturbs women's metabolism, mood and quality of life. Apigenin (4',5,7-trihydroxyflavone) is a natural flavonoid that exhibits antioxidant, anti-inflammatory and anticancer effects. The present study aims to investigate the effect of apigenin on perimenopausal syndrome by combining bioinformatics analysis with in vivo experiments. The mouse model with perimenopausal syndrome was established using 4-vinylcyclohexene diepoxide (VCD) treatment. Apigenin alleviated VCD-induced disorder of estrous cycle and shrinkage of ovarian tissue. The reduction of anti-Muller hormone and the increase of follicle stimulation hormone and luteinizing hormone triggered by VCD were reversed by apigenin in a dose-dependent manner. Apigenin suppressed the VCD-induced decrease of primordial, primary, secondary and antral follicle number in ovarian tissue. Oxidative stress in ovarian tissue was activated by VCD treatment through increasing the reactive oxygen species production. High concentration of apigenin significantly reversed the alteration induced by VCD. Apigenin alleviated VCD-induced cell apoptosis through regulating Bax, Bcl-2, cleaved PARP1 and caspase-3. Furthermore, the phosphorylation of AKT and FOXO3a was inhibited by VCD and activated by apigenin in a dose-dependent manner. Collectively, apigenin effectively mitigates the ovarian dysfunction through suppressing oxidative stress and apoptosis via the AKT/FOXO3a signaling pathway.
Collapse
Affiliation(s)
- Yang Yu
- College of Acupuncture Moxibustion and Tuina, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Gynecology, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianchan Zhang
- College of Acupuncture Moxibustion and Tuina, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Gynecology, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xin Li
- Department of Organization, Qiqihar Medical University, Qiqihar, China
| | - Tianyang Yu
- College of Acupuncture Moxibustion and Tuina, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Acupuncture, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fanci Meng
- College of Acupuncture Moxibustion and Tuina, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yifeng Luan
- Department of Gynecology, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Huifang Cong
- College of Acupuncture Moxibustion and Tuina, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Gynecology, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiuhong Wu
- College of Acupuncture Moxibustion and Tuina, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Knox N, Yasrebi A, Caramico D, Wiersielis K, Samuels BA, Roepke TA. The Interaction Of Diet-Induced Obesity And Chronic Stress In A Mouse Model Of Menopause. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622997. [PMID: 39605499 PMCID: PMC11601223 DOI: 10.1101/2024.11.11.622997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Menopause is characterized by the cessation of ovarian hormone production. During postmenopause, cisgender women face increased risks of obesity, cognitive decline, and mood disorder. Mood disorders are associated with exposure to chronic stress. We investigated the combined effects of a high-fat diet (HFD) and chronic stress exposure in a mouse model of menopause using 4-vinylcyclohexene diepoxide (VCD), a selective ovotoxicant that gradually depletes ovarian follicles and hormones. Starting at 6 months, 82 female WT C57BL/6J mice received saline or VCD (130 mg/kg i.p.) 5 days per week for 3 weeks. One month after injection, mice were fed either low-fat diet (LFD) or HFD for 8 weeks followed by 6 weeks of chronic variable mild stress (CVMS). Post-CVMS, mice were either processed for gene expression of the anterodorsal BNST or behavior tests to assess cognitive and anxiety-related behaviors. Plasma samples were collected to analyze metabolic hormones and corticosterone levels. VCD-treated HFD-fed mice had higher fat and body mass, and elevated fasting glucose levels compared to controls and more pronounced avoidance behaviors and cognitive impairments. LFD-fed, VCD-treated mice exhibited less exploration of novel objects and open spaces compared to OIL and HFD counterparts. VCD elevated corticosterone levels on LFD and increased BNST Pacap gene expression on HFD. These findings highlight cognitive repercussions of estrogen deficiency and suggest a potential protective effect of a HFD against some of the adverse outcomes associated with menopause. Our study emphasizes the importance of considering dietary and hormonal interactions in the development of therapeutic strategies.
Collapse
|
4
|
Ramli NZ, Yahaya MF, Fahami NAM, Hamezah HS, Bakar ZHA, Arrozi AP, Yanagisawa D, Tooyama I, Singh M, Damanhuri HA. Spatial learning and memory impairment at the post-follicular depletion state is associated with reduced hippocampal glucose uptake. Exp Gerontol 2024; 197:112607. [PMID: 39389279 DOI: 10.1016/j.exger.2024.112607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The menopausal transition is a complex neuroendocrine aging process affecting brain structure and metabolic function. Such changes are consistent with neurological sequelae noted following the menopausal transition, including cognitive deficits. Although studies in rodent models of the menopause revealed changes in learning and memory, little is known about the structural and metabolic changes in the brain regions serving the cognitive function in these models. The administration 4-vinylcyclohexene diepoxide (VCD) in laboratory animals results in follicular depletion, and thus, is a powerful translational tool that models the human menopause. In the studies presented here, we evaluated behavior, brain structure, and metabolism in young female rats administered with either VCD or vehicle for 15 days across the early, mid, and post-follicular depletion states at 1-, 2-, and 3-months post-final injection, respectively. Additionally, we evaluated the serum hormonal profile and ovarian follicles based on the estrous cycle pattern. Positron emission tomography (PET) was utilized to determine regional brain glucose metabolism in the hippocampus, medial prefrontal cortex, and striatum. Subsequently, the rats were euthanized for ex-vivo magnetic resonance imaging (MRI) to assess regional brain volumes. VCD-induced rats at the post-follicular depleted time points had diminished spatial learning and memory as well as reduced hippocampal glucose uptake. Additionally, VCD-induced rats at post-follicular depletion time points had marked reductions in estradiol, progesterone, and anti-mullerian hormone with an increase in follicle-stimulating hormone. These rats also exhibited fewer ovarian follicles, indicating that substantial ovarian function loss during post-follicular time points impairs the female rats' spatial learning/memory abilities and triggers the metabolic changes in the hippocampus.
Collapse
Affiliation(s)
- Nur Zuliani Ramli
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Jalan Hospital, 47000 Sungai Buloh, Selangor, Malaysia.
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia.
| | - Nur Azlina Mohd Fahami
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia.
| | - Hamizah Shahirah Hamezah
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia.
| | - Zulzikry Hafiz Abu Bakar
- Medical Innovation Research Centre, Shiga University of Medical Sciences, Seta Tsukinowacho, Otsu 520-2192, Shiga, Japan.
| | - Aslina Pahrudin Arrozi
- Medical Innovation Research Centre, Shiga University of Medical Sciences, Seta Tsukinowacho, Otsu 520-2192, Shiga, Japan.
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Centre, Shiga University of Medical Sciences, Seta Tsukinowacho, Otsu 520-2192, Shiga, Japan.
| | - Ikuo Tooyama
- Medical Innovation Research Centre, Shiga University of Medical Sciences, Seta Tsukinowacho, Otsu 520-2192, Shiga, Japan.
| | - Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago Maywood, IL 60153, USA.
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia.
| |
Collapse
|
5
|
Sommer G, Rodríguez López C, Hirschkorn A, Calimano G, Marques-Lopes J, Milner TA, Glass MJ. Estrogen Receptor Beta Agonist Influences Presynaptic NMDA Receptor Distribution in the Paraventricular Hypothalamic Nucleus Following Hypertension in a Mouse Model of Perimenopause. BIOLOGY 2024; 13:819. [PMID: 39452127 PMCID: PMC11505520 DOI: 10.3390/biology13100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/01/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Women become susceptible to hypertension as they transition to menopause (i.e., perimenopause); however, the underlying mechanisms are unclear. Animal studies using an accelerated ovarian failure (AOF) model of peri-menopause (peri-AOF) demonstrate that peri-AOF hypertension is associated with increased postsynaptic NMDA receptor plasticity in the paraventricular hypothalamic nucleus (PVN), a brain area critical for blood pressure regulation. However, recent evidence indicates that presynaptic NMDA receptors also play a role in neural plasticity. Here, using immuno-electron microscopy, we examine the influence of peri-AOF hypertension on the subcellular distribution of the essential NMDA GluN1 receptor subunit in PVN axon terminals in peri-AOF and in male mice. Hypertension was produced by 14-day slow-pressor angiotensin II (AngII) infusion. The involvement of estrogen signaling was investigated by co-administering an estrogen receptor beta (ERß) agonist. Although AngII induced hypertension in both peri-AOF and male mice, peri-AOF females showed higher cytoplasmic GluN1 levels. In peri-AOF females, activation of ERß blocked hypertension and increased plasmalemmal GluN1 in axon terminals. In contrast, stimulation of ERß did not inhibit hypertension or influence presynaptic GluN1 localization in males. These results indicate that sex-dependent recruitment of presynaptic NMDA receptors in the PVN is influenced by ERß signaling in mice during early ovarian failure.
Collapse
Affiliation(s)
- Garrett Sommer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Claudia Rodríguez López
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Adi Hirschkorn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Gianna Calimano
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
- Center for Translational Health and Medical Biotechnology Research (TBIO)/Health Research Network (RISE-HEALTH), ESS, Polytechnic of Porto, R. Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| |
Collapse
|
6
|
Chen ZK, Liu YY, Zhou JC, Chen GH, Liu CF, Qu WM, Huang ZL. Insomnia-related rodent models in drug discovery. Acta Pharmacol Sin 2024; 45:1777-1792. [PMID: 38671193 PMCID: PMC11335876 DOI: 10.1038/s41401-024-01269-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/24/2024] [Indexed: 04/28/2024] Open
Abstract
Despite the widespread prevalence and important medical impact of insomnia, effective agents with few side effects are lacking in clinics. This is most likely due to relatively poor understanding of the etiology and pathophysiology of insomnia, and the lack of appropriate animal models for screening new compounds. As the main homeostatic, circadian, and neurochemical modulations of sleep remain essentially similar between humans and rodents, rodent models are often used to elucidate the mechanisms of insomnia and to develop novel therapeutic targets. In this article, we focus on several rodent models of insomnia induced by stress, diseases, drugs, disruption of the circadian clock, and other means such as genetic manipulation of specific neuronal activity, respectively, which could be used to screen for novel hypnotics. Moreover, important advantages and constraints of some animal models are discussed. Finally, this review highlights that the rodent models of insomnia may play a crucial role in novel drug development to optimize the management of insomnia.
Collapse
Affiliation(s)
- Ze-Ka Chen
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yuan-Yuan Liu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ji-Chuan Zhou
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Saeki K, Ha D, Chang G, Mori H, Yoshitake R, Wu X, Wang J, Wang YZ, Wang X, Tzeng T, Shim HJ, Neuhausen SL, Chen S. Perimenopausal and Menopausal Mammary Glands In A 4-Vinylcyclohexene Diepoxide Mouse Model. J Mammary Gland Biol Neoplasia 2024; 29:15. [PMID: 39017946 PMCID: PMC11254995 DOI: 10.1007/s10911-024-09569-x] [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: 02/02/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
As both perimenopausal and menopausal periods are recognized critical windows of susceptibility for breast carcinogenesis, development of a physiologically relevant model has been warranted. The traditional ovariectomy model causes instant removal of the entire hormonal repertoire produced by the ovary, which does not accurately approximate human natural menopause with gradual transition. Here, we characterized the mammary glands of 4-vinylcyclohexene diepoxide (VCD)-treated animals at different time points, revealing that the model can provide the mammary glands with both perimenopausal and menopausal states. The perimenopausal gland showed moderate regression in ductal structure with no responsiveness to external hormones, while the menopausal gland showed severe regression with hypersensitivity to hormones. Leveraging the findings on the VCD model, effects of a major endocrine disruptor (polybrominated diphenyl ethers, PBDEs) on the mammary gland were examined during and after menopausal transition, with the two exposure modes; low-dose, chronic (environmental) and high-dose, subacute (experimental). All conditions of PBDE exposure did not augment or compromise the macroscopic ductal reorganization resulting from menopausal transition and/or hormonal treatments. Single-cell RNA sequencing revealed that the experimental PBDE exposure during the post-menopausal period caused specific transcriptomic changes in the non-epithelial compartment such as Errfi1 upregulation in fibroblasts. The environmental PBDE exposure resulted in similar transcriptomic changes to a lesser extent. In summary, the VCD mouse model provides both perimenopausal and menopausal windows of susceptibility for the breast cancer research community. PBDEs, including all tested models, may affect the post-menopausal gland including impacts on the non-epithelial compartments.
Collapse
Affiliation(s)
- Kohei Saeki
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Desiree Ha
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Gregory Chang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Hitomi Mori
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Ryohei Yoshitake
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Jinhui Wang
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Yuan-Zhong Wang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Xiaoqiang Wang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Tony Tzeng
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Hyun Jeong Shim
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Shiuan Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
8
|
Cakir C, Kuspinar G, Aslan K, Bozyigit C, Kasapoglu I, Dirican M, Uncu G, Avci B. Dehydroepiandrosterone modulates the PTEN/PI3K/AKT signaling pathway to alleviate 4-vinylcyclohexene diepoxide-induced premature ovarian insufficiency in rats. Exp Anim 2024; 73:319-335. [PMID: 38494723 PMCID: PMC11254495 DOI: 10.1538/expanim.23-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Dehydroepiandrosterone (DHEA) is frequently integrated as an adjuvant in over a quarter of controlled ovarian hyperstimulation (COH) protocols, despite the ongoing debate regarding its impact. This study aimed to evaluate the efficacy and mechanism of action of DHEA on ovarian follicular development and ovarian response in rats with varying ovarian reserves. The study involved 75 rats categorized into 15 distinct groups. The ovarian tissues of rats in both the normal ovarian reserve group and the premature ovarian insufficiency (POI) group, induced by 4-vinylcyclohexene diepoxide (VCD) injection, were subjected to histomorphological and biochemical analyses following the administration of DHEA, either alone or in combination with COH. Follicle counting was performed on histological sections obtained from various tissues. Serum concentrations of anti-Müllerian hormone (AMH) and the quantification of specific proteins in ovarian tissue, including phosphatase and tensin homolog of chromosome 10 (PTEN), phosphoinositide 3-kinase (PI3K), phosphorylated protein kinase B (pAKT), cyclooxygenase 2 (COX-2), caspase-3, as well as assessments of total antioxidant status and total oxidant status, were conducted employing the ELISA method. The impact of DHEA exhibited variability based on ovarian reserve. In the POI model, DHEA augmented follicular development and ovarian response to the COH protocol by upregulating the PTEN/PI3K/AKT signaling pathway, mitigating apoptosis, inflammation, and oxidative stress, contrary to its effects in the normal ovarian reserve group. In conclusion, it has been determined that DHEA may exert beneficial effects on ovarian stimulation response by enhancing the initiation of primordial follicles and supporting antral follicle populations.
