1
|
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
|
2
|
White AM, Craig AJ, Richie DL, Corley C, Sadek SM, Barton HN, Gipson CD. Nicotine is an Immunosuppressant: Implications for Women's Health and Disease. J Neuroimmunol 2024; 397:578468. [PMID: 39461120 DOI: 10.1016/j.jneuroim.2024.578468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
A plethora of evidence supports that nicotine, the primary alkaloid in tobacco products that is generally accepted for maintaining use, is immunoregulatory and may function as an immunosuppressant. Women have unique experiences with use of nicotine-containing products and also undergo significant reproductive transitions throughout their lifespan which may be impacted by nicotine use. Within the extant literature, there is conflicting evidence that nicotine may confer beneficial health effects in specific disease states (e.g., in ulcerative colitis). Use prevalence of nicotine-containing products is exceptionally high in individuals presenting with some comorbid disease states that impact immune system health and can be a risk factor for the development of diseases which disproportionately impact women; however, the mechanisms underlying these relationships are largely unclear. Further, little is known regarding the impacts of nicotine's immunosuppressive effects on women's health during the menopausal transition, which is arguably an inflammatory event characterized by a pro-inflammatory peri-menopause period. Given that post-menopausal women are at a higher risk than men for the development of neurodegenerative diseases such as Alzheimer's disease and are also more vulnerable to negative health effects associated with diseases such as HIV-1 infection, it is important to understand how use of nicotine-containing products may impact the immune milieu in women. In this review, we define instances in which nicotine use confers immunosuppressive, anti-inflammatory, or pro-inflammatory effects in the context of comorbid disease states, and focus on how nicotine impacts neuroimmune signaling to maintain use. We posit that regardless of potential health benefits, nicotine use cessation should be a priority in the clinical care of women. The synthesis of this review demonstrates the importance of systematically defining the relationships between volitional nicotine use, immune system function, and comorbid disease states in women to better understand how nicotine impacts women's health and disease.
Collapse
Affiliation(s)
- Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley J Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Daryl L Richie
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah M Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Heather N Barton
- Beebe Health, Gastroenterology and Internal Medicine, Lewes, Delaware, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
3
|
Cornelius SA, Basu U, Zimmern PE, De Nisco NJ. Overcoming challenges in the management of recurrent urinary tract infections. Expert Rev Anti Infect Ther 2024:1-13. [PMID: 39387179 DOI: 10.1080/14787210.2024.2412628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Urinary tract infection (UTI) is a major global health concern. While acute UTIs can usually be effectively treated, recurrent UTIs (rUTIs) impact patients for years, causing significant morbidity and can become refractory to front-line antibiotics. AREAS COVERED This review discusses the risk factors associated with rUTI, current rUTI treatment paradigms, prophylactic strategies, and challenges in rUTI diagnostics. We specifically discuss common risk factors for rUTI, including biological sex, age, menopause status, and diabetes mellitus. We also review recently available evidence for commonly used treatments, from oral antibiotic therapy to intravesical antimicrobials, electrofulguration of chronic cystitis, and the last-resort treatment, cystectomy. We discuss the most current literature evaluating prophylactic strategies for rUTI including long-term antibiotic prophylaxis, estrogen hormone therapy, and dietary supplements. Finally, we address the important role of UTI diagnostics in effective rUTI management and review the strengths and limitations of both current and emerging UTI diagnostic platforms as well as their ability to operate at point-of-care. EXPERT OPINION We discuss the current challenges faced by clinicians in managing rUTI in women and the steps that should be taken so that clinicians, scientists, and patients can work together to better understand the disease and develop better strategies for its management.
Collapse
Affiliation(s)
- Samuel A Cornelius
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Ujjaini Basu
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Philippe E Zimmern
- Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicole J De Nisco
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
- Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Corley C, Craig A, Sadek S, Marusich JA, Chehimi SN, White AM, Holdiness LJ, Reiner BC, Gipson CD. Enhancing translation: A need to leverage complex preclinical models of addictive drugs to accelerate substance use treatment options. Pharmacol Biochem Behav 2024; 243:173836. [PMID: 39067531 PMCID: PMC11344688 DOI: 10.1016/j.pbb.2024.173836] [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: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Preclinical models of addictive drugs have been developed for decades to model aspects of the clinical experience in substance use disorders (SUDs). These include passive exposure as well as volitional intake models across addictive drugs and have been utilized to also measure withdrawal symptomatology and potential neurobehavioral mechanisms underlying relapse to drug seeking or taking. There are a number of Food and Drug Administration (FDA)-approved medications for SUDs, however, many demonstrate low clinical efficacy as well as potential sex differences, and we also note gaps in the continuum of care for certain aspects of clinical experiences in individuals who use drugs. In this review, we provide a comprehensive update on both frequently utilized and novel behavioral models of addiction with a focus on translational value to the clinical experience and highlight the need for preclinical research to follow epidemiological trends in drug use patterns to stay abreast of clinical treatment needs. We then note areas in which models could be improved to enhance the medications development pipeline through efforts to enhance translation of preclinical models. Next, we describe neuroscience efforts that can be leveraged to identify novel biological mechanisms to enhance medications development efforts for SUDs, focusing specifically on advances in brain transcriptomics approaches that can provide comprehensive screening and identification of novel targets. Together, the confluence of this review demonstrates the need for careful selection of behavioral models and methodological parameters that better approximate the clinical experience combined with cutting edge neuroscience techniques to advance the medications development pipeline for SUDs.