Collapse
Affiliation(s)
- Cihan Cakir
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Goktan Kuspinar
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Kiper Aslan
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Cengiz Bozyigit
- Department of Medical Biochemistry, Bursa City Hospital, Doğanköy District, Nilüfer Bursa, 16110, Türkiye
| | - Isil Kasapoglu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Melahat Dirican
- Department of Medical Biochemistry, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Gurkan Uncu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Berrin Avci
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| |
Collapse
|
9
|
Hinks A, Dalton BE, Mashouri P, Flewwelling LD, Pyle WG, Cheng AJ, Power GA. Time course changes in in vivo muscle mechanical function and Ca 2+ regulation of force following experimentally induced gradual ovarian failure in mice. Exp Physiol 2024; 109:711-728. [PMID: 38500268 PMCID: PMC11061627 DOI: 10.1113/ep091735] [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: 12/21/2023] [Accepted: 02/05/2024] [Indexed: 03/20/2024]
Abstract
The abrupt cessation of ovarian hormone release is associated with declines in muscle contractile function, yet the impact of gradual ovarian failure on muscle contractility across peri-, early- and late-stage menopause remains unclear. In this study, a 4-vinylcyclohexene diepoxide (VCD)-induced ovarian failure mouse model was used to examine time course changes in muscle mechanical function. Plantar flexors of female mice (VCD: n = 10; CON: n = 8) were assessed at 40 (early perimenopause), 80 (late perimenopause), 120 (menopause onset) and 176 (late menopause) days post-initial VCD injection. A torque-frequency relationship was established across a range of frequencies (10-200 Hz). Isotonic dynamic contractions were elicited against relative loads (10-80% maximal isometric torque) to determine the torque-velocity-power relationship. Mice then performed a fatigue task using intermittent 100 Hz isometric contractions until torque dropped by 60%. Recovery of twitch, 10 Hz and 100 Hz torque were tracked for 10 min post-task failure. Additionally, intact muscle fibres from the flexor digitorum brevis underwent a fatigue task (50 repetitions at 70 Hz), and 10 and 100 Hz tetanic [Ca2+] were monitored for 10 min afterward. VCD mice exhibited 16% lower twitch torque than controls across all time points. Apart from twitch torque, 10 Hz torque and 10 Hz tetanic [Ca2+], where VCD showed greater values relative to pre-fatigue during recovery, no significant differences were observed between control and VCD mice during recovery. These results indicate that gradual ovarian failure has minimal detriments to in vivo muscle mechanical function, with minor alterations observed primarily for low-frequency stimulation during recovery from fatigue.
Collapse
Affiliation(s)
- Avery Hinks
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| | - Benjamin E. Dalton
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| | - Parastoo Mashouri
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| | - Luke D. Flewwelling
- Muscle Health Research Centre, School of Kinesiology and Health Sciences, Faculty of HealthYork UniversityTorontoCanada
| | - William Glen Pyle
- IMPART Team Canada, Dalhousie MedicineDalhousie UniversitySaint JohnNew BrunswickCanada
| | - Arthur J. Cheng
- Muscle Health Research Centre, School of Kinesiology and Health Sciences, Faculty of HealthYork UniversityTorontoCanada
| | - Geoffrey A. Power
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| |
Collapse
|
10
|
Li Y, He R, Qin X, Zhu Q, Ma L, Liang X. Transcriptome analysis during 4-vinylcyclohexene diepoxide exposure-induced premature ovarian insufficiency in mice. PeerJ 2024; 12:e17251. [PMID: 38646488 PMCID: PMC11032656 DOI: 10.7717/peerj.17251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/26/2024] [Indexed: 04/23/2024] Open
Abstract
The occupational chemical 4-Vinylcyclohexene diepoxide (VCD) is a reproductively toxic environmental pollutant that causes follicular failure, leading to premature ovarian insufficiency (POI), which significantly impacts a woman's physical health and fertility. Investigating VCD's pathogenic mechanisms can offer insights for the prevention of ovarian impairment and the treatment of POI. This study established a mouse model of POI through intraperitoneal injection of VCD into female C57BL/6 mice for 15 days. The results were then compared with those of the control group, including a comparison of phenotypic characteristics and transcriptome differences, at two time points: day 15 and day 30. Through a comprehensive analysis of differentially expressed genes (DEGs), key genes were identified and validated some using RT-PCR. The results revealed significant impacts on sex hormone levels, follicle number, and the estrous cycle in VCD-induced POI mice on both day 15 and day 30. The DEGs and enrichment results obtained on day 15 were not as significant as those obtained on day 30. The results of this study provide a preliminary indication that steroid hormone synthesis, DNA damage repair, and impaired oocyte mitosis are pivotal in VCD-mediated ovarian dysfunction. This dysfunction may have been caused by VCD damage to the primordial follicular pool, impairing follicular development and aggravating ovarian damage over time, making it gradually difficult for the ovaries to perform their normal functions.
Collapse
Affiliation(s)
- Yi Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ruifen He
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Qin
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Qinying Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Liangjian Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaolei Liang
- Gansu Provincial Clinical Research Center for Gynecological Oncology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Mashouri P, Saboune J, Pyle WG, Power GA. Effects of chemically induced ovarian failure on single muscle fiber contractility in a mouse model of menopause. Maturitas 2024; 180:107885. [PMID: 38061310 DOI: 10.1016/j.maturitas.2023.107885] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE Menopause is associated with impaired skeletal muscle contractile function. The temporal and mechanistic bases of this dysfunction are unknown. Using a mouse model of menopause, we identified how gradual ovarian failure affects single muscle fiber contractility. STUDY DESIGN Ovarian failure was chemically induced over 120 days, representing the perimenopausal transition. Mice were sacrificed and soleus and extensor digitorum longus muscles were dissected and chemically permeabilized for single fiber mechanical testing. MAIN OUTCOME MEASURES Muscle fiber contractility was assessed via force, rate of force redevelopment, instantaneous stiffness, and calcium sensitivity. RESULTS Peak force and cross-sectional area of the soleus were, respectively, ~33 % and ~24 % greater following ovarian failure compared with controls (p < 0.05) with no differences in force produced by the extensor digitorum longus across groups (p > 0.05). Upon normalizing force to cross-sectional area there were no differences across groups (p > 0.05). Following ovarian failure, rate of force redevelopment of single fibers from the soleus was ~33 % faster compared with controls. There was no shift in the midpoint of the force‑calcium curve between groups or muscles (p > 0.05). However, following ovarian failure, Type I fibers from the soleus had a higher calcium sensitivity between pCa values of 4.5 and 6.2 compared with controls (p < 0.05), with no differences for Type II fibers or the extensor digitorum longus (p > 0.05). CONCLUSIONS In our model of menopause, alterations to muscle contractility were less evident than in ovariectomized models. This divergence across models highlights the importance of better approximating the natural trajectory of menopause during and after the transitional phase of ovarian failure on neuromuscular function.
Collapse
Affiliation(s)
- Parastoo Mashouri
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada.
| | - Jinan Saboune
- IMPART Team Canada, Dalhousie Medicine, Dalhousie University, Saint John, New Brunswick, Canada; Laboratory of Molecular Cardiology, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
| | - W Glen Pyle
- IMPART Team Canada, Dalhousie Medicine, Dalhousie University, Saint John, New Brunswick, Canada; Laboratory of Molecular Cardiology, Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
| | - Geoffrey A Power
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada.
| |
Collapse
|
12
|
Abi-Ghanem C, Salinero AE, Smith RM, Kelly RD, Belanger KM, Richard RN, Paul AS, Herzog AA, Thrasher CA, Rybka KA, Riccio D, Gannon OJ, Kordit D, Kyaw NR, Mansour FM, Groom E, Brooks HL, Robison LS, Pumiglia K, Zuloaga DG, Zuloaga KL. Effects of Menopause and High Fat Diet on Metabolic Outcomes in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 101:1177-1194. [PMID: 39302361 DOI: 10.3233/jad-231332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Background About two-thirds of those with Alzheimer's disease (AD) are women, most of whom are post-menopausal. Menopause accelerates dementia risk by increasing the risk for metabolic, cardiovascular, and cerebrovascular diseases. Mid-life metabolic disease (obesity, diabetes/prediabetes) is a well-known risk factor for dementia. A high fat diet can lead to poor metabolic health in both humans and rodents. Objective Our goal was to determine the effects of a high fat diet on metabolic outcomes in the AppNL-F knock-in mouse model of AD and assess the effects of menopause. Methods First, 3-month-old AppNL-F and WT female mice were placed on either a control or a high fat diet until 10 months of age then assessed for metabolic outcomes. Next, we did a more extensive assessment in AppNL-F mice that were administered VCD (4-vinylcyclohexene diepoxide) or vehicle (oil) and placed on a control or high fat diet for 7 months. VCD was used to model menopause by causing accelerated ovarian failure. Results Compared to WT controls, AD female mice had worse glucose intolerance. Menopause led to metabolic impairment (weight gain and glucose intolerance) and further exacerbated obesity in response to a high fat diet. There were interactions between diet and menopause on some metabolic health serum biomarkers and the expression of hypothalamic markers related to energy balance. Conclusions This work highlights the need to model endocrine aging in animal models of dementia and will contribute to further understanding the interaction between menopause and metabolic health in the context of AD.
Collapse
Affiliation(s)
- Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Rachel M Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Kasey M Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Riane N Richard
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Aaron S Paul
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Ava A Herzog
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Christina A Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Krystyna A Rybka
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - David Riccio
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - David Kordit
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Febronia M Mansour
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Emily Groom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Heddwen L Brooks
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lisa S Robison
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Kevin Pumiglia
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
13
|
Zucon Bacelar AC, Momesso NR, Pederro FHM, Gonçalves A, Ervolino E, Chaves-Neto AH, Biguetti CC, Matsumoto MA. Aged and induced-premature ovarian failure mouse models affect diestrus profile and ovarian features. PLoS One 2023; 18:e0284887. [PMID: 38064437 PMCID: PMC10707698 DOI: 10.1371/journal.pone.0284887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/11/2023] [Indexed: 12/18/2023] Open
Abstract
Sex hormones exert a wide influence on several systems of the human body, especially in women, who undergo intense changes in the trans and postmenopausal periods. Different experimental models are used to mimic these conditions; however, the impact on hormonal profile may be different. This study aimed to analyze and compare vaginal cytology of different post-estropausal mice models, along with their microscopical ovarian features. Forty-six C57BL/6J female mice with the ages of 4, 6 and 18 months at the beginning of the experiment, weighing about 25-28 grams, constituted five groups: NC-(negative control) animals with no treatment, OVX-SHAM-sham ovariectomized, OVX-ovariectomized, VCD-medicated with 160 mg/kg/day of 4-vinylcyclohexene diepoxide via IP for 20 consecutive days, and Aged-senescent mice under physiological estropause. Euthanasia was performed at different periods for the removal of the ovaries, and after diestrus was confirmed by vaginal cytology for 10 consecutive days. For daily vaginal cytology, morphological and histomorphometric microscopic analyzes were performed. Aged mice presented significant increased neutrophils when compared to VCD group, as well as increased cornified epithelial cells when compared to OVX mice, and also increased nucleated epithelial cells when compared to VCD and OVX. NC and OVX-SHAM ovaries presented innumerous follicles at different stages of development, while VCD showed marked follicular atresia, depleted of primordial or developing follicles and a predominance of interstitial cells. The ovaries of aged mice were predominantly constituted by corpus luteum degenerated into corpus albicans, with rare antral follicles. All analyzed models led to different permanent diestrus profiles caused by each model, as indicated by ovarian features. This should be carefully considered when choosing a post-estropausal experimental model, in order to better correlate this challenging phase of female's life with physiological/pathological conditions.
Collapse
Affiliation(s)
- Ana Carolina Zucon Bacelar
- Department of Diagnostics and Surgery, São Paulo State University—Unesp, Araçatuba, School of Dentistry, São Paulo, Brazil
| | - Nataira Regina Momesso
- Department of Diagnostics and Surgery, São Paulo State University—Unesp, Araçatuba, School of Dentistry, São Paulo, Brazil
| | - Felipe Haddad Martim Pederro
- Department of Basic Sciences, São Paulo State University—Unesp, Araçatuba School of Dentistry, São Paulo, Brazil
| | - Alaíde Gonçalves
- Department of Basic Sciences, São Paulo State University—Unesp, Araçatuba School of Dentistry, São Paulo, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, São Paulo State University—Unesp, Araçatuba School of Dentistry, São Paulo, Brazil
| | | | - Claudia Cristina Biguetti
- School of Podiatric Medicine, The University of Texas at Rio Grande Valley (UTRGV), Rio Grande Valley, Texas, United States of America
| | - Mariza Akemi Matsumoto
- Department of Basic Sciences, São Paulo State University—Unesp, Araçatuba School of Dentistry, São Paulo, Brazil
| |
Collapse
|
14
|
Youngblood H, Schoenlein PV, Pasquale LR, Stamer WD, Liu Y. Estrogen dysregulation, intraocular pressure, and glaucoma risk. Exp Eye Res 2023; 237:109725. [PMID: 37956940 PMCID: PMC10842791 DOI: 10.1016/j.exer.2023.109725] [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: 09/01/2023] [Revised: 10/20/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Characterized by optic nerve atrophy due to retinal ganglion cell (RGC) death, glaucoma is the leading cause of irreversible blindness worldwide. Of the major risk factors for glaucoma (age, ocular hypertension, and genetics), only elevated intraocular pressure (IOP) is modifiable, which is largely regulated by aqueous humor outflow through the trabecular meshwork. Glucocorticoids such as dexamethasone have long been known to elevate IOP and lead to glaucoma. However, several recent studies have reported that steroid hormone estrogen levels inversely correlate with glaucoma risk, and that variants in estrogen signaling genes have been associated with glaucoma. As a result, estrogen dysregulation may contribute to glaucoma pathogenesis, and estrogen signaling may protect against glaucoma. The mechanism for estrogen-related protection against glaucoma is not completely understood but likely involves both regulation of IOP homeostasis and neuroprotection of RGCs. Based upon its known activities, estrogen signaling may promote IOP homeostasis by affecting extracellular matrix turnover, focal adhesion assembly, actin stress fiber formation, mechanosensation, and nitric oxide production. In addition, estrogen receptors in the RGCs may mediate neuroprotective functions. As a result, the estrogen signaling pathway may offer a therapeutic target for both IOP control and neuroprotection. This review examines the evidence for a relationship between estrogen and IOP and explores the possible mechanisms by which estrogen maintains IOP homeostasis.
Collapse
Affiliation(s)
- Hannah Youngblood
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Patricia V Schoenlein
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA; Department of Radiology and Georgia Cancer Center, Augusta University, Augusta, GA, USA; Department of Surgery, Augusta University, Augusta, GA, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W Daniel Stamer
- Department of Ophthalmology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA; Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA.
| |
Collapse
|
15
|
Platholi J, Marongiu R, Park L, Yu F, Sommer G, Weinberger R, Tower W, Milner TA, Glass MJ. Hippocampal glial inflammatory markers are differentially altered in a novel mouse model of perimenopausal cerebral amyloid angiopathy. Front Aging Neurosci 2023; 15:1280218. [PMID: 38035277 PMCID: PMC10684955 DOI: 10.3389/fnagi.2023.1280218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Dementia is often characterized by age-dependent cerebrovascular pathology, neuroinflammation, and cognitive deficits with notable sex differences in risk, disease onset, progression and severity. Women bear a disproportionate burden of dementia, and the onset of menopause (i.e., perimenopause) may be a critical period conferring increased susceptibility. However, the contribution of early ovarian decline to the neuroinflammatory processes associated with cerebrovascular dementia risks, particularly at the initial stages of pathology that may be more amenable to proactive intervention, is unknown. To better understand the influence of early ovarian failure on dementia-associated neuroinflammation we developed a model of perimenopausal cerebral amyloid angiopathy (CAA), an important contributor to dementia. For this, accelerated ovarian failure (AOF) was induced by 4-vinylcyclohexene diepoxide (VCD) treatment to isolate early-stage ovarian failure comparable to human perimenopause (termed "peri-AOF") in transgenic SWDI mice expressing human vasculotropic mutant amyloid beta (Aβ) precursor protein, that were also tested at an early stage of amyloidosis. We found that peri-AOF SWDI mice showed increased astrocyte activation accompanied by elevated Aβ in select regions of the hippocampus, a brain system involved in learning and memory that is severely impacted during dementia. However, although SWDI mice showed signs of increased hippocampal microglial activation and impaired cognitive function, this was not further affected by peri-AOF. In sum, these results suggest that elevated dysfunction of key elements of the neurovascular unit in select hippocampal regions characterizes the brain pathology of mice at early stages of both CAA and AOF. However, neurovascular unit pathology may not yet have passed a threshold that leads to further behavioral compromise at these early periods of cerebral amyloidosis and ovarian failure. These results are consistent with the hypothesis that the hormonal dysregulation associated with perimenopause onset represents a stage of emerging vulnerability to dementia-associated neuropathology, thus providing a selective window of opportunity for therapeutic intervention prior to the development of advanced pathology that has proven difficult to repair or reverse.
Collapse
Affiliation(s)
- Jimcy Platholi
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
- Anesthesiology Department, Weill Cornell Medicine, New York, NY, United States
| | - Roberta Marongiu
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
- Neurological Surgery Department, Weill Cornell Medicine, New York, NY, United States
- Genetic Medicine Department, Weill Cornell Medicine, New York, NY, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Laibaik Park
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Fangmin Yu
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Garrett Sommer
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Rena Weinberger
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - William Tower
- Neurological Surgery Department, Weill Cornell Medicine, New York, NY, United States
| | - Teresa A. Milner
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
- Harold and Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States
| | - Michael J. Glass
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| |
Collapse
|
16
|
Gannon OJ, Naik JS, Riccio D, Mansour FM, Abi-Ghanem C, Salinero AE, Kelly RD, Brooks HL, Zuloaga KL. Menopause causes metabolic and cognitive impairments in a chronic cerebral hypoperfusion model of vascular contributions to cognitive impairment and dementia. Biol Sex Differ 2023; 14:34. [PMID: 37221553 DOI: 10.1186/s13293-023-00518-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/08/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND The vast majority of women with dementia are post-menopausal. Despite clinical relevance, menopause is underrepresented in rodent models of dementia. Before menopause, women are less likely than men to experience strokes, obesity, and diabetes-known risk factors for vascular contributions to cognitive impairment and dementia (VCID). During menopause, ovarian estrogen production stops and the risk of developing these dementia risk factors spikes. Here, we aimed to determine if menopause worsens cognitive impairment in VCID. We hypothesized that menopause would cause metabolic dysfunction and increase cognitive impairment in a mouse model of VCID. METHODS We performed a unilateral common carotid artery occlusion surgery to produce chronic cerebral hypoperfusion and model VCID in mice. We used 4-vinylcyclohexene diepoxide to induce accelerated ovarian failure and model menopause. We evaluated cognitive impairment using behavioral tests including novel object recognition, Barnes maze, and nest building. To assess metabolic changes, we measured weight, adiposity, and glucose tolerance. We explored multiple aspects of brain pathology including cerebral hypoperfusion and white matter changes (commonly observed in VCID) as well as changes to estrogen receptor expression (which may mediate altered sensitivity to VCID pathology post-menopause). RESULTS Menopause increased weight gain, glucose intolerance, and visceral adiposity. VCID caused deficits in spatial memory regardless of menopausal status. Post-menopausal VCID specifically led to additional deficits in episodic-like memory and activities of daily living. Menopause did not alter resting cerebral blood flow on the cortical surface (assessed by laser speckle contrast imaging). In the white matter, menopause decreased myelin basic protein gene expression in the corpus callosum but did not lead to overt white matter damage (assessed by Luxol fast blue). Menopause did not significantly alter estrogen receptor expression (ERα, ERβ, or GPER1) in the cortex or hippocampus. CONCLUSIONS Overall, we have found that the accelerated ovarian failure model of menopause caused metabolic impairment and cognitive deficits in a mouse model of VCID. Further studies are needed to identify the underlying mechanism. Importantly, the post-menopausal brain still expressed estrogen receptors at normal (pre-menopausal) levels. This is encouraging for any future studies attempting to reverse the effects of estrogen loss by activating brain estrogen receptors.
Collapse
Affiliation(s)
- Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Janvie S Naik
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - David Riccio
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Febronia M Mansour
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
17
|
Yu S, Zhang L, Wang Y, Yan J, Wang Q, Bian H, Huang L. Mood, hormone levels, metabolic and sleep across the menopausal transition in VCD-induced ICR mice. Physiol Behav 2023; 265:114178. [PMID: 37001841 DOI: 10.1016/j.physbeh.2023.114178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/27/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
AIMS Menopausal transition is the transitional period before menopause in women, often accompanied by abnormal fluctuations in hormone levels that increase the risk of aging-related diseases. 4-vinylcyclohexene dioxide (VCD) is a chemical agent that induces gradual depletion of ovarian follicles, which can mimic the natural human process of transition from menopausal transition to post-menopause. Previous studies have shown that the onset of menopausal transition or menopause in VCD-injected mice is associated with a specific strain, even in inbred animals. Institute of Cancer Research (ICR) mice constitute general purpose outbred population, which has not been well-characterized in the VCD-induced model. Thus, the current study aimed to explore the characteristic features, including sleep, mood, and metabolism, of the model by examining the effect of timing of VCD injection in ICR mice to extend the applications of this model. MATERIALS AND METHODS ICR mice were randomly divided into six groups: 20d VCD and 20d Control, 35d VCD and 35d Control, 52d VCD and 52d Control. VCD mice were intraperitoneally injected with VCD (160 mg/kg), while Control mice were injected intraperitoneally with sesame oil for 4 consecutive weeks, five times a week daily. A vaginal smear was used to observe the estrous cycle of the mice. On the 20th, 35th, and 52nd day after VCD or sesame oil injection, the ovarian morphology, the number of atretic cells, hormone levels, anxiety, depression-like behaviors, sleep phase, and energy metabolism were observed. KEY FINDINGS The menopausal transition model was successfully replicated by injecting VCD into ICR mice. On the specific days after VCD treatment, the number of atretic follicles increased, the level of E2 decreased and FSH increased, the depressive- and anxiety-like behavior increased, the time of REM and NREM sleep time decreased, and energy metabolism was reduced. SIGNIFICANCE These results suggested that the ICR mice model has human-like characteristics during the menopause transition. Moreover, the ICR model has a long menopausal transition duration.
Collapse
Affiliation(s)
- Shuang Yu
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Lixin Zhang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Yanyan Wang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Jinming Yan
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Qi Wang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Hongsheng Bian
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Lili Huang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China.
| |
Collapse
|
18
|
Massei G. Fertility Control for Wildlife: A European Perspective. Animals (Basel) 2023; 13:428. [PMID: 36766317 PMCID: PMC9913817 DOI: 10.3390/ani13030428] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Trends of human population growth and landscape development in Europe show that wildlife impacts are escalating. Lethal methods, traditionally employed to mitigate these impacts, are often ineffective, environmentally hazardous and face increasing public opposition. Fertility control is advocated as a humane tool to mitigate these impacts. This review describes mammalian and avian wildlife contraceptives' effect on reproduction of individuals and populations, delivery methods, potential costs and feasibility of using fertility control in European contexts. These contexts include small, isolated wildlife populations and situations in which lethal control is either illegal or socially unacceptable, such as urban settings, national parks and areas where rewilding occurs. The review highlights knowledge gaps, such as impact of fertility control on recruitment, social and spatial behaviour and on target and non-target species, provides a decision framework to assist decisions about the potential use of wildlife fertility control, and suggests eight reasons for Europe to invest in this area. Although developing and registering contraceptives in Europe will have substantial costs, these are relatively small when compared to wildlife's economic and environmental impact. Developing safe and effective contraceptives will be essential if European countries want to meet public demand for methods to promote human-wildlife coexistence.
Collapse
Affiliation(s)
- Giovanna Massei
- Botstiber Institute for Wildlife Fertility Control Europe, Department of Environment and Geography, University of York, 290 Wentworth Way, Heslington, York YO10 5NG, UK
| |
Collapse
|
19
|
Sui K, Yasrebi A, Longoria CR, MacDonell AT, Jaffri ZH, Martinez SA, Fisher SE, Malonza N, Jung K, Tveter KM, Wiersielis KR, Uzumcu M, Shapses SA, Campbell SC, Roepke TA, Roopchand DE. Coconut Oil Saturated Fatty Acids Improved Energy Homeostasis but not Blood Pressure or Cognition in VCD-Treated Female Mice. Endocrinology 2023; 164:bqad001. [PMID: 36626144 PMCID: PMC11009791 DOI: 10.1210/endocr/bqad001] [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: 10/04/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
Obesity, cardiometabolic disease, cognitive decline, and osteoporosis are symptoms of postmenopause, which can be modeled using 4-vinylcyclohexene diepoxide (VCD)-treated mice to induce ovarian failure and estrogen deficiency combined with high-fat diet (HFD) feeding. The trend of replacing saturated fatty acids (SFAs), for example coconut oil, with seed oils that are high in polyunsaturated fatty acids, specifically linoleic acid (LA), may induce inflammation and gut dysbiosis, and worsen symptoms of estrogen deficiency. To investigate this hypothesis, vehicle (Veh)- or VCD-treated C57BL/6J mice were fed a HFD (45% kcal fat) with a high LA:SFA ratio (22.5%: 8%), referred to as the 22.5% LA diet, or a HFD with a low LA:SFA ratio (1%: 31%), referred to as 1% LA diet, for a period of 23 to 25 weeks. Compared with VCD-treated mice fed the 22.5% LA diet, VCD-treated mice fed the 1% LA diet showed lower weight gain and improved glucose tolerance. However, VCD-treated mice fed the 1% LA diet had higher blood pressure and showed evidence of spatial cognitive impairment. Mice fed the 1% LA or 22.5% LA diets showed gut microbial taxa changes that have been associated with a mix of both beneficial and unfavorable cognitive and metabolic phenotypes. Overall, these data suggest that consuming different types of dietary fat from a variety of sources, without overemphasis on any particular type, is the optimal approach for promoting metabolic health regardless of estrogen status.