Collapse
Affiliation(s)
- Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lexi J Holdiness
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
5
|
Eitmann S, Füredi N, Gaszner B, Kormos V, Berta G, Pólai F, Kovács DK, Balaskó M, Pétervári E. Activity of the hypothalamic neuropeptide Y increases in adult and decreases in old rats. Sci Rep 2024; 14:22676. [PMID: 39349740 PMCID: PMC11442438 DOI: 10.1038/s41598-024-73825-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Middle-aged obesity and aging anorexia with muscle loss (sarcopenia) of old people present public health burden. These alterations may appear both in humans and rodents suggesting the role for regulatory alterations. Previously, we demonstrated that biphasic changes in the weight-reducing (catabolic) effects of neuropeptides of the hypothalamus-adipose tissue axis (e.g. leptin) may contribute to both trends. With regard to the anabolic effects of the hypothalamic neuropeptide Y (NPY) inhibited by leptin, we hypothesized non-linear age-related changes with shifts in the opposite directions. We investigated the orexigenic and hypometabolic effects of intracerebroventricularly administered NPY (hyperphagia induced by NPY injection or changes in food intake, body weight, heart rate, body temperature, locomotor activity during a 7-day NPY infusion), the immunoreactivity and gene expression of NPY in the hypothalamic arcuate nucleus of male Wistar rats of five age groups from young to old. The orexigenic/hypometabolic efficacy and the immunoreactivity of NPY increased in middle-aged animals preceding the peak of adiposity observed in aging rats, then decreased preceding anorexia and weight loss in old rats. These shifts may contribute to the development of both age-related obesity and aging anorexia, sarcopenia, and should be considered in future drug development targeting the NPY system.
Collapse
Affiliation(s)
- Szimonetta Eitmann
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti street, Pécs, 7624, Hungary
| | - Nóra Füredi
- Research Group for Mood Disorders, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Balázs Gaszner
- Research Group for Mood Disorders, Centre for Neuroscience, Medical School, University of Pécs, Pécs, Hungary
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscopic Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Fanni Pólai
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti street, Pécs, 7624, Hungary
| | - Dóra K Kovács
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti street, Pécs, 7624, Hungary
| | - Márta Balaskó
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti street, Pécs, 7624, Hungary
| | - Erika Pétervári
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti street, Pécs, 7624, Hungary.
| |
Collapse
|
6
|
Feng J, Rouse CD, Coogan I, Byrd O, Nguyen E, Taylor L, Garcia S, Lee H, Berchuck A, Murphy SK, Huang Z. Regulation of Age-Related Lipid Metabolism in Ovarian Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611709. [PMID: 39314468 PMCID: PMC11418935 DOI: 10.1101/2024.09.06.611709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Although a lot of effort has been dedicated to ovarian cancer (OC) research, the mortality rate is still among the highest in female gynecologic malignancies. The effects of the aged tumor microenvironment are still being undermined despite age being the highest risk factor in ovarian cancer development and progression. In this study, we have conducted RNA sequencing and lipidomics analysis of gonadal adipose tissues from young and aged rat xenografts before and after ovarian cancer formation. We have found significantly higher tumor formation rates and volumes in aged OC xenograft rat models compared to their young counterparts (p<0.05), suggesting the aged adipose microenvironment (AME) is more susceptible to OC outgrowth. We have revealed significant shifts in the gene expression enrichment from groups of young vs. aged rats before tumor formation, groups of young vs. aged rats when the tumor formed, and groups of aged rats before and after tumor formation. We also observed shifts in the lipid components of the gonadal adipose tissues between young and aged rat xenografts when tumors were generated. Additionally, we found that the aged AME was associated with age-related changes in the immune cell composition, especially inflammation-related cells. The top hits showing the most differences between aged and young adipose tissues were eight genes including S100a8, S100a9, Il1rl1, Lcn2, C3, Hba-a1, Fcna, and Pnpla3, 22 lipids including multiple isoforms of free fatty acids (FFA) and triglyceride (TG), as well as four immune cells including neutrophil, myeloid dendritic cell, T cell CD4+ (non-regulatory), and mast cell activation. The functional correlation among S100a8, S100a9, neutrophil, and FFA (18:3) was also determined. Furthermore, FFA (18:3), which was shown to be downregulated in aged xenograft rats, was capable of inhibiting OC cell proliferation. In conclusion, our study suggested that aging promoted OC proliferation through changes in genes/pathways, lipid metabolism, and immune cells. Targeting the aging adipose microenvironment, particularly lipid metabolism reprogramming, holds promise as a therapeutic strategy for OC, which warrants further investigation. Significance Aging microenvironment of OC may be regulated by S100a8 and S100a9 secreted by adipocytes, preadipocytes, or neutrophils through affecting the lipid metabolism, such as FFA (18:3).
Collapse
|
7
|
Hernandez PA. Response to commentary by Dvir-Ginzberg et al. Osteoarthritis Cartilage 2024; 32:1059. [PMID: 38527664 DOI: 10.1016/j.joca.2024.03.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Affiliation(s)
- Paula A Hernandez
- Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
8
|
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
|
9
|
Bajaj P, Kaur T, Singh AP, Kaur G. Acute sleep deprivation-induced hepatotoxicity and dyslipidemia in middle-aged female rats and its amelioration by butanol extract of Tinospora cordifolia. Lab Anim Res 2024; 40:29. [PMID: 39164744 PMCID: PMC11337769 DOI: 10.1186/s42826-024-00216-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Sleep deprivation (SD) due to an unhealthy lifestyle poses an oxidative challenge and is closely associated with an increased risk and prevalence of different metabolic disorders. Although the negative consequences of SD are well reported on mental health little is known about its detrimental effects on liver function and lipid metabolism. Tinospora cordifolia is reported for its hepatoprotective activity in different pre-clinical model systems. The current study was designed to elucidate the cumulative effects of aging and acute SD on liver functions, oxidative stress, and lipid metabolism, and their management by butanol extract of T. cordifolia (B-TCE) using middle-aged female acyclic rats as the model system. RESULTS Rats were divided into 4 groups: (1) Vehicle-undisturbed (VUD) (2) Vehicle-sleep deprived (VSD) (3) B-TCE pre-treated sleep-deprived (TSD) (4) B-TCE pre-treated undisturbed sleep (TUD). TSD and TUD groups were given 35 mg/kg of B-TCE once daily for 15 days followed by 12 h of sleep deprivation (6 a.m.-6 p.m.) of VSD and TSD group animals using the gentle-handling method while VUD and TUD group animals were left undisturbed. SD of VSD group animals increased oxidative stress, liver function disruption, and dyslipidemia which were ameliorated by B-TCE pre-treatment. Further, B-TCE was observed to target AMPK and its downstream lipid metabolism pathways as well as the p-Akt/cyclinD1/p-bad pathway of cell survival as possible underlying mechanisms of its hepatoprotective activity. CONCLUSIONS These findings suggest that B-TCE being a multi-component extract may be a potential agent in curtailing sleep-related problems and preventing SD-associated hepatotoxicity and dyslipidemia in postmenopausal women.