Collapse
Affiliation(s)
- Ke Sui
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ali Yasrebi
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Candace R Longoria
- Department of Kinesiology and Applied Physiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Avery T MacDonell
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Zehra H Jaffri
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Savannah A Martinez
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Samuel E Fisher
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Natasha Malonza
- Department of Kinesiology and Applied Physiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Katie Jung
- Department of Nutritional Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Kevin M Tveter
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Kimberly R Wiersielis
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Mehmet Uzumcu
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Sue A Shapses
- Department of Nutritional Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Sara C Campbell
- Department of Kinesiology and Applied Physiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Troy A Roepke
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Diana E Roopchand
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
20
|
Blackwell JA, Silva JF, Louis EM, Savu A, Largent-Milnes TM, Brooks HL, Pires PW. Cerebral arteriolar and neurovascular dysfunction after chemically induced menopause in mice. Am J Physiol Heart Circ Physiol 2022; 323:H845-H860. [PMID: 36149767 PMCID: PMC9602916 DOI: 10.1152/ajpheart.00276.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 12/14/2022]
Abstract
Cognitive decline is linked to decreased cerebral blood flow, particularly in women after menopause. Impaired cerebrovascular function precedes the onset of dementia, possibly because of reduced functional dilation in parenchymal arterioles. These vessels are bottlenecks of the cerebral microcirculation, and dysfunction can limit functional hyperemia in the brain. Large-conductance Ca2+-activated K+ channels (BKCa) are the final effectors of several pathways responsible for functional hyperemia, and their expression is modulated by estrogen. However, it remains unknown whether BKCa function is altered in cerebral parenchymal arterioles after menopause. Using a chemically induced model of menopause, the 4-vinylcyclohexene diepoxide (VCD) model, which depletes follicles while maintaining intact ovaries, we hypothesized that menopause would be associated with reduced functional vasodilatory responses in cerebral parenchymal arterioles of wild-type mice via reduced BKCa function. Using pressure myography of isolated parenchymal arterioles, we observed that menopause (Meno) induced a significant increase in spontaneous myogenic tone. Endothelial function, assessed as nitric oxide production and dilation after cholinergic stimulation or endothelium-dependent hyperpolarization pathways, was unaffected by Meno. BKCa function was significantly impaired in Meno compared with control, without changes in voltage-gated K+ channel activity. Cerebral functional hyperemia, measured by laser-speckle contrast imaging during whisker stimulation, was significantly blunted in Meno mice, without detectable changes in basal perfusion. However, behavioral testing identified no change in cognition. These findings suggest that menopause induces cerebral microvascular and neurovascular deficits.NEW & NOTEWORTHY Cerebral parenchymal arterioles from menopause mice showed increased myogenic tone. We identified an impairment in smooth muscle cell BKCa channel activity, without a reduction in endothelium-dependent dilation or nitric oxide production. Microvascular dysfunction was associated with a reduction in neurovascular responses after somatosensory stimulation. Despite the neurovascular impairment, cognitive abilities were maintained in menopausal mice.
Collapse
Affiliation(s)
- Jade A Blackwell
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Josiane F Silva
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Emma M Louis
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Andrea Savu
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Tally M Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Paulo W Pires
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
21
|
van As C, Koedam M, McLuskey A, Kramer P, Lahlou N, van der Eerden BCJ, Visser JA. Loss of Anti-Müllerian Hormone Signaling in Mice Affects Trabecular Bone Mass in a Sex- and Age-Dependent Manner. Endocrinology 2022; 163:6717957. [PMID: 36155779 PMCID: PMC9576916 DOI: 10.1210/endocr/bqac157] [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: 02/28/2022] [Indexed: 11/19/2022]
Abstract
Ovariectomy-induced osteoporosis in mice results from an abrupt loss of ovarian sex steroids. Anti-Müllerian hormone knockout (AMHKO) mice show a gradual but accelerated ovarian aging, and therefore may better resemble osteoporosis following natural menopause. To study the impact of AMH signaling deficiency on bone, we compared trabecular and cortical bone parameters in 2-, 4-, 10-, and 16-month-old male and female wild-type (WT), AMHKO, and AMH type II receptor knockout (MRKI) mice using micro computed tomography (microCT). Goldner's staining was performed to confirm the observed bone phenotype. Both male and female AMHKO and MRKI mice showed age-dependent loss of trabecular bone (P < 0.001). However, reproductive-aged female AMHKO and MRKI mice had higher BV/TV compared with WT (P < 0.001), coinciding with increased growing follicle numbers (P < 0.05) and increased estrus inhibin B levels (AMHKO: P < 0.001; MRKI: P < 0.05) but normal inhibin A, estrogen, and progesterone levels. In aged female AMHKO and MRKI mice BV/TV did not differ from WT mice due to greater trabecular bone loss between 10 and 16 months compared with WT mice. At these ages, AMHKO and MRKI mice had reduced growing follicle numbers (P < 0.05) and reduced inhibin B levels (P < 0.001). At age 10 months, female MRKI mice had increased cortical bone parameters compared with WT mice (P < 0.01). Bone parameters of male AMHKO and MRKI mice did not differ from male WT mice. In conclusion, AMH signaling deficiency results in a sex- and age-dependent effect on predominantly trabecular bone. Our results further suggest that reproductive hormones beyond estrogen may contribute to bone homeostasis.
Collapse
Affiliation(s)
- Christiane van As
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Marijke Koedam
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Anke McLuskey
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Piet Kramer
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Najiba Lahlou
- Department of Hormone Biology and Metabolic Disorders, BPR-AS, 45700 Pannes, France
| | | | - Jenny A Visser
- Correspondence: Jenny A. Visser, PhD, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Vrontou S, Bédécarrats A, Wei X, Ayodeji M, Brassai A, Molnár L, Mody I. Altered brain rhythms and behaviour in the accelerated ovarian failure mouse model of human menopause. Brain Commun 2022; 4:fcac166. [PMID: 35794872 PMCID: PMC9253886 DOI: 10.1093/braincomms/fcac166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 03/30/2022] [Accepted: 06/20/2022] [Indexed: 11/14/2022] Open
Abstract
To date, potential mechanisms of menopause-related memory and cognitive deficits have not been elucidated. Therefore, we studied brain oscillations, their phase–amplitude coupling, sleep and vigilance state patterns, running wheel use and other behavioural measures in a translationally valid mouse model of menopause, the 4-vinylcyclohexene-diepoxide-induced accelerated ovarian failure. After accelerated ovarian failure, female mice show significant alterations in brain rhythms, including changes in the frequencies of θ (5–12 Hz) and γ (30–120 Hz) oscillations, a reversed phase–amplitude coupling, altered coupling of hippocampal sharp-wave ripples to medial prefrontal cortical sleep spindles and reduced δ oscillation (0.5–4 Hz) synchrony between the two regions during non-rapid eye movement sleep. In addition, we report on significant circadian variations in the frequencies of θ and γ oscillations, and massive synchronous δ oscillations during wheel running. Our results reveal novel and specific network alterations and feasible signs for diminished brain connectivity in the accelerated ovarian failure mouse model of menopause. Taken together, our results may have identified changes possibly responsible for some of the memory and cognitive deficits previously described in this model. Corresponding future studies in menopausal women could shed light on fundamental mechanisms underlying the neurological and psychiatric comorbidities present during this important transitional phase in women’s lives.
Collapse
Affiliation(s)
- Sophia Vrontou
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| | - Alexis Bédécarrats
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| | | | - Attila Brassai
- Department of Pharmacology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology , Târgu Mureş 540139 , Romania
| | - László Molnár
- Department of Electrical Engineering, Sapientia Hungarian University of Transylvania , Târgu Mureş 540485 , Romania
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
- Department of Physiology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| |
Collapse
|
23
|
Marchant IC, Chabert S, Martínez-Pinto J, Sotomayor-Zárate R, Ramírez-Barrantes R, Acevedo L, Córdova C, Olivero P. Estrogen, Cognitive Performance, and Functional Imaging Studies: What Are We Missing About Neuroprotection? Front Cell Neurosci 2022; 16:866122. [PMID: 35634466 PMCID: PMC9133497 DOI: 10.3389/fncel.2022.866122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 01/20/2023] Open
Abstract
Menopause transition can be interpreted as a vulnerable state characterized by estrogen deficiency with detrimental systemic effects as the low-grade chronic inflammation that appears with aging and partly explains age-related disorders as cancer, diabetes mellitus and increased risk of cognitive impairment. Over the course of a lifetime, estrogen produces several beneficial effects in healthy neurological tissues as well as cardioprotective effects, and anti-inflammatory effects. However, clinical evidence on the efficacy of hormone treatment in menopausal women has failed to confirm the benefit reported in observational studies. Unambiguously, enhanced verbal memory is the most robust finding from longitudinal and cross-sectional studies, what merits consideration for future studies aiming to determine estrogen neuroprotective efficacy. Estrogen related brain activity and functional connectivity remain, however, unexplored. In this context, the resting state paradigm may provide valuable information about reproductive aging and hormonal treatment effects, and their relationship with brain imaging of functional connectivity may be key to understand and anticipate estrogen cognitive protective effects. To go in-depth into the molecular and cellular mechanisms underlying rapid-to-long lasting protective effects of estrogen, we will provide a comprehensive review of cognitive tasks used in animal studies to evaluate the effect of hormone treatment on cognitive performance and discuss about the tasks best suited to the demonstration of clinically significant differences in cognitive performance to be applied in human studies. Eventually, we will focus on studies evaluating the DMN activity and responsiveness to pharmacological stimulation in humans.
Collapse
Affiliation(s)
- Ivanny Carolina Marchant
- Laboratorio de Modelamiento en Medicina, Escuela de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
- Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
- *Correspondence: Ivanny Carolina Marchant
| | - Stéren Chabert
- Millennium Nucleus in Cardiovascular Magnetic Resonance, Santiago, Chile
- Escuela de Ingeniería Biomédica, Universidad de Valparaiso, Valparaíso, Chile
- Centro de Investigación y Desarrollo en Ingeniería en Salud, Universidad de Valparaíso, Valparaíso, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa, Valparaíso, Chile
- Laboratorio de Neuroquímica y Neurofarmacología, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa, Valparaíso, Chile
- Laboratorio de Neuroquímica y Neurofarmacología, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | - Lilian Acevedo
- Servicio de Neurología Hospital Carlos van Buren, Valparaíso, Chile
| | - Claudio Córdova
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Olivero
- Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
- Pablo Olivero
| |
Collapse
|
24
|
Aging-related changes in metabolic indicators in female rats and their management with Tinospora cordifolia. Biogerontology 2022; 23:363-380. [PMID: 35488997 DOI: 10.1007/s10522-022-09962-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
Conflicting reports of HRT necessitates exploration of therapeutic interventions with the least side effects to preserve metabolic homeodynamics in women later in life. The current study was designed to elucidate the cumulative effects of aging and/or high fat diet (HFD) on some metabolic indicators and their management by Tinospora cordifolia stem powder (TCP) using middle-aged acyclic and young adult cyclic female rats as the model system. Animals were fed on either normal chow or HFD supplemented with or without TCP. Blood and liver tissue were collected for biochemical, and histological studies as well as for expression of proteins regulating lipid metabolism. Animals fed with TCP supplemented normal chow feed showed bodyweight management over 12-weeks despite their high feed and calories intake compared to young and age-matched controls as well as HFD-fed animals. TCP dose used was not toxic and rather prevented age-associated liver dysfunctions and ameliorated dyslipidemia and oxidative stress, normalized blood glucose, insulin, leptin, and secretary pro-inflammatory cytokines. Further, bodyweight management effect of TCP was observed to target AMPK signalling pathway as the mediator of lipogenesis, sterol biosynthesis, lipolysis, and β-oxidation of fatty acids. These findings suggest that TCP supplementation in diet may be a potential interventional strategy to ameliorate aging-associated hepatic and metabolic dysfunctions and to promote healthy aging.
Collapse
|
25
|
Han G, Choi J, Cha SY, Kim BI, Kho HK, Jang MJ, Kim MA, Maeng S, Hong H. Effects of Radix Polygalae on Cognitive Decline and Depression in Estradiol Depletion Mouse Model of Menopause. Curr Issues Mol Biol 2021; 43:1669-1684. [PMID: 34698102 PMCID: PMC8929121 DOI: 10.3390/cimb43030118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/22/2022] Open
Abstract
Postmenopausal syndrome refers to symptoms caused by the gradual decrease in female hormones after mid-40 years. As a target organ of estrogen, decrease in estrogen causes various changes in brain function such as a decrease in choline acetyltransferase and brain-derived neurotrophic factor; thus, postmenopausal women experience cognitive decline and more depressive symptoms than age-matched men. Radix Polygalae has been used for memory boosting and as a mood stabilizer and its components have shown neuroprotective, antidepressant, and stress relief properties. In a mouse model of estrogen depletion induced by 4-vinylcyclohexene diepoxide, Radix Polygalae was orally administered for 3 weeks. In these animals, cognitive and depression-related behaviors and molecular changes related to these behaviors were measured in the prefrontal cortex and hippocampus. Radix Polygalae improved working memory and contextual memory and despair-related behaviors in 4-vinylcyclohexene diepoxide-treated mice without increasing serum estradiol levels in this model. In relation to these behaviors, choline acetyltransferase and brain-derived neurotrophic factor in the prefrontal cortex and hippocampus and bcl-2-associated athanogene expression increased in the hippocampus. These results implicate the possible benefit of Radix Polygalae in use as a supplement of estrogen to prevent conditions such as postmenopausal depression and cognitive decline.
Collapse
Affiliation(s)
- Gaeul Han
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Junhyuk Choi
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Seung-Yun Cha
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Byung Il Kim
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Hee Kyung Kho
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Maeng-Jin Jang
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Mi Ae Kim
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
| | - Sungho Maeng
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea; (G.H.); (J.C.); (S.-Y.C.); (B.I.K.); (H.K.K.); (M.-J.J.); (M.A.K.)
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si 17104, Korea
- Correspondence: (S.M.); (H.H.); Tel.: +82-31-201-2916 (S.M.); +82-2-2049-6274 (H.H.)
| | - Heeok Hong
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.M.); (H.H.); Tel.: +82-31-201-2916 (S.M.); +82-2-2049-6274 (H.H.)
| |
Collapse
|
26
|
Jacoblinnert K, Jacob J, Zhang Z, Hinds LA. The status of fertility control for rodents-recent achievements and future directions. Integr Zool 2021; 17:964-980. [PMID: 34549512 DOI: 10.1111/1749-4877.12588] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Management of overabundant rodents at a landscape scale is complex but often required to sustainably reduce rodent abundance below damage thresholds. Current conventional techniques such as poisoning are not species specific, with some approaches becoming increasingly unacceptable to the general public. Fertility control, first proposed for vertebrate pest management over 5 decades ago, has gained public acceptance because it is perceived as a potentially more species-specific and humane approach compared with many lethal methods. An ideal fertility control agent needs to induce infertility across one or more breeding seasons, be easily delivered to an appropriate proportion of the population, be species specific with minimal side-effects (behavioral or social structure changes), and be environmentally benign and cost effective. To date, effective fertility control of rodents has not been demonstrated at landscape scales and very few products have achieved registration. Reproductive targets for fertility control include disrupting the hormonal feedback associated with the hypothalamic-pituitary-gonadal axis, gonad function, fertilization, and/or early implantation. We review progress on the oral delivery of various agents for which laboratory studies have demonstrated efficacy in females and/or males and synthesize progress with the development and/or use of synthetic steroids, plant extracts, ovarian specific peptides, and immunocontraceptive vaccines. There are promising results for field application of synthetic steroids (levonorgestrel, quinestrol), chemosterilants (4-vinylcyclohexene diepoxide), and some plant extracts (triptolide). For most fertility control agents, more research is essential to enable their efficient and cost-effective delivery such that rodent impacts at a population level are mitigated and food security is improved.
Collapse
Affiliation(s)
- Kyra Jacoblinnert
- Julius Kühn-Institute (JKI), Federal Research Centre for Cultivated Plants, Institute for Plant Protection in Horticulture and Forests, Vertebrate Research, Münster, Germany.,Department of Behavioral Biology, University Osnabrück, Osnabrück, Germany
| | - Jens Jacob
- Julius Kühn-Institute (JKI), Federal Research Centre for Cultivated Plants, Institute for Plant Protection in Horticulture and Forests, Vertebrate Research, Münster, Germany
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management on Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Lyn A Hinds
- CSIRO Health and Biosecurity, Canberra, ACT, Australia
| |
Collapse
|
27
|
Koebele SV, Hiroi R, Plumley ZMT, Melikian R, Prakapenka AV, Patel S, Carson C, Kirby D, Mennenga SE, Mayer LP, Dyer CA, Bimonte-Nelson HA. Clinically Used Hormone Formulations Differentially Impact Memory, Anxiety-Like, and Depressive-Like Behaviors in a Rat Model of Transitional Menopause. Front Behav Neurosci 2021; 15:696838. [PMID: 34366807 PMCID: PMC8335488 DOI: 10.3389/fnbeh.2021.696838] [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: 04/17/2021] [Accepted: 06/04/2021] [Indexed: 01/29/2023] Open
Abstract
A variety of U.S. Food and Drug Administration-approved hormone therapy options are currently used to successfully alleviate unwanted symptoms associated with the changing endogenous hormonal milieu that occurs in midlife with menopause. Depending on the primary indication for treatment, different hormone therapy formulations are utilized, including estrogen-only, progestogen-only, or combined estrogen plus progestogen options. There is little known about how these formulations, or their unique pharmacodynamics, impact neurobiological processes. Seemingly disparate pre-clinical and clinical findings regarding the cognitive effects of hormone therapies, such as the negative effects associated with conjugated equine estrogens and medroxyprogesterone acetate vs. naturally circulating 17β-estradiol (E2) and progesterone, signal a critical need to further investigate the neuro-cognitive impact of hormone therapy formulations. Here, utilizing a rat model of transitional menopause, we administered either E2, progesterone, levonorgestrel, or combinations of E2 with progesterone or with levonorgestrel daily to follicle-depleted, middle-aged rats. A battery of assessments, including spatial memory, anxiety-like behaviors, and depressive-like behaviors, as well as endocrine status and ovarian follicle complement, were evaluated. Results indicate divergent outcomes for memory, anxiety, and depression, as well as unique physiological profiles, that were dependent upon the hormone regimen administered. Overall, the combination hormone treatments had the most consistently favorable profile for the domains evaluated in rats that had undergone experimentally induced transitional menopause and remained ovary-intact. The collective results underscore the importance of investigating variations in hormone therapy formulation as well as the menopause background upon which these formulations are delivered.
Collapse
Affiliation(s)
- Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Ryoko Hiroi
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Zachary M. T. Plumley
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Ryan Melikian
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Alesia V. Prakapenka
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Catherine Carson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Destiney Kirby
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Sarah E. Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | | | | | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| |
Collapse
|
28
|
Milner TA, Contoreggi NH, Yu F, Johnson MA, Wang G, Woods C, Mazid S, Van Kempen TA, Waters EM, McEwen BS, Korach KS, Glass MJ. Estrogen Receptor β Contributes to Both Hypertension and Hypothalamic Plasticity in a Mouse Model of Peri-Menopause. J Neurosci 2021; 41:5190-5205. [PMID: 33941651 PMCID: PMC8211546 DOI: 10.1523/jneurosci.0164-21.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertension susceptibility in women increases at the transition to menopause, termed perimenopause, a state characterized by erratic estrogen fluctuation and extended hormone cycles. Elucidating the role of estrogen signaling in the emergence of hypertension during perimenopause has been hindered by animal models that are confounded by abrupt estrogen cessation or effects of aging. In the present study, accelerated ovarian failure (AOF) in estrogen receptor β (ERβ) reporter mice was induced by 4-vinylcyclohexene diepoxide in young mice to model early-stage ovarian failure (peri-AOF) characteristic of peri-menopause. It was found that administering ERβ agonists suppressed elevated blood pressure in a model of neurogenic hypertension induced by angiotensin II (AngII) in peri-AOF, but not in age-matched male mice. It was also found that ERβ agonist administration in peri-AOF females, but not males, suppressed the heightened NMDAR signaling and reactive oxygen production in ERβ neurons in the hypothalamic paraventricular nucleus (PVN), a critical neural regulator of blood pressure. It was further shown that deleting ERβ in the PVN of gonadally intact females produced a phenotype marked by a sensitivity to AngII hypertension. These results suggest that ERβ signaling in the PVN plays an important role in blood pressure regulation in female mice and contributes to hypertension susceptibility in females at an early stage of ovarian failure comparable to human perimenopause.
Collapse
Affiliation(s)
- Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
- Harold and Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, New York 10065
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Fangmin Yu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Megan A Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Clara Woods
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Elizabeth M Waters
- Harold and Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, New York 10065
| | - Bruce S McEwen
- Harold and Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, New York 10065
| | - Kenneth S Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, North Carolina 27709
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
29
|
Bimonte-Nelson HA, Bernaud VE, Koebele SV. Menopause, hormone therapy and cognition: maximizing translation from preclinical research. Climacteric 2021; 24:373-381. [PMID: 33977823 DOI: 10.1080/13697137.2021.1917538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Menopause-associated and hormone-associated cognitive research has a rich history built from varied disciplines and species. This review discusses landmark rodent and human work addressing cognitive outcomes associated with varied experiences of menopause and hormone therapy. Critical variables in menopause and cognitive aging research are considered, including menopause etiology, background hormone milieu and parameters of exposure to estrogens and progestogens. Recent preclinical research has identified that menopause and ovarian hormone fluctuations across many neurobiological systems affect cognitive aging, mapping novel avenues for future research. Preclinical models provide insight into complex interdisciplinary relationships in a systematic and highly controlled fashion. We highlight that acknowledging the strengths and weaknesses for both preclinical and clinical research approaches is vital to accurate interpretation, optimal translation and the direction of future research. There is great value in collaboration and communication across preclinical and clinical realms, especially regarding reciprocal feedback of findings to advance preclinical models, improve experimental designs and enrich basic science translation to the clinic. In searching for biological mechanisms underlying the cognitive consequences of menopause and hormone therapies, it is noteworthy that clinical and preclinical scientists are grounded in the same fundamental goal of optimizing health outcomes for women across the lifespan.
Collapse
Affiliation(s)
- H A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - V E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - S V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, USA.,Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| |
Collapse
|
30
|
Arikawe AP, Rorato RC, Gomes N, Elias LL, Anselmo-Franci J. Hormonal and neural responses to restraint stress in an animal model of perimenopause in female rats. J Neuroendocrinol 2021; 33:e12976. [PMID: 33900672 DOI: 10.1111/jne.12976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 01/21/2023]
Abstract
The present study investigated the hormonal and neural responses to stress in a perimenopause animal model induced by 4-vinylcyclohexene diepoxide (VCD), which induces progressive follicular depletion in rodents, allowing studies on the transition to ovarian failure. Female rats, aged 28 days old, were s.c. injected for 15 consecutive days with corn oil or VCD. At 85 ± 5 days after the onset of treatment, the jugular vein was cannulated in the afternoon of metoestrus and in next morning (dioestrus) at 10.00 am, rats were subjected to 30 minutes of restraint stress. Blood samples were withdrawn before (-5 minutes), during (2, 5, 15 and 30 minutes) and after (45, 60 and 90 minutes) stress and plasma prolactin, progesterone and corticosterone levels were measured. Animals were perfused, brains processed for c-Fos/tyrosine hydroxylase (TH) in the locus coeruleus (LC) and c-Fos/corticotrophin-releasing factor (CRF) in the paraventricular nucleus (PVN). In unstressed rats the density of β-endorphin fibres was assessed in LC and PVN. In VCD-treated rats, stress-induced prolactin peak was higher, basal and peak progesterone levels were lower, and both levels of corticosterone were similar to controls. However, the recovery period was longer for both adrenal hormones. In VCD-treated rats the number of c-Fos/TH and c-Fos/CRF-immunoreactive neurones was higher whereas the density of β-endorphin fibres was lower in LC and PVN. We surmise that the hyperactivity of the LC and PVN neurones in VCD-treated rats may be a result of the lower progesterone levels that resulted in the decrease of β-endorphin content in both nuclei, thus impairing the negative-feedback mechanism in the recovery period.
Collapse
Affiliation(s)
- Adesina Paul Arikawe
- Laboratory of Neuroendocrinology, Department of Basic and Oral Biology Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Rodrigo César Rorato
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
- Departmento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nathali Gomes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Lucila Leico Elias
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Janete Anselmo-Franci
- Laboratory of Neuroendocrinology, Department of Basic and Oral Biology Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Young KF, Gardner R, Sariana V, Whitman SA, Bartlett MJ, Falk T, Morrison HW. Can quantifying morphology and TMEM119 expression distinguish between microglia and infiltrating macrophages after ischemic stroke and reperfusion in male and female mice? J Neuroinflammation 2021; 18:58. [PMID: 33618737 PMCID: PMC7901206 DOI: 10.1186/s12974-021-02105-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/05/2021] [Indexed: 12/30/2022] Open
Abstract
Background Ischemic stroke is an acquired brain injury with gender-dependent outcomes. A persistent obstacle in understanding the sex-specific neuroinflammatory contributions to ischemic brain injury is distinguishing between resident microglia and infiltrating macrophages—both phagocytes—and determining cell population-specific contributions to injury evolution and recovery processes. Our purpose was to identify microglial and macrophage populations regulated by ischemic stroke using morphology analysis and the presence of microglia transmembrane protein 119 (TMEM119). Second, we examined sex and menopause differences in microglia/macrophage cell populations after an ischemic stroke. Methods Male and female, premenopausal and postmenopausal, mice underwent either 60 min of middle cerebral artery occlusion and 24 h of reperfusion or sham surgery. The accelerated ovarian failure model was used to model postmenopause. Brain tissue was collected to quantify the infarct area and for immunohistochemistry and western blot methods. Ionized calcium-binding adapter molecule, TMEM119, and confocal microscopy were used to analyze the microglia morphology and TMEM119 area in the ipsilateral brain regions. Western blot was used to quantify protein quantity. Results Post-stroke injury is increased in male and postmenopause female mice vs. premenopause female mice (p < 0.05) with differences primarily occurring in the caudal sections. After stroke, the microglia underwent a region, but not sex group, dependent transformation into less ramified cells (p < 0.0001). However, the number of phagocytic microglia was increased in distal ipsilateral regions of postmenopausal mice vs. the other sex groups (p < 0.05). The number of TMEM119-positive cells was decreased in proximity to the infarct (p < 0.0001) but without a sex group effect. Two key findings prevented distinguishing microglia from systemic macrophages. First, morphological data were not congruent with TMEM119 immunofluorescence data. Cells with severely decreased TMEM119 immunofluorescence were ramified, a distinguishing microglia characteristic. Second, whereas the TMEM119 immunofluorescence area decreased in proximity to the infarcted area, the TMEM119 protein quantity was unchanged in the ipsilateral hemisphere regions using western blot methods. Conclusions Our findings suggest that TMEM119 is not a stable microglia marker in male and female mice in the context of ischemic stroke. Until TMEM119 function in the brain is elucidated, its use to distinguish between cell populations following brain injury with cell infiltration is cautioned. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02105-2.