Collapse
Affiliation(s)
- Payal Bajaj
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Tajpreet Kaur
- Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, 143005, India
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Gurcharan Kaur
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
10
|
Salinero AE, Abi-Ghanem C, Venkataganesh H, Sura A, Smith RM, Thrasher CA, Kelly RD, Hatcher KM, NyBlom V, Shamlian V, Kyaw NR, Belanger KM, Gannon OJ, Stephens SBZ, Zuloaga DG, Zuloaga KL. Treatment with brain specific estrogen prodrug ameliorates cognitive effects of surgical menopause in mice. Horm Behav 2024; 164:105594. [PMID: 38917776 PMCID: PMC11330726 DOI: 10.1016/j.yhbeh.2024.105594] [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: 01/16/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Menopause is an endocrine shift leading to increased vulnerability for cognitive impairment and dementia risk factors, in part due to loss of neuroprotective circulating estrogens. Systemic replacement of estrogen post-menopause has limitations, including risk for estrogen-sensitive cancers. A promising therapeutic approach therefore might be to deliver estrogen only to the brain. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in ovariectomized mice (a surgical menopause model). We treated mice with the prodrug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17β-estradiol only in the brain. Young and middle-aged C57BL/6 J mice received ovariectomy and subcutaneous implant containing vehicle or DHED and underwent cognitive testing to assess memory after 1-3.5 months of treatment. Low and medium doses of DHED did not alter metabolic status in middle-aged mice. In both age groups, DHED treatment improved spatial memory in ovariectomized mice. Additional testing in middle-aged mice showed that DHED treatment improved working and recognition memory in ovariectomized mice. These results lay the foundation for future studies determining if this intervention is as efficacious in models of dementia with comorbid risk factors.
Collapse
Affiliation(s)
- Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Harini Venkataganesh
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Avi Sura
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Rachel M Smith
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Christina A Thrasher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Richard D Kelly
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Katherine M Hatcher
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Vanessa NyBlom
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA; Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY 12222, USA
| | - Victoria Shamlian
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Nyi-Rein Kyaw
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Kasey M Belanger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Shannon B Z Stephens
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA
| | - Damian G Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, 1400 Washington Ave, Biology 325, Albany, NY 12222, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY 12208, USA.
| |
Collapse
|
11
|
Pajski ML, Maroto R, Byrd C, Graber TG. Longitudinal Decline of Exercise Capacity in Male and Female Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605646. [PMID: 39131298 PMCID: PMC11312590 DOI: 10.1101/2024.07.29.605646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The population of older adults is exponentially expanding. Alongside aging comes the onset of chronic disease, decline of functional capacity, and reduced quality of life. Thus, this population increase will stress the capacity and financial viability of health and long-term care systems. Developing pre-clinical models for age-related functional decline is imperative to advancing therapies that extend healthspan and prolong independence. Previously in a cross-sectional study, we established a powerful composite scoring system we termed CFAB (comprehensive functional assessment battery). CFAB measures physical function and exercise capacity using well-validated determinants to measure overall motor function, fore-limb strength, four-limb strength/endurance, aerobic capacity, and volitional exercise/activity rate. In the current work, we used CFAB to track cohorts of male and female C57BL/6 mice over the lifespan (measuring CFAB at 6, 12, 18, 24, and 28 months of age). Overall, we found statistically significantly declining function as the mice aged, with some differences between males and females in trajectory and slope. We also determined that body mass changes presented differently between sexes, and tracked body composition (fat percentage, using magnetic resonance imagery) in females. In a subset of mice, we tracked in vivo contractile physiology noting declines in plantar flexor maximum isometric torque. In summary, our data suggest that males and females declined at different rates. We confirmed the efficacy of CFAB to track longitudinal changes in exercise capacity and physical fitness in both males and females, further validating the system to track age-related functional decline.
Collapse
Affiliation(s)
| | - Rosario Maroto
- University of Texas Medical Branch, Dept. of Biochemistry & Molecular Biology
| | - Chris Byrd
- East Carolina University (ECU) Dept. of Physical Therapy
| | - Ted G. Graber
- East Carolina University (ECU) Dept. of Physical Therapy
- ECU Dept. of Kinesiology
- ECU Dept. of Physiology
- East Carolina Obesity and Diabetes Institute
| |
Collapse
|
12
|
Herrera-Pérez JJ, Hernández-Hernández OT, Flores-Ramos M, Cueto-Escobedo J, Rodríguez-Landa JF, Martínez-Mota L. The intersection between menopause and depression: overview of research using animal models. Front Psychiatry 2024; 15:1408878. [PMID: 39081530 PMCID: PMC11287658 DOI: 10.3389/fpsyt.2024.1408878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Menopausal women may experience symptoms of depression, sometimes even progressing clinical depression requiring treatment to improve quality of life. While varying levels of estrogen in perimenopause may contribute to an increased biological vulnerability to mood disturbances, the effectiveness of estrogen replacement therapy (ERT) in the relief of depressive symptoms remains controversial. Menopausal depression has a complex, multifactorial etiology, that has limited the identification of optimal treatment strategies for the management of this psychiatric complaint. Nevertheless, clinical evidence increasingly supports the notion that estrogen exerts neuroprotective effects on brain structures related to mood regulation. Indeed, research using preclinical animal models continues to improve our understanding of menopause and the effectiveness of ERT and other substances at treating depression-like behaviors. However, questions regarding the efficacy of ERT in perimenopause have been raised. These questions may be answered by further investigation using specific animal models of reduced ovarian function. This review compares and discusses the advantages and pitfalls of different models emulating the menopausal stages and their relationship with the onset of depressive-like signs, as well as the efficacy and mechanisms of conventional and novel ERTs in treating depressive-like behavior. Ovariectomized young rats, middle-to-old aged intact rats, and females treated with reprotoxics have all been used as models of menopause, with stages ranging from surgical menopause to perimenopause. Additionally, this manuscript discusses the impact of organistic and therapeutic variables that may improve or reduce the antidepressant response of females to ERT. Findings from these models have revealed the complexity of the dynamic changes occurring in brain function during menopausal transition, reinforcing the idea that the best approach is timely intervention considering the opportunity window, in addition to the careful selection of treatment according to the presence or absence of reproductive tissue. Additionally, data from animal models has yielded evidence to support new promising estrogens that could be considered as ERTs with antidepressant properties and actions in endocrine situations in which traditional ERTs are not effective.
Collapse
Affiliation(s)
- José Jaime Herrera-Pérez
- Laboratorio de Farmacología Conductual, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Olivia Tania Hernández-Hernández
- Consejo Nacional de Humanidades, Ciencias y Tecnologías Research Fellow. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Mónica Flores-Ramos
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Jonathan Cueto-Escobedo
- Departamento de Investigación Clínica, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa-Enríquez, Mexico
| | | | - Lucía Martínez-Mota
- Laboratorio de Farmacología Conductual, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| |
Collapse
|
13
|
Kim MR, Kim DI, Park SY, Kang HJ, Park SD, Lee JH. The Protective Role of Magnoliae Flos in Preventing Ovotoxicity and Managing Ovarian Function: An In Vitro and In Vivo Study. Int J Mol Sci 2024; 25:6456. [PMID: 38928161 PMCID: PMC11203778 DOI: 10.3390/ijms25126456] [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: 05/02/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Magnoliae Flos (MF) is a medicinal herb widely employed in traditional medicine for relieving sinusitis, allergic rhinitis, headaches, and toothaches. Here, we investigated the potential preventive effects of MF extract (MFE) against 4-vinylcyclohexene diepoxide (VCD)-induced ovotoxicity in ovarian cells and a mouse model of premature ovarian insufficiency (POI). The cytoprotective effects of MFE were assessed using CHO-K1 or COV434 cells. In vivo, B6C3F1 female mice were intraperitoneally injected with VCD for two weeks to induce POI, while MFE was orally administered for four weeks, beginning one week before VCD administration. VCD led to a significant decline in the viabilities of CHO-K1 and COV434 cells and triggered excessive reactive oxygen species (ROS) production and apoptosis specifically in CHO-K1 cells. However, pretreatment with MFE effectively prevented VCD-induced cell death and ROS generation, while also activating the Akt signaling pathway. In vivo, MFE increased relative ovary weights, follicle numbers, and serum estradiol and anti-Müllerian hormone levels versus controls under conditions of ovary failure. Collectively, our results demonstrate that MFE has a preventive effect on VCD-induced ovotoxicity through Akt activation. These results suggest that MFE may have the potential to prevent and manage conditions such as POI and diminished ovarian reserve.
Collapse
Affiliation(s)
- Mi Ra Kim
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea; (M.R.K.); (D.-I.K.); (S.Y.P.)
| | - Dong-Il Kim
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea; (M.R.K.); (D.-I.K.); (S.Y.P.)
| | - Sung Yun Park
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea; (M.R.K.); (D.-I.K.); (S.Y.P.)
| | - Hyo Jin Kang
- Department of Biomedical Laboratory Science, Honam University, Gwangju 62399, Republic of Korea;
| | - Sun-Dong Park
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea; (M.R.K.); (D.-I.K.); (S.Y.P.)
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea; (M.R.K.); (D.-I.K.); (S.Y.P.)
| |
Collapse
|
14
|
Park J, Hu R, Qian Y, Xiong S, El-Sabbagh AS, Ibrahim M, Wang J, Xu Z, Chen Z, Song Q, Song Z, Yan G, Mahmoud AM, He Y, Layden BT, Chen J, Ong SG, Xu P, Jiang Y. Estrogen counteracts age-related decline in beige adipogenesis through the NAMPT-regulated ER stress response. NATURE AGING 2024; 4:839-853. [PMID: 38858606 DOI: 10.1038/s43587-024-00633-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/17/2024] [Indexed: 06/12/2024]
Abstract
Thermogenic beige adipocytes are recognized as potential therapeutic targets for combating metabolic diseases. However, the metabolic advantages that they offer are compromised with aging. Here we show that treating mice with estrogen (E2), a hormone that decreases with age, can counteract the age-related decline in beige adipogenesis when exposed to cold temperature while concurrently enhancing energy expenditure and improving glucose tolerance in mice. Mechanistically, we found that nicotinamide phosphoribosyl transferase (NAMPT) plays a pivotal role in facilitating the formation of E2-induced beige adipocytes, which subsequently suppresses the onset of age-related endoplasmic reticulum (ER) stress. Furthermore, we found that targeting NAMPT signaling, either genetically or pharmacologically, can restore the formation of beige adipocytes by increasing the number of perivascular adipocyte progenitor cells. Conversely, the absence of NAMPT signaling prevents this process. Together, our findings shed light on the mechanisms regulating the age-dependent impairment of beige adipocyte formation and underscore the E2-NAMPT-controlled ER stress pathway as a key regulator of this process.
Collapse
Affiliation(s)
- Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Ruoci Hu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Yanyu Qian
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Shaolei Xiong
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Asma Sana El-Sabbagh
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Meram Ibrahim
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Jaden Wang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Ziqiao Xu
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA
| | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA
- Biostatistics Shared Resource, University of Illinois Cancer Center, Chicago, IL, USA
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Gege Yan
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Abeer M Mahmoud
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Brian T Layden
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
- Jesse Brown Medical VA Medical Center, Chicago, IL, USA
| | - Jiwang Chen
- Division of Pulmonary, Critical Care, Sleep & Allergy, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA.
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA.
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
15
|
Baudin J, Hernandez-Baixauli J, Quesada-Vázquez S, Mulero F, Puiggròs F, Arola L, Caimari A. Combined supplementation with hesperidin, phytosterols and curcumin decreases adiposity and improves metabolic health in ovariectomized rats. Food Funct 2024; 15:4905-4924. [PMID: 38598180 DOI: 10.1039/d3fo05122f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
In recent years many women have looked for alternative therapies to address menopause. Hesperidin, phytosterols and curcumin are bioactive compounds that can ameliorate some cardiovascular risk factors associated with menopause, although there are no data concerning the effects of their combined supplementation. We used ovariectomized (OVX) rats, a postmenopausal model with oestrogen deficiency, to evaluate whether supplementation with a multi-ingredient (MI) including hesperidin, phytosterols and curcumin for 57 days would display beneficial effects against fat mass accretion and metabolic disturbances associated with menopause. Twenty OVX rats were orally supplemented with either MI (OVX-MI) or vehicle (OVX). Furthermore, 10 OVX rats orally received the vehicle along with subcutaneous injections of 17β-oestradiol biweekly (OVX-E2), whereas 10 rats were sham operated and received oral and injected vehicles (control group; SH). MI supplementation partly counteracted the fat mass accretion observed in OVX animals, which was evidenced by decreased total fat mass, adiposity index, the weight of retroperitoneal, inguinal and mesenteric white adipose tissue (MWAT) depots and MWAT adipocyte hypertrophy. These effects were accompanied by a significant decrease in the circulating levels of leptin and the mRNA levels of the fatty acid uptake-related genes Lpl and Cd36 in MWAT. These results were very similar to those observed in OVX-E2 animals. OVX-MI rats also displayed a higher lean body mass, lean/fat mass ratio, adiponectin-to-leptin ratio and insulin sensitivity than their OVX counterparts. Our findings can pave the way for using this MI formulation as an alternative therapy to manage obesity and to improve the cardiometabolic health of menopausal women.