Collapse
Affiliation(s)
- Kimberly F Young
- College of Nursing, University of Arizona, 1305 N. Martin Ave., Tucson, AZ, 85721, USA.,Current affiliation: Department of Psychology, University of Arizona, 1503 E University Blvd., Tucson, AZ, USA.,University of Arizona Evelyn F. McKnight Brain Institute, 1333 N. Martin Ave., Tucson, AZ, USA
| | - Rebeca Gardner
- College of Science, University of Arizona, 1040 4th St., Tucson, AZ, USA
| | - Victoria Sariana
- College of Nursing, University of Arizona, 1305 N. Martin Ave., Tucson, AZ, 85721, USA
| | - Susan A Whitman
- College of Nursing, University of Arizona, 1305 N. Martin Ave., Tucson, AZ, 85721, USA
| | - Mitchell J Bartlett
- College of Medicine, Department of Neurology, University of Arizona, 1501 N. Campbell Ave., Tucson, AZ, USA
| | - Torsten Falk
- College of Medicine, Department of Neurology, University of Arizona, 1501 N. Campbell Ave., Tucson, AZ, USA.,College of Medicine, Department of Pharmacology, University of Arizona, 1501 N. Campbell Ave., Tucson, AZ, USA
| | - Helena W Morrison
- College of Nursing, University of Arizona, 1305 N. Martin Ave., Tucson, AZ, 85721, USA.
| |
Collapse
|
32
|
Koebele SV, Mennenga SE, Poisson ML, Hewitt LT, Patel S, Mayer LP, Dyer CA, Bimonte-Nelson HA. Characterizing the effects of tonic 17β-estradiol administration on spatial learning and memory in the follicle-deplete middle-aged female rat. Horm Behav 2020; 126:104854. [PMID: 32949557 PMCID: PMC8032560 DOI: 10.1016/j.yhbeh.2020.104854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 01/19/2023]
Abstract
17β-estradiol (E2)-containing hormone therapy is a safe, effective way to alleviate unwanted menopause symptoms. Preclinical research has focused upon the role of E2 in learning and memory using a surgically menopausal rodent model whereby the ovaries are removed. Given that most women retain their reproductive tract and undergo a natural menopause transition, it is necessary to understand how exogenous E2 impacts a structurally intact, but follicle-deplete, system. In the current study, 8 month old female rats were administered the ovatoxin 4-vinylcyclohexene diepoxide (VCD), which accelerates ovarian follicular depletion, to model the human menopause transition. After follicular depletion, at 11 months old, rats were administered Vehicle or tonic E2 treatment for 12 days prior to behavioral evaluation on spatial working and reference memory tasks. Results demonstrated that E2 had both enhancing and impairing effects on taxed working memory depending upon the learning or retention phases of the water radial-arm maze, with no impact on reference memory. Relationships between memory scores and circulating estrogen levels were specific to follicle-depleted rats without E2 treatment. Collectively, findings demonstrate the complexity of E2 administration in a follicle-depleted background, with cognitive effects specific to working memory; furthermore, E2 administration altered circulating hormonal milieu and relationships between hormone profiles and memory. In sum, menopausal etiology impacts the parameters of E2 effects on cognition, complementing prior work with other estrogen compounds. Deciphering estrogenic actions in a system wherein the reproductive tract remains intact with follicle-depleted ovaries, thus modeling the majority or menopausal women, is critical for translational perspectives.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Sarah E Mennenga
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Mallori L Poisson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Lauren T Hewitt
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | - Shruti Patel
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America
| | | | - Cheryl A Dyer
- FYXX Foundation, Flagstaff, AZ, United States of America
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States of America; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States of America.
| |
Collapse
|
33
|
Kirshner ZZ, Yao JK, Li J, Long T, Nelson D, Gibbs RB. Impact of estrogen receptor agonists and model of menopause on enzymes involved in brain metabolism, acetyl-CoA production and cholinergic function. Life Sci 2020; 256:117975. [PMID: 32565251 PMCID: PMC7448522 DOI: 10.1016/j.lfs.2020.117975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022]
Abstract
Our goal is to understand how loss of circulating estrogens and estrogen replacement affect brain physiology and function, particularly in brain regions involved in cognitive processes. We recently conducted a large metabolomics study characterizing the effects of rodent models of menopause and treatment with estrogen receptor (ER) agonists on neurochemical targets in hippocampus, frontal cortex, and striatum. Here we characterize effects on levels of several key enzymes involved in glucose utilization and energy production, specifically phosphofructokinase, glyceraldehyde 3-phosphate dehydrogenase, and pyruvate dehydrogenase. We also evaluated effects on levels of β-actin and α-tubulin, choline acetyltransferase (ChAT) activity, and levels of ATP citrate lyase. All experiments were conducted in young adult rats. Experiment 1 compared the effects of ovariectomy (OVX), a model of surgical menopause, and 4-vinylcyclohexene diepoxide (VCD)-treatments, a model of transitional menopause, with tissues collected at proestrus and at diestrus. Experiment 2 used a separate cohort of rats to evaluate the same targets in OVX and VCD-treated rats treated with estradiol or with selective ER agonists. Differences in the expression of metabolic enzymes between cycling animals and models of surgical and transitional menopause were detected. These differences were model-, region- and time- dependent, and were modulated by selective ER agonists. Collectively, the findings demonstrate that loss of ovarian function and ER agonist treatments have differing effects in OVX vs. VCD-treated rats. Differences may help to explain differences in the effects of estrogen treatments on brain function and cognition in women who have experienced surgical vs. transitional menopause.
Collapse
Affiliation(s)
- Z Z Kirshner
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Jeffrey K Yao
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Junyi Li
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Tao Long
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - Doug Nelson
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - R B Gibbs
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
34
|
Ahmadian S, Sheshpari S, Pazhang M, Bedate AM, Beheshti R, Abbasi MM, Nouri M, Rahbarghazi R, Mahdipour M. Intra-ovarian injection of platelet-rich plasma into ovarian tissue promoted rejuvenation in the rat model of premature ovarian insufficiency and restored ovulation rate via angiogenesis modulation. Reprod Biol Endocrinol 2020; 18:78. [PMID: 32758249 PMCID: PMC7405361 DOI: 10.1186/s12958-020-00638-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Premature Ovarian Insufficiency (POI) is viewed as a type of infertility in which the menopausal status occurs before the physiological age. Several therapeutic strategies have been introduced in clinic for POI treatment, although the outputs are not fully convincing. Platelet-rich plasma (PRP) is a unique blood product widely applied in regenerative medicine, which is based on the releasing of the growth factors present in platelets α-granules. In the current investigation, we examined the effectiveness of PRP as a therapeutic alternative for POI animals. POI in Wistar albino rats was induced by daily intraperitoneal (IP) administration of gonadotoxic chemical agent, 4-vinylcyclohexene dioxide (VCD) (160 mg/ kg) for 15 consecutive days. After POI induction, the PRP solution was directly injected intra-ovarian in two concentrations via a surgical intervention. Every two weeks post-injection, pathological changes were monitored in the ovaries using Hematoxylin-Eosin staining method, until eight weeks. Follicle Stimulating Hormone (FSH) content in serum was measured, together with the expression of the angiogenic-related transcripts ANGPT2 and KDR by real-time qPCR. Furthermore the fertility status of the treated rats was evaluated by mating trials. Histopathological examination revealed successful POI induction via the depletion of morphologically normal follicles in rats following VCD treatment compared to the control rats. The injection of PRP at two concentrations reduced the number and extent of the follicular atresia and inflammatory responses (p < 0.05). The expression of both ANGPT2 and KDR transcripts were significantly increased in POI rats due to enhanced inflammation, while these values were modulated after PRP administration (p < 0.05) compared to POI rats. FSH showed a decreased trend in concentration eight weeks after PRP treatment, but not statistically significant (p > 0.05). Nevertheless, a clear improvement in litter counts was found in POI rats receiving PRP compared to the non-treated POI group, being able to consider PRP as a facile, quick, accessible, safe and relatively cheap alternative therapeutic strategy to revert POI-related pathologies.
Collapse
Affiliation(s)
- Shahin Ahmadian
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, 5138663134, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, 537517169, Iran
| | - Sepideh Sheshpari
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, 5138947977, Iran
| | - Mohammad Pazhang
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, 537517169, Iran
| | - Alberto Miranda Bedate
- Laboratory for Translational Immunology (LTI), Universitair Medisch Centrum Utrecht, (UMCU), Utrecht, Heidelberglaan 100, 3584, CX, The Netherlands
| | - Rahim Beheshti
- Department of Veterinary Science, Islamic Azad University Shabestar Branch, Shabestar, 5381637181, Iran
| | | | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 5166653431, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 5166653431, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran.
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 5166653431, Iran.
| |
Collapse
|
35
|
Konhilas JP, Sanchez JN, Regan JA, Constantopoulos E, Lopez-Pier M, Cannon DK, Skaria R, McKee LA, Chen H, Lipovka Y, Pollow D, Brooks HL. Using 4-vinylcyclohexene diepoxide as a model of menopause for cardiovascular disease. Am J Physiol Heart Circ Physiol 2020; 318:H1461-H1473. [PMID: 32383991 PMCID: PMC7311698 DOI: 10.1152/ajpheart.00555.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a sharp rise in cardiovascular disease (CVD) risk and progression with the onset of menopause. The 4-vinylcyclohexene diepoxide (VCD) model of menopause recapitulates the natural, physiological transition through perimenopause to menopause. We hypothesized that menopausal female mice were more susceptible to CVD than pre- or perimenopausal females. Female mice were treated with VCD or vehicle for 20 consecutive days. Premenopausal, perimenopausal, and menopausal mice were administered angiotensin II (ANG II) or subjected to ischemia-reperfusion (I/R). Menopausal females were more susceptible to pathological ANG II-induced cardiac remodeling and cardiac injury from a myocardial infarction (MI), while perimenopausal, like premenopausal, females remained protected. Specifically, ANG II significantly elevated diastolic (130.9 ± 6.0 vs. 114.7 ± 6.2 mmHg) and systolic (156.9 ± 4.8 vs. 141.7 ± 5.0 mmHg) blood pressure and normalized cardiac mass (15.9 ± 1.0 vs. 7.7 ± 1.5%) to a greater extent in menopausal females compared with controls, whereas perimenopausal females demonstrated a similar elevation of diastolic (93.7 ± 2.9 vs. 100.5 ± 4.1 mmHg) and systolic (155.9 ± 7.3 vs. 152.3 ± 6.5 mmHg) blood pressure and normalized cardiac mass (8.3 ± 2.1 vs. 7.5 ± 1.4%) compared with controls. Similarly, menopausal females demonstrated a threefold increase in fibrosis measured by Picrosirus red staining. Finally, hearts of menopausal females (41 ± 5%) showed larger infarct sizes following I/R injury than perimenopausal (18.0 ± 5.6%) and premenopausal (16.2 ± 3.3, 20.1 ± 4.8%) groups. Using the VCD model of menopause, we provide evidence that menopausal females were more susceptible to pathological cardiac remodeling. We suggest that the VCD model of menopause may be critical to better elucidate cellular and molecular mechanisms underlying the transition to CVD susceptibility in menopausal women.NEW & NOTEWORTHY Before menopause, women are protected against cardiovascular disease (CVD) compared with age-matched men; this protection is gradually lost after menopause. We present the first evidence that demonstrates menopausal females are more susceptible to pathological cardiac remodeling while perimenopausal and cycling females are not. The VCD model permits appropriate examination of how increased susceptibility to the pathological process of cardiac remodeling accelerates from pre- to perimenopause to menopause.
Collapse
Affiliation(s)
- John P Konhilas
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Jessica N Sanchez
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Jessica A Regan
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Eleni Constantopoulos
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Marissa Lopez-Pier
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | | | - Rinku Skaria
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Laurel A McKee
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Hao Chen
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Yulia Lipovka
- Department of Physiology, University of Arizona, Tucson, Arizona.,Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona
| | - Dennis Pollow
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona
| |
Collapse
|
36
|
Carolino ROG, Barros PT, Kalil B, Anselmo-Franci J. Endocrine profile of the VCD-induced perimenopausal model rat. PLoS One 2019; 14:e0226874. [PMID: 31887176 PMCID: PMC6936812 DOI: 10.1371/journal.pone.0226874] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/06/2019] [Indexed: 11/19/2022] Open
Abstract
During the transition to menopause, women experience a variety of physical and psychological symptoms that are directly or indirectly linked to changes in hormone secretion. Establishing animal models with intact ovaries is essential for understanding these interactions and finding new therapeutic targets. In this study, we assessed the endocrine profile, as well as the estrous cycle, in the 4-vinylcyclohexene diepoxide (VCD)-induced follicular depletion rat model in 10-day intervals over 1 month to accurately establish the best period for studies of the transition period. Twenty-eight-day-old female rats were injected daily with VCD or oil s.c. for 15 days and euthanized in the diestrus phase approximately 70, 80, 90 and 100 days after the onset of treatment. The percentage of rats showing irregular cycles and the plasma level of FSH increased only in the 100-day VCD group. Plasma anti-Müllerian hormone (AMH) and progesterone were lower in all VCD groups compared to control groups, while estradiol remained unchanged or higher. As in control groups, dihydrotestosterone (DHT) progressively decreased in the 70-90-day VCD groups; however, it was followed by a sharp increase only in the 100-day VCD group. No changes were found in plasma corticosterone, prolactin, thyroid hormones or luteinizing hormone. Based on the estrous cycle and endocrine profile, we conclude that 1) the time window from 70 to 100 days is suitable to study a perimenopause-like state in this model, and 2) regular cycles with low progesterone and AMH and normal FSH can be used as markers of the early/mid-transition period, whereas irregular cycles associated with higher FSH and DHT can be used as markers of the late transition period to estropause.