Collapse
Affiliation(s)
- Julio Baudin
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain.
| | - Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
| | - Sergio Quesada-Vázquez
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
| | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain.
| | - Lluís Arola
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain.
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain.
| |
Collapse
|
16
|
Yang SH, Kim YJ, Yang HR, Park SU, Kim JG, Kim JK. Metabolic Profiling in Plasma and Brain Induced by 17β-Estradiol Supplementation in Ovariectomized Mice. ACS OMEGA 2024; 9:18212-18223. [PMID: 38680363 PMCID: PMC11044158 DOI: 10.1021/acsomega.3c10399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024]
Abstract
17β-Estradiol is an ovarian hormone that regulates energy circulation and storage by acting on the central nervous system. However, the metabolic differences between the blood and brain when stimulated by 17β-estradiol are poorly understood. Moreover, research using menopause-induced models to investigate primary metabolites in the blood and brain is limited. Thus, this study aimed to identify metabolic changes in the plasma and brain resulting from 17β-estradiol supplementation in an estrogen-deficient mouse model. Three groups of mice were utilized: sham-operated mice (Sham), ovariectomized mice (OVX), and ovariectomized mice that received a weekly supplementation of 17β-estradiol (E2). Plasma and brain samples from these mice were subjected to metabolic analysis using gas chromatography-time-of-flight-mass spectrometry. Compared with the plasma samples from the Sham and OVX groups, the plasma samples from the E2 group contained higher contents of branched-chain amino acids (BCAAs), such as valine, isoleucine, and leucine. Meanwhile, the brain samples from the E2 group contained higher contents of most metabolites, including BCAAs, neurotransmitters, tricarboxylic acid cycle intermediates, and fatty acids, than those from the two other groups. This study is the first to reveal differences in energy metabolism induced by 17β-estradiol supplementation through brain metabolic profiling of ovariectomized mice, emphasizing the importance of brain metabolic profiling in menopausal hormone research.
Collapse
Affiliation(s)
- So Hwi Yang
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Ye Jin Kim
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Hye Rim Yang
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Sang Un Park
- Department
of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jae Geun Kim
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jae Kwang Kim
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Convergence
Research Center for Insect Vectors, Incheon
National University, Incheon 22012, Republic
of Korea
| |
Collapse
|
17
|
Pereira AAR, Pinto AM, Malerba HN, Toricelli M, Buck HS, Viel TA. Microdose lithium improves behavioral deficits and modulates molecular mechanisms of memory formation in female SAMP-8, a mouse model of accelerated aging. PLoS One 2024; 19:e0299534. [PMID: 38574297 PMCID: PMC10994667 DOI: 10.1371/journal.pone.0299534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/12/2024] [Indexed: 04/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neuronal disorder that leads to the development of dementia. Until nowadays, some therapies may alleviate the symptoms, but there is no pharmacological treatment. Microdosing lithium has been used to modify the pathological characteristics of the disease, with effects in both experimental and clinical conditions. The present work aimed to analyze the effects of this treatment on spatial memory, anxiety, and molecular mechanisms related to long-term memory formation during the aging process of a mouse model of accelerated aging (SAMP-8). Female SAMP-8 showed learning and memory impairments together with disruption of memory mechanisms, neuronal loss, and increased density of senile plaques compared to their natural control strain, the senescence-accelerated mouse resistant (SAMR-1). Chronic treatment with lithium promoted memory maintenance, reduction in anxiety, and maintenance of proteins related to memory formation and neuronal density. The density of senile plaques was also reduced. An increase in the density of gamma-aminobutyric acid A (GABAA) and α7 nicotinic cholinergic receptors was also observed and related to neuroprotection and anxiety reduction. In addition, this microdose of lithium inhibited the activation of glycogen synthase kinase-3beta (GSK-3β), the classical mechanism of lithium cell effects, which could contribute to the preservation of the memory mechanism and reduction in senile plaque formation. This work shows that lithium effects in neuroprotection along the aging process are not related to a unique cellular mechanism but produce multiple effects that slowly protect the brain along the aging process.