Collapse
Affiliation(s)
- Ruither O. G. Carolino
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Paulo T. Barros
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Bruna Kalil
- Department of Physiology, Institute of Biomedical Science, Federal University of Alfenas, Alfenas, MG, Brazil
| | - Janete Anselmo-Franci
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
37
|
Rubin BR, Milner TA, Pickel VM, Coleman CG, Marques-Lopes J, Van Kempen TA, Kazim SF, McEwen BS, Gray JD, Pereira AC. Sex and age differentially affect GABAergic neurons in the mouse prefrontal cortex and hippocampus following chronic intermittent hypoxia. Exp Neurol 2019; 325:113075. [PMID: 31837319 DOI: 10.1016/j.expneurol.2019.113075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/22/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Obstructive sleep apnea (OSA), a chronic sleep disorder characterized by repetitive reduction or cessation of airflow during sleep, is widely prevalent and is associated with adverse neurocognitive sequelae including increased risk of Alzheimer's disease (AD). In humans, OSA is more common in elderly males. OSA is characterized by sleep fragmentation and chronic intermittent hypoxia (CIH), and recent epidemiological studies point to CIH as the best predictor of neurocognitive sequelae associated with OSA. The sex- and age- specific effects of OSA-associated CIH on specific cell populations such as γ-aminobutyric acid (GABA)-ergic neurons in the hippocampus and the medial prefrontal cortex (mPFC), regions important for cognitive function, remain largely unknown. The present study examined the effect of 35 days of either moderate (10% oxygen) or severe (5% oxygen) CIH on GABAergic neurons in the mPFC and hippocampus of young and aged male and female mice as well as post-accelerated ovarian failure (AOF) female mice. In the mPFC and hippocampus, the number of GABA-labeled neurons increased in aged and young severe CIH males compared to controls but not in young moderate CIH males. This change was not representative of the individual GABAergic cell subpopulations, as the number of parvalbumin-labeled neurons decreased while the number of somatostatin-labeled neurons increased in the hippocampus of severe CIH young males only. In all female groups, the number of GABA-labeled cells was not different between CIH and controls. However, in the mPFC, CIH increased the number of parvalbumin-labeled neurons in young females and the number of somatostatin-labeled cells in AOF females but decreased the number of somatostatin-labeled cells in aged females. In the hippocampus, CIH decreased the number of somatostatin-labeled neurons in young females. CIH decreased the density of vesicular GABA transporter in the mPFC of AOF females only. These findings suggest sex-specific changes in GABAergic neurons in the hippocampus and mPFC with males showing an increase of this cell population as compared to their female counterparts following CIH. Age at exposure and severity of CIH also differentially affect the GABAergic cell population in mice.
Collapse
Affiliation(s)
- Batsheva R Rubin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Teresa A Milner
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Virginia M Pickel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Christal G Coleman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, United States of America
| | - Syed Faraz Kazim
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America
| | - Ana C Pereira
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, United States of America; Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| |
Collapse
|
38
|
Ovalles AC, Contoreggi NH, Marques-Lopes J, Van Kempen TA, Iadecola C, Waters EM, Glass MJ, Milner TA. Plasma Membrane Affiliated AMPA GluA1 in Estrogen Receptor β-containing Paraventricular Hypothalamic Neurons Increases Following Hypertension in a Mouse Model of Post-menopause. Neuroscience 2019; 423:192-205. [PMID: 31682817 DOI: 10.1016/j.neuroscience.2019.09.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
Sex and ovarian function contribute to hypertension susceptibility, however, the mechanisms are not well understood. Prior studies show that estrogens and neurogenic factors, including hypothalamic glutamatergic NMDA receptor plasticity, play significant roles in rodent hypertension. Here, we investigated the role of sex and ovarian failure on AMPA receptor plasticity in estrogen-sensitive paraventricular nucleus (PVN) neurons in naïve and angiotensin II (AngII) infused male and female mice and female mice at early and late stages of accelerated ovarian failure (AOF). High-resolution electron microscopy was used to assess the subcellular distribution of AMPA GluA1 in age-matched male and female estrogen receptor beta (ERβ) enhanced green fluorescent protein (EGFP) reporter mice as well as female ERβ-EGFP mice treated with 4-vinylcyclohexene diepoxide. In the absence of AngII, female mice at a late stage of AOF displayed higher levels of GluA1 on the plasma membrane, indicative of functional protein, in ERβ-expressing PVN dendrites when compared to male, naïve female and early stage AOF mice. Following slow-pressor AngII infusion, males, as well as early and late stage AOF females had elevated blood pressure. Significantly, only late stage-AOF female mice infused with AngII had an increase in GluA1 near the plasma membrane in dendrites of ERβ-expressing PVN neurons. In contrast, prior studies reported that plasmalemmal NMDA GluN1 increased in ERβ-expressing PVN dendrites in males and early, but not late stage AOF females. Together, these findings reveal that early and late stage AOF female mice display unique molecular signatures of long-lasting synaptic strength prior to, and following hypertension.
Collapse
Affiliation(s)
- Astrid C Ovalles
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA.
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
39
|
Long T, Yao JK, Li J, Kirshner ZZ, Nelson D, Dougherty GG, Gibbs RB. Estradiol and selective estrogen receptor agonists differentially affect brain monoamines and amino acids levels in transitional and surgical menopausal rat models. Mol Cell Endocrinol 2019; 496:110533. [PMID: 31394142 PMCID: PMC6717664 DOI: 10.1016/j.mce.2019.110533] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/18/2019] [Accepted: 08/03/2019] [Indexed: 10/26/2022]
Abstract
Estrogens have many beneficial effects in the brain. Previously, we evaluated the effects of two models of menopause (surgical vs. transitional) on multiple monoaminergic endpoints in different regions of the adult rat brain in comparison with levels in gonadally intact rats. Here we evaluated the effects of estrogen receptor (ER) agonist treatments in these same two models of menopause. Neurochemical endpoints were evaluated in the hippocampus (HPC), frontal cortex (FCX), and striatum (STR) of adult ovariectomized (OVX) rats and in rats that underwent selective and gradual ovarian follicle depletion by daily injection of 4-vinylcyclohexene-diepoxide (VCD), after 1- and 6-weeks treatment with 17β-estradiol (E2), or with selective ERα (PPT), ERβ (DPN), or GPR30 (G-1) agonists. Endpoints included serotonin (5-HT) and 5-Hydroxyindoleacetic acid, dopamine (DA), 3,4-Dihydroxyphenylacetic acid and homovanillic acid, norepinephrine (NE) and epinephrine, as well as the amino acids tryptophan (TRP) and tyrosine (TYR). Significant differences between the models were detected. OVX rats were much more sensitive to ER agonist treatments than VCD-treated rats. Significant differences between brain regions also were detected. Within OVX rats, more agonist effects were detected in the HPC than in any other region. One interesting finding was the substantial decrease in TRP and TYR detected in the HPC and FCX in response to agonist treatments, particularly in OVX rats. This is on top of the substantial decreases in TRP and TYR previously reported one week after OVX or VCD-treatments in comparison with gonadally intact controls. Other interesting findings included increases in the levels of 5-HT, DA, and NE in the HPC of OVX rats treated with DPN, increases in DA detected in the FCX of OVX rats treated with any of the ER agonists, and increases in 5-HT and DA detected in the STR of OVX rats treated with E2. Many effects that were observed after 1-week of treatment were no longer observed after 6-weeks of treatment, demonstrating that effects were temporary despite continued agonist treatment. Collectively, the results demonstrate significant differences in the effects of ER agonists on monoaminergic endpoints in OVX vs. VCD-treated rats that also were brain region-specific and time dependent. The fact that agonist treatments had lesser effects in VCD treated rats than in OVX rats may help to explain reports of lesser effects of estrogen replacement on cognitive performance in women that have undergone transitional vs. surgical menopause.
Collapse
Affiliation(s)
- Tao Long
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Jeffrey K Yao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Junyi Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ziv Z Kirshner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Doug Nelson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - George G Dougherty
- Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Robert B Gibbs
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
40
|
Marongiu R. Accelerated Ovarian Failure as a Unique Model to Study Peri-Menopause Influence on Alzheimer's Disease. Front Aging Neurosci 2019; 11:242. [PMID: 31551757 PMCID: PMC6743419 DOI: 10.3389/fnagi.2019.00242] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Despite decades of extensive research efforts, efficacious therapies for Alzheimer's disease (AD) are lacking. The multi-factorial nature of AD neuropathology and symptomatology has taught us that a single therapeutic approach will most likely not fit all. Women constitute ~70% of the affected AD population, and pathology and rate of symptoms progression are 2-3 times higher in women than men. Epidemiological data suggest that menopausal estrogen loss may be causative of the more severe symptoms observed in AD women, however, results from clinical trials employing estrogen replacement therapy are inconsistent. AD pathological hallmarks-amyloid β (Aβ), neurofibrillary tangles (NFTs), and chronic gliosis-are laid down during a 20-year prodromal period before clinical symptoms appear, which coincides with the menopause transition (peri-menopause) in women (~45-54-years-old). Peri-menopause is marked by widely fluctuating estrogen levels resulting in periods of irregular hormone-receptor interactions. Recent studies showed that peri-menopausal women have increased indicators of AD phenotype (brain Aβ deposition and hypometabolism), and peri-menopausal women who used hormone replacement therapy (HRT) had a reduced AD risk. This suggests that neuroendocrine changes during peri-menopause may be a trigger that increases risk of AD in women. Studies on sex differences have been performed in several AD rodent models over the years. However, it has been challenging to study the menopause influence on AD due to lack of optimal models that mimic the human process. Recently, the rodent model of accelerated ovarian failure (AOF) was developed, which uniquely recapitulates human menopause, including a transitional peri-AOF period with irregular estrogen fluctuations and a post-AOF stage with low estrogen levels. This model has proven useful in hypertension and cognition studies with wild type animals. This review article will highlight the molecular mechanisms by which peri-menopause may influence the female brain vulnerability to AD and AD risk factors, such as hypertension and apolipoprotein E (APOE) genotype. Studies on these biological mechanisms together with the use of the AOF model have the potential to shed light on key molecular pathways underlying AD pathogenesis for the development of precision medicine approaches that take sex and hormonal status into account.
Collapse
Affiliation(s)
- Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Weill Cornell Medicine, Department of Neurosurgery, Cornell University, New York, NY, United States
| |
Collapse
|
41
|
Kodama L, Gan L. Do Microglial Sex Differences Contribute to Sex Differences in Neurodegenerative Diseases? Trends Mol Med 2019; 25:741-749. [PMID: 31171460 PMCID: PMC7338035 DOI: 10.1016/j.molmed.2019.05.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/23/2019] [Accepted: 05/01/2019] [Indexed: 12/12/2022]
Abstract
Sex differences have been clinically documented in numerous neurodegenerative diseases and yet the reasons for these differences are not well understood. Recent studies have found that microglia, the innate immune cells of the central nervous system, are a key cell type involved in neurodegenerative diseases. This cell type displays sex differences in their expression profiles and function. Could these sex differences in microglia explain the sex differences seen in neurodegenerative diseases? How can we further probe these differences to better understand disease mechanisms? In this Opinion, we highlight the recent evidence that microglia have sex differences, factors that contribute to these differences, and how these differences could shed new light on the pathophysiology of neurological diseases.
Collapse
Affiliation(s)
- Lay Kodama
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Helen and Robert Appel Alzheimer Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
42
|
Ahmadian S, Sheshpari S, Mahdipour M, Pazhang M, Tsai PSJ, Nouri M, Rahbarghazi R, Shahnazi M. Toxic effects of VCD on kidneys and liver tissues: a histopathological and biochemical study. BMC Res Notes 2019; 12:446. [PMID: 31331386 PMCID: PMC6647313 DOI: 10.1186/s13104-019-4490-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE We explored detrimental effects of VCD on non-ovarian tissues such as kidneys and liver 14 days post-drug administration. Twelve rats were randomly assigned into two groups. In VCD group, rats received 160 mg/kgbw VCD intraperitoneally for 15 consequent days. Control rats were injected with VCD-free normal saline. At the respective time point, rats were euthanized, blood and tissue samples were collected. H&E staining was performed to evaluate pathological changes. Serum level of ALT, AST, creatinine and urea were also measured. RESULTS Histological analysis revealed hyperemia and follicular atresia in the ovaries, indicating successful POF induction in rats. In renal tissue, extensive tubular necrosis, focal hemorrhage, hyaline casts, and interstitial nephritis were observed. Analysis of hepatic tissue showed numerous hemorrhagic foci, chronic cholangitis, and hepatocyte necrosis, indicating apparent VCD toxicity of both hepatic and renal tissues. The biochemical evaluation revealed a tendency of increase in ALT, AST, creatinine, and Urea in VCD-treated rats; however, the values did not reach significant level. In conclusion, the induction of POF in rats by VCD correlates with renal and hepatic damages. Commensurate with data from this study, any conclusions from experiments based on VCD-induced premature ovarian failure rats should be reported with caution.
Collapse
Affiliation(s)
- Shahin Ahmadian
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Sepideh Sheshpari
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Shariati St., Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran. .,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran.
| | - Mohammad Pazhang
- Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Pei-Shiue Jason Tsai
- Center for Developmental Biology and Regenerative Medicine Research, National Taiwan University/NTU, Taipei, Taiwan.,Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University/NTU, Taipei, Taiwan
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Daneshgah St., Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Shahnazi
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Shariati St., Tabriz, Iran.
| |
Collapse
|
43
|
Kano M, Hsu JY, Saatcioglu HD, Nagykery N, Zhang L, Morris Sabatini ME, Donahoe PK, Pépin D. Neoadjuvant Treatment With Müllerian-Inhibiting Substance Synchronizes Follicles and Enhances Superovulation Yield. J Endocr Soc 2019; 3:2123-2134. [PMID: 31687639 PMCID: PMC6821214 DOI: 10.1210/js.2019-00190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/16/2019] [Indexed: 12/25/2022] Open
Abstract
Müllerian-inhibiting substance (MIS), also known as anti-Müllerian hormone, is thought to be a negative regulator of primordial follicle activation. We have previously reported that treatment with exogenous MIS can induce complete ovarian suppression within 5 weeks of treatment in mice. To investigate the kinetics of the return of folliculogenesis following the reversal of suppression, we treated animals with recombinant human MIS (rhMIS) protein for 40 days in adult female Nu/Nu mice and monitored the recovery of each follicle type over time. Following cessation of MIS therapy, secondary, and antral follicles returned within 30 days, along with the normalization of reproductive hormones, including LH, FSH, MIS, and Inhibin B. Furthermore, 30 days following MIS pretreatment, the number of antral follicles were significantly higher than controls, and superovulation with timed pregnant mare serum gonadotropin and human chorionic gonadotropin stimulation at this time point resulted in an approximately threefold increased yield of eggs. Use of the combined rhMIS-gonadotropin superovulation regimen in a diminished ovarian reserve (DOR) mouse model, created by 4-vinylcyclohexene dioxide treatment, also resulted in a twofold improvement in the yield of eggs. In conclusion, treatment with rhMIS can induce a reversible ovarian suppression, following which a rapid and synchronized large initial wave of growing follicles can be harnessed to enhance the response to superovulation. Therapies modulating MIS signaling may therefore augment the response to current ovarian stimulation protocols and could be particularly useful to women with DOR or poor responders to controlled ovarian hyperstimulation during in vitro fertilization.