Collapse
Affiliation(s)
- Arthur Antonio Ruiz Pereira
- Department of Pharmacology, Institute of Biomedical Sciences, Graduate Course on Pharmacology, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Alessandra Macedo Pinto
- Graduate Course on Gerontology, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Helena Nascimento Malerba
- Department of Pharmacology, Institute of Biomedical Sciences, Graduate Course on Pharmacology, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Mariana Toricelli
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Hudson Sousa Buck
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
- Department of Physiology, University of Mogi das Cruzes, Mogi das Cruzes, Sao Paulo, Brazil
| | - Tania Araujo Viel
- Department of Pharmacology, Institute of Biomedical Sciences, Graduate Course on Pharmacology, Universidade de Sao Paulo, Sao Paulo, Sao Paulo, Brazil
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
- Graduate Course on Gerontology, School of Arts, Sciences and Humanities, Universidade de São Paulo, Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
18
|
Yu J, Joo IL, Bazzigaluppi P, Koletar MM, Cherin E, Stanisz AG, Graham JWC, Demore C, Stefanovic B. Micro-ultrasound based characterization of cerebrovasculature following focal ischemic stroke and upon short-term rehabilitation. J Cereb Blood Flow Metab 2024; 44:461-476. [PMID: 37974304 PMCID: PMC10981404 DOI: 10.1177/0271678x231215004] [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: 03/27/2023] [Revised: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
Notwithstanding recanalization treatments in the acute stage of stroke, many survivors suffer long-term impairments. Physical rehabilitation is the only widely available strategy for chronic-stage recovery, but its optimization is hindered by limited understanding of its effects on brain structure and function. Using micro-ultrasound, behavioral testing, and electrophysiology, we investigated the impact of skilled reaching rehabilitation on cerebral hemodynamics, motor function, and neuronal activity in a rat model of focal ischemic stroke. A 50 MHz micro-ultrasound transducer and intracortical electrophysiology were utilized to characterize neurovascular changes three weeks following focal ischemia elicited by endothelin-1 injection into the sensorimotor cortex. Sprague-Dawley rats were rehabilitated through tray reaching, and their fine skilled reaching was assessed via the Montoya staircase. Focal ischemia led to a sustained deficit in forelimb reaching; and increased tortuosity of the penetrating vessels in the perilesional cortex; with no lateralization of spontaneous neuronal activity. Rehabilitation improved skilled reaching; decreased cortical vascularity; was associated with elevated peri- vs. contralesional hypercapnia-induced flow homogenization and increased perilesional spontaneous cortical neuronal activity. Our study demonstrated neurovascular plasticity accompanying rehabilitation-elicited functional recovery in the subacute stage following stroke, and multiple micro-ultrasound-based markers of cerebrovascular structure and function modified in recovery from ischemia and upon rehabilitation.
Collapse
Affiliation(s)
- Johnson Yu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Illsung L Joo
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Paolo Bazzigaluppi
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- MetaCell, Cagliari, Italy
| | - Margaret M Koletar
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Emmanuel Cherin
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Andrew G Stanisz
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - James WC Graham
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Christine Demore
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Bojana Stefanovic
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Kumari R, Ponte ME, Franczak E, Prom JC, O'Neil MF, Sardiu ME, Lutkewitte AJ, Christenson LK, Shankar K, Morris EM, Thyfault JP. VCD-induced menopause mouse model reveals reprogramming of hepatic metabolism. Mol Metab 2024; 82:101908. [PMID: 38432400 PMCID: PMC10944007 DOI: 10.1016/j.molmet.2024.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Menopause adversely impacts systemic energy metabolism and increases the risk of metabolic disease(s) including hepatic steatosis, but the mechanisms are largely unknown. Dosing female mice with vinyl cyclohexene dioxide (VCD) selectively causes follicular atresia in ovaries, leading to a murine menopause-like phenotype. METHODS In this study, we treated female C57BL6/J mice with VCD (160 mg/kg i.p. for 20 consecutive days followed by verification of the lack of estrous cycling) to investigate changes in body composition, energy expenditure (EE), hepatic mitochondrial function, and hepatic steatosis across different dietary conditions. RESULTS VCD treatment induced ovarian follicular loss and increased follicle-stimulating hormone (FSH) levels in female mice, mimicking a menopause-like phenotype. VCD treatment did not affect body composition, or EE in mice on a low-fat diet (LFD) or in response to a short-term (1-week) high-fat, high sucrose diet (HFHS). However, the transition to a HFHS lowered cage activity in VCD mice. A chronic HFHS diet (16 weeks) significantly increased weight gain, fat mass, and hepatic steatosis in VCD-treated mice compared to HFHS-fed controls. In the liver, VCD mice showed suppressed hepatic mitochondrial respiration on LFD, while chronic HFHS resulted in compensatory increases in hepatic mitochondrial respiration. Also, liver RNA sequencing revealed that VCD promoted global upregulation of hepatic lipid/cholesterol synthesis pathways. CONCLUSION Our findings suggest that the VCD-induced menopause model compromises hepatic mitochondrial function and lipid/cholesterol homeostasis that sets the stage for HFHS diet-induced steatosis while also increasing susceptibility to obesity.
Collapse
Affiliation(s)
- Roshan Kumari
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA
| | - Michael E Ponte
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - Edziu Franczak
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - John C Prom
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maura F O'Neil
- Department of Pathology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mihaela E Sardiu
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew J Lutkewitte
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lane K Christenson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kartik Shankar
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - E Matthew Morris
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA; KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, KS, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA; Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.
| |
Collapse
|
20
|
Gunderson ML, Heer S, Klahr AC. A Pilot Systematic Review and Meta-analysis of Neuroprotective Studies in Female Rodent Models of Ischemic Stroke. Transl Stroke Res 2024; 15:364-377. [PMID: 36763321 DOI: 10.1007/s12975-023-01134-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/29/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Most ischemic stroke (IS) patients go untreated due to limited treatment windows, restrictive eligibility criteria, and poor availability of current clinical therapies. Neuroprotective treatments targeting protracted neurodegeneration are needed yet keep failing in clinical trials. Over half of IS patients are female, and the scarcity of neuroprotective studies using female animals hinders translational success. This pilot review and meta-analysis assessed the relationship between the risk of bias and efficacy of studies testing post-ischemic neuroprotective therapies using female rodent models of IS. We carried out a systematic search of the PubMed database for studies published between 1999 and May 2022, used the CAMARADES checklist to evaluate study quality, and extracted data pertaining to lesion volume and behavioral assessment. We found that 34 studies met our inclusion criteria, with pooled effect sizes depicting a significant treatment effect. However, researchers used mostly healthy young females, administered therapies within short time windows, ignored hormonal influences, and did not assess long-term outcomes. Interestingly, studies failing to report factors impacting internal validity, such as blinding and random allocation, had inflated effect sizes or did not reach statistical significance. There was also a relationship between low study quality and larger effect sizes for functional outcome, stressing the need to follow the existing translational design, reporting, and data analysis guidelines. In this review, we cover previous recommendations and offer our own in hopes that rigorous and meticulous research using female animal models of IS will increase our chances of successful bench-to-bedside translation.