Collapse
Affiliation(s)
- Motohiro Kano
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Y Hsu
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hatice D Saatcioglu
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Nicholas Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - LiHua Zhang
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Mary E Morris Sabatini
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Ovarian failure induced by 4-vinylcyclohexene diepoxide worsens the autonomic cardiovascular response to chronic unpredictable stress in rats. Life Sci 2019; 226:130-139. [DOI: 10.1016/j.lfs.2019.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 11/24/2022]
|
45
|
Ma Y, He X, Qi K, Wang T, Qi Y, Cui L, Wang F, Song M. Effects of environmental contaminants on fertility and reproductive health. J Environ Sci (China) 2019; 77:210-217. [PMID: 30573085 DOI: 10.1016/j.jes.2018.07.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/20/2018] [Accepted: 07/30/2018] [Indexed: 06/09/2023]
Abstract
Recent research indicates that the human infertility rate is increasing. Although various reasons have been hypothesized for the growing infertility rate, environmental contaminants are potentially important causal agents associated with this change. Chemical contaminants are widespread throughout our environment and human exposure is virtually unavoidable. The overall contribution of environmental exposure to infertility is unknown, but studies involving occupational exposure, together with results from animal experiments, suggest that environmental contaminants may adversely affect fertility. We reviewed the adverse effects of environmental exposure on fertility and related reproductive outcomes. Environmental contaminants covered in this review include heavy metals, organic solvents, pesticides and endocrine disrupting chemicals. It is hoped that this review will highlight the need for further research in this area.
Collapse
Affiliation(s)
- Yanmin Ma
- Reproductive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Xin He
- Reproductive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Keyan Qi
- Department of Clinical Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Tong Wang
- Reproductive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Yongchao Qi
- Reproductive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Lele Cui
- Reproductive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China
| | - Fengbang Wang
- Key Laboratory of Environmental Nanotechnology and Health Effects, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Maoyong Song
- Key Laboratory of Environmental Nanotechnology and Health Effects, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
46
|
Hoyer PB, Rice PF, Howard CC, Koevary JW, Dominguez Cooks JP, Hutchens GV, Chambers SK, Craig ZR, Connolly DC, Barton JK. Comparison of Reproductive Function in Female TgMISIIR-TAg Transgenic and Wildtype C57BL/6 Mice. Comp Med 2019; 69:16-21. [PMID: 30591091 PMCID: PMC6382048 DOI: 10.30802/aalas-cm-18-000008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/05/2018] [Accepted: 07/17/2018] [Indexed: 01/23/2023]
Abstract
Transgenic TgMISIIR-TAg (TAg) mice express the oncogenic virus SV40 in Mullerian epithelial cells. Female TAg mice spontaneously develop epithelial ovarian carcinoma, the most common type of ovarian cancer in women. Female TAg mice are infertile, but the reason has not been determined. We therefore investigated whether female TAg mice undergo puberty, demonstrate follicular development, maintain regular cycles, and ovulate. Ovarian cancers in women commonly develop after menopause. The occupational chemical 4-vinylcyclohexene diepoxide (VCD) accelerates follicle degeneration in the ovaries of rats and mice, causing early ovarian failure. We therefore used VCD dosing of mice to develop an animal model for menopause. The purpose of this study was to characterize reproductive parameters in female TAg mice and to investigate whether the onset of ovarian failure due VCD dosing differed between female TAg and WT C57BL/6 mice. As in WT female mice, TAg female mice underwent puberty (vaginal opening) and developed cyclicity in patterns that were similar between the groups. Vehicle-only TAg mice had fewer ovulations (numbers of corpora lutea) than WT animals. VCD exposure delayed the onset of puberty (day of first estrus) in TAg as compared with WT mice. Morphologic evaluation of ovaries revealed many more degenerating follicles in TAg mice than WT mice, and more VCD-dosed TAg mice were in ovarian failure than VCD-dosed WT mice. These results suggest that despite showing similar onset of sexual maturation, TAg mice have increased follicular degeneration and fewer ovulations than WT. These features may contribute to the inability of female TAg mice to reproduce.
Collapse
Affiliation(s)
| | - Photini F Rice
- Biomedical Engineering, The University of Arizona, Tucson, Arizona
| | - Caitlin C Howard
- Biomedical Engineering, The University of Arizona, Tucson, Arizona
| | | | | | | | | | - Zelieann R Craig
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona
| | | | | |
Collapse
|
47
|
Long T, Yao JK, Li J, Kirshner ZZ, Nelson D, Dougherty GG, Gibbs RB. Comparison of transitional vs surgical menopause on monoamine and amino acid levels in the rat brain. Mol Cell Endocrinol 2018; 476:139-147. [PMID: 29738870 PMCID: PMC6120792 DOI: 10.1016/j.mce.2018.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 01/09/2023]
Abstract
Loss of ovarian function has important effects on neurotransmitter production and release with corresponding effects on cognitive performance. To date, there has been little direct comparison of the effects of surgical and transitional menopause on neurotransmitter pathways in the brain. In this study, effects on monoamines, monoamine metabolites, and the amino acids tryptophan (TRP) and tyrosine (TYR) were evaluated in adult ovariectomized (OVX) rats and in rats that underwent selective and gradual ovarian follicle depletion by daily injection of 4-vinylcyclohexene-diepoxide (VCD). Tissues from the hippocampus (HPC), frontal cortex (FCX), and striatum (STR) were dissected and analyzed at 1- and 6-weeks following OVX or VCD treatments. Tissues from gonadally intact rats were collected at proestrus and diestrus to represent neurochemical levels during natural states of high and low estrogens. In gonadally intact rats, higher levels of serotonin (5-HT) were detected at proestrus than at diestrus in the FCX. In addition, the ratio of 5-hydroxyindoleacetic acid (5-HIAA)/5HT in the FCX and HPC was lower at proestrus than at diestrus, suggesting an effect on 5-HT turnover in these regions. No other significant differences between proestrus and diestrus were observed. In OVX- and VCD-treated rats, changes were observed which were both brain region- and time point-dependent. In the HPC levels of norepinephrine, 5-HIAA, TRP and TYR were significantly reduced at 1 week, but not 6 weeks, in both OVX and VCD-treated rats relative to proestrus and diestrus. In the FCX, dopamine levels were elevated at 6 weeks after OVX relative to diestrus. A similar trend was observed at 1 week (but not 6 weeks) following VCD treatment. In the STR, norepinephrine levels were elevated at 1 week following OVX, and HVA levels were elevated at 1 week, but not 6 weeks, following VCD treatment, relative to proestrus and diestrus. Collectively, these data provide the first comprehensive analysis comparing the effects of two models of menopause on multiple neuroendocrine endpoints in the brain. These effects likely contribute to effects of surgical and transitional menopause on brain function and cognitive performance that have been reported.
Collapse
Affiliation(s)
- Tao Long
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Jeffrey K Yao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Junyi Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ziv Z Kirshner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Doug Nelson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - George G Dougherty
- Veterans Affairs Pittsburgh Healthcare System, Medical Research Service, Pittsburgh, PA, 15240, USA
| | - Robert B Gibbs
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
48
|
Shuster SM, Pyzyna B, Mayer LP, Dyer CA. The opportunity for sexual selection and the evolution of non-responsiveness to pesticides, sterility inducers and contraceptives. Heliyon 2018; 4:e00943. [PMID: 30761364 PMCID: PMC6275691 DOI: 10.1016/j.heliyon.2018.e00943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 09/24/2018] [Accepted: 11/14/2018] [Indexed: 11/28/2022] Open
Abstract
We illustrate a method for delaying and possibly eliminating the evolution of non-responsiveness to the treatments now used to control pest populations. Using simulations and estimates of the variance in relative fitness, i.e., the opportunity for selection, in a rat-like mammal, we show that the selection responsible for the evolution of non-responsiveness to pesticides and sterility-inducers, is similar in its action to sexual selection, and for this reason can be orders of magnitude stronger than that which exists for untreated populations. In contrast, we show that when contraceptives are used to reduce the fertility of a pest species, with non-responders embedded within such populations, the opportunity for selection favoring non-responsiveness is reduced to that which is expected by chance alone. In pest species with separate sexes, we show that efforts to control pest populations or to mitigate selection favoring non-responsiveness, are likely to be ineffective when members of one sex are sterilized or killed. We also show that while mating preferences can impede the rate at which resistance evolves, they are more likely to accelerate this process, arguing against the use of sterile male approaches for controlling pests. Our results suggest that contraceptives are more effective at controlling pest populations and slowing the evolution of non-responsiveness than treatments that cause sterilization or death in target species. Furthermore, our results indicate that contraceptives that work differentially on each sex will be most effective in mitigating selection favoring non-responders. Our results have significant implications for the development and application of treatments to manage pests, now and into the future.
Collapse
Affiliation(s)
- Stephen M. Shuster
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011-5640, USA
| | - Brandy Pyzyna
- Senestech, Inc., 3140 N Caden Court, Suite #1, Flagstaff, AZ 86004, USA
| | - Loretta P. Mayer
- Senestech, Inc., 3140 N Caden Court, Suite #1, Flagstaff, AZ 86004, USA
| | - Cheryl A. Dyer
- Senestech, Inc., 3140 N Caden Court, Suite #1, Flagstaff, AZ 86004, USA
| |
Collapse
|
49
|
Kirshner ZZ, Gibbs RB. Use of the REVERT ® total protein stain as a loading control demonstrates significant benefits over the use of housekeeping proteins when analyzing brain homogenates by Western blot: An analysis of samples representing different gonadal hormone states. Mol Cell Endocrinol 2018; 473:156-165. [PMID: 29396126 PMCID: PMC6045444 DOI: 10.1016/j.mce.2018.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/21/2018] [Accepted: 01/22/2018] [Indexed: 01/01/2023]
Abstract
Western blot is routinely used to quantify differences in the levels of target proteins in tissues. Standard methods typically use measurements of housekeeping proteins to control for variations in loading and protein transfer. This is problematic, however, when housekeeping proteins also are affected by experimental conditions such as injury, disease, and/or gonadal hormone manipulations. Our goal was to evaluate an alternative and perhaps superior method for conducting Western blot analysis of brain tissue homogenates from rats with distinct physiologically relevant gonadal hormone states. Tissues were collected from the hippocampus, frontal cortex, and striatum of young adult female rats that either were ovariectomized to model surgical menopause, or were treated with the ovatotoxin 4-vinylcyclohexene diepoxide (VCD) to model transitional menopause. Tissues also were collected from rats with a normal estrous cycle killed at proestrus when estradiol levels are high, and at diestrus when estradiol levels are low. Western blot detection of α-tubulin, β-actin, and GAPDH was performed and were compared for sensitivity and reliability with a fluorescent total protein stain (REVERT®). Results show that the total protein stain was much less variable across samples and had a greater linear range than α-tubulin, β-actin, or GAPDH. The stain was stable and easy to use, and did not interfere with the immunodetection or multiplexed detection of the housekeeping proteins. In addition, we show that normalization of our data to total protein, but not to GAPDH, revealed significant differences in α-tubulin expression in the hippocampus as a function of treatment, and that gel-to-gel consistency in measuring differences between paired samples run on multiple gels was significantly better when data were normalized to total protein than when normalized to GAPDH. These results demonstrate that the REVERT® total protein stain can be used in Western blot analysis of brain tissue homogenates to control for variations in loading and protein transfer, and provides significant advantages over the use of housekeeping proteins for quantifying changes in the levels of multiple target proteins.
Collapse
Affiliation(s)
- Z Z Kirshner
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| | - R B Gibbs
- University of Pittsburgh, Department of Pharmaceutical Sciences, 1004 Salk Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
50
|
Meijer BJ, Wielenga MCB, Hoyer PB, Amos-Landgraf JM, Hakvoort TBM, Muncan V, Heijmans J, van den Brink GR. Colorectal tumor prevention by the progestin medroxyprogesterone acetate is critically dependent on postmenopausal status. Oncotarget 2018; 9:30561-30567. [PMID: 30093969 PMCID: PMC6078141 DOI: 10.18632/oncotarget.25703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/01/2018] [Indexed: 11/25/2022] Open
Abstract
The large randomized placebo controlled trials of the Women’s Health Initiative have shown that the combination of estrogen and progestin medroxyprogesterone acetate (MPA) protects from colorectal cancer in postmenopausal women. No effect was observed in women treated with estrogen alone. This suggests that progesterone, or more specifically the progestin MPA may have chemopreventive activity. The effect of MPA on colorectal carcinogenesis has been difficult to study in animal models. Most models are not affected by either depleting female hormones by ovariectomy or treatment with MPA. Importantly, an ovariectomy fails to reproduce one of the hall marks of the postmenopausal state in women with intact ovaries. That is, the continued production of androgens by the atrophic postmenopausal ovaries. Here we show that adenoma incidence is increased in the vinyl cylcohexene diepoxide (VCD) mouse model of the menopause compared to age matched fertile female mice. Treatment with MPA protected VCD treated mice from adenomagenesis, but had no effect on adenoma numbers in age-matched fertile female mice. Our data show that the protective effect of MPA depends on the postmenopausal state and suggest that MPA monotherapy may be studied as a chemopreventive agent in postmenopausal women.
Collapse
Affiliation(s)
- Bartolomeus J Meijer
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Mattheus C B Wielenga
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Patricia B Hoyer
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | | | - Theodorus B M Hakvoort
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Vanesa Muncan
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jarom Heijmans
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.,Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Gijs R van den Brink
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.,Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands.,GlaxoSmithKline, Medicines Research Center, Stevenage, UK
| |
Collapse
|