Collapse
Affiliation(s)
- Morgen L Gunderson
- Department of Social Sciences, Augustana Faculty, University of Alberta, Camrose, Canada
| | - Sukhmani Heer
- Department of Social Sciences, Augustana Faculty, University of Alberta, Camrose, Canada
| | - Ana C Klahr
- Department of Social Sciences, Augustana Faculty, University of Alberta, Camrose, Canada.
| |
Collapse
|
21
|
Baek DC, Kang JY, Lee JS, Lee EJ, Son CG. Linking alterations in estrogen receptor expression to memory deficits and depressive behavior in an ovariectomy mouse model. Sci Rep 2024; 14:6854. [PMID: 38514828 PMCID: PMC10958029 DOI: 10.1038/s41598-024-57611-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/20/2024] [Indexed: 03/23/2024] Open
Abstract
The high risk of neurological disorders in postmenopausal women is an emerging medical issue. Based on the hypothesis of altered estrogen receptors (ERα and β) after the decline of estrogen production, we investigated the changes in ERs expressions across brain regions and depressive/amnesic behaviors. C57BL/6J female mice were ovariectomized (OVX) to establish a menopausal condition. Along with behavior tests (anxiety, depression, and memory), the expression of ERs, microglial activity, and neuronal activity was measured in six brain regions (hippocampus, prefrontal cortex, striatum, raphe nucleus, amygdala, and hypothalamus) from 4 to 12 weeks after OVX. Mice exhibited anxiety- and depressive-like behaviors, as well as memory impairment. These behavioral alterations have been linked to a suppression in the expression of ERβ. The decreased ERβ expression coincided with microglial-derived neuroinflammation, as indicated by notable activations of Ionized calcium-binding adapter molecule 1 and Interleukin-1beta. Additionally, the activity of brain-derived neurotrophic factor (BDNF), particularly in the hippocampus, decreased in a time-dependent manner from 4 to 12 weeks post-OVX. Our study provides evidence shedding light on the susceptibility to memory impairment and depression in women after menopause. This susceptibility is associated with the suppression of ERβ and alteration of ERα in six brain regions.
Collapse
Affiliation(s)
- Dong-Cheol Baek
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Ji-Yun Kang
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Jin-Seok Lee
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Eun-Jung Lee
- Department of Korean Rehabilitation Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea
| | - Chang-Gue Son
- Institute of Bioscience & Integrative Medicine, Daejeon Korean Hospital of Daejeon University, Daedukdae-ro 176 bun-gil 75, Daejeon, 35235, Republic of Korea.
| |
Collapse
|
22
|
Arellano JI, Duque A, Rakic P. A coming-of-age story: adult neurogenesis or adolescent neurogenesis in rodents? Front Neurosci 2024; 18:1383728. [PMID: 38505771 PMCID: PMC10948509 DOI: 10.3389/fnins.2024.1383728] [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: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 03/21/2024] Open
Abstract
It is surprising that after more than a century using rodents for scientific research, there are no clear, consensual, or consistent definitions for when a mouse or a rat becomes adult. Specifically, in the field of adult hippocampal neurogenesis, where this concept is central, there is a trend to consider that puberty marks the start of adulthood and is not uncommon to find 30-day-old mice being described as adults. However, as others discussed earlier, this implies an important bias in the perceived importance of this trait because functional studies are normally done at very young ages, when neurogenesis is at its peak, disregarding middle aged and old animals that exhibit very little generation of new neurons. In this feature article we elaborate on those issues and argue that research on the postnatal development of mice and rats in the last 3 decades allows to establish an adolescence period that marks the transition to adulthood, as occurs in other mammals. Adolescence in both rat and mice ends around postnatal day 60 and therefore this age can be considered the onset of adulthood in both species. Nonetheless, to account for inter-individual, inter-strain differences in maturation and for possible delays due to environmental and social conditions, 3 months of age might be a safer option to consider mice and rats bona fide adults, as suggested by The Jackson Labs.
Collapse
Affiliation(s)
- Jon I. Arellano
- Department of Neuroscience, Yale University, New Haven, CT, United States
| | - Alvaro Duque
- Department of Neuroscience, Yale University, New Haven, CT, United States
| | - Pasko Rakic
- Department of Neuroscience, Yale University, New Haven, CT, United States
- Kavli Institute for Neuroscience at Yale, Yale University, New Haven, CT, United States
| |
Collapse
|
23
|
Oknińska M, Duda MK, Czarnowska E, Bierła J, Paterek A, Mączewski M, Mackiewicz U. Sex- and age-dependent susceptibility to ventricular arrhythmias in the rat heart ex vivo. Sci Rep 2024; 14:3460. [PMID: 38342936 PMCID: PMC10859380 DOI: 10.1038/s41598-024-53803-9] [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: 11/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024] Open
Abstract
The incidence of life-threatening ventricular arrhythmias, the most common cause of sudden cardiac death (SCD), depends largely on the arrhythmic substrate that develops in the myocardium during the aging process. There is a large deficit of comparative studies on the development of this substrate in both sexes, with a particular paucity of studies in females. To identify the substrates of arrhythmia, fibrosis, cardiomyocyte hypertrophy, mitochondrial density, oxidative stress, antioxidant defense and intracellular Ca2+ signaling in isolated cardiomyocytes were measured in the hearts of 3- and 24-month-old female and male rats. Arrhythmia susceptibility was assessed in ex vivo perfused hearts after exposure to isoproterenol (ISO) and hydrogen peroxide (H2O2). The number of ventricular premature beats (PVBs), ventricular tachycardia (VT) and ventricular fibrillation (VF) episodes, as well as intrinsic heart rate, QRS and QT duration, were measured in ECG signals recorded from the surfaces of the beating hearts. After ISO administration, VT/VFs were formed only in the hearts of males, mainly older ones. In contrast, H2O2 led to VT/VF formation in the hearts of rats of both sexes but much more frequently in older males. We identified several components of the arrhythmia substrate that develop in the myocardium during the aging process, including high spontaneous ryanodine receptor activity in cardiomyocytes, fibrosis of varying severity in different layers of the myocardium (nonheterogenic fibrosis), and high levels of oxidative stress as measured by nitrated tyrosine levels. All of these elements appeared at a much greater intensity in male individuals during the aging process. On the other hand, in aging females, antioxidant defense at the level of H2O2 detoxification, measured as glutathione peroxidase expression, was weaker than that in males of the same age. We showed that sex has a significant effect on the development of an arrhythmic substrate during aging. This substrate determines the incidence of life-threatening ventricular arrhythmias in the presence of additional stimuli with proarrhythmic potential, such as catecholamine stimulation or oxidative stress, which are constant elements in the pathomechanism of most cardiovascular diseases.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Monika Katarzyna Duda
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-736, Warsaw, Poland
- Department of Pathology, Medical University of Warsaw, Żwirki i Wigury 61, 02-091, Warsaw, Poland
| | - Joanna Bierła
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-736, Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland.
| |
Collapse
|
24
|
Holter KM, Lekander AD, Pierce BE, Sands LP, Gould RW. Use of Quantitative Electroencephalography to Inform Age- and Sex-Related Differences in NMDA Receptor Function Following MK-801 Administration. Pharmaceuticals (Basel) 2024; 17:237. [PMID: 38399452 PMCID: PMC10892193 DOI: 10.3390/ph17020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Sex- and age-related differences in symptom prevalence and severity have been widely reported in patients with schizophrenia, yet the underlying mechanisms contributing to these differences are not well understood. N-methyl-D-aspartate (NMDA) receptor hypofunction contributes to schizophrenia pathology, and preclinical models often use NMDA receptor antagonists, including MK-801, to model all symptom clusters. Quantitative electroencephalography (qEEG) represents a translational approach to measure neuronal activity, identify targetable biomarkers in neuropsychiatric disorders and evaluate possible treatments. Abnormalities in gamma power have been reported in patients with schizophrenia and correspond to psychosis and cognitive impairment. Further, as gamma power reflects cortical glutamate and GABA signaling, it is highly sensitive to changes in NMDA receptor function, and NMDA receptor antagonists aberrantly increase gamma power in rodents and humans. To evaluate the role of sex and age on NMDA receptor function, MK-801 (0.03-0.3 mg/kg, SC) was administered to 3- and 9-month-old male and female Sprague-Dawley rats that were implanted with wireless EEG transmitters to measure cortical brain function. MK-801-induced elevations in gamma power were observed in 3-month-old male and female and 9-month-old male rats. In contrast, 9-month-old female rats demonstrated blunted maximal elevations across a wide dose range. Importantly, MK-801-induced hyperlocomotor effects, a common behavioral screen used to examine antipsychotic-like activity, were similar across all groups. Overall, sex-by-age-related differences in gamma power support using qEEG as a translational tool to evaluate pathological progression and predict treatment response across a heterogeneous population.
Collapse
Affiliation(s)
| | | | | | | | - Robert W. Gould
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.M.H.)
| |
Collapse
|
25
|
Prokai-Tatrai K, Prokai L. The impact of 17β-estradiol on the estrogen-deficient female brain: from mechanisms to therapy with hot flushes as target symptoms. Front Endocrinol (Lausanne) 2024; 14:1310432. [PMID: 38260155 PMCID: PMC10800853 DOI: 10.3389/fendo.2023.1310432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Sex steroids are essential for whole body development and functions. Among these steroids, 17β-estradiol (E2) has been known as the principal "female" hormone. However, E2's actions are not restricted to reproduction, as it plays a myriad of important roles throughout the body including the brain. In fact, this hormone also has profound effects on the female brain throughout the life span. The brain receives this gonadal hormone from the circulation, and local formation of E2 from testosterone via aromatase has been shown. Therefore, the brain appears to be not only a target but also a producer of this steroid. The beneficial broad actions of the hormone in the brain are the end result of well-orchestrated delayed genomic and rapid non-genomic responses. A drastic and steady decline in circulating E2 in a female occurs naturally over an extended period of time starting with the perimenopausal transition, as ovarian functions are gradually declining until the complete cessation of the menstrual cycle. The waning of endogenous E2 in the blood leads to an estrogen-deficient brain. This adversely impacts neural and behavioral functions and may lead to a constellation of maladies such as vasomotor symptoms with varying severity among women and, also, over time within an individual. Vasomotor symptoms triggered apparently by estrogen deficiency are related to abnormal changes in the hypothalamus particularly involving its preoptic and anterior areas. However, conventional hormone therapies to "re-estrogenize" the brain carry risks due to multiple confounding factors including unwanted hormonal exposure of the periphery. In this review, we focus on hot flushes as the archetypic manifestation of estrogen deprivation in the brain. Beyond our current mechanistic understanding of the symptoms, we highlight the arduous process and various obstacles of developing effective and safe therapies for hot flushes using E2. We discuss our preclinical efforts to constrain E2's beneficial actions to the brain by the DHED prodrug our laboratory developed to treat maladies associated with the hypoestrogenic brain.
Collapse
Affiliation(s)
- Katalin Prokai-Tatrai
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | | |
Collapse
|
26
|
Chessum JE, Shaya SA, Rajab D, Aftabjahani A, Zhou J, Weitz JI, Gross PL, Kim PY. Thrombin-activatable fibrinolysis inhibitor and sex modulate thrombus stability and pulmonary embolism burden in a murine model. J Thromb Haemost 2024; 22:263-270. [PMID: 37751849 DOI: 10.1016/j.jtha.2023.09.016] [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: 04/29/2022] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Thrombin-activatable fibrinolysis inhibitor (TAFI) levels are positively correlated with the risk of thrombosis. The mechanism of how TAFI affects venous thromboembolism (VTE) remains uncertain. In addition, the role of sex on the risk of VTE has also been studied. However, their association also remains unclear. OBJECTIVES To investigate how TAFI and/or sex affect venous thrombus stability and consequent pulmonary embolism (PE). METHODS Ferric chloride-induced thrombi were formed within the femoral veins of male and female wild-type (WT) or TAFI-knockout (Cpb2-/-) mice. Thrombi were imaged over 2 hours using intravital videomicroscopy to quantify embolization and thrombus size over time. Lungs were examined by immunohistochemistry to quantify (a) emboli and (b) fibrin composition of these emboli. RESULTS Embolization events in female mice were higher than in males (7.9-fold in WT and 3.1-fold in Cpb2-/- mice). Although the maximal thrombus sizes were not different across groups, Cpb2-/- mice had thrombi that were, on average, 24% smaller at the end of the 2-hour experiment than WT mice. Loss of TAFI led to a 4.0- and 2.8-fold increase in PE burden in males and females, respectively, while sex had no influence. Pulmonary emboli in Cpb2-/- mice had higher fibrin composition compared with WT mice. CONCLUSION Female mice had less stable venous thrombi than male mice, suggesting a higher risk of PE in females with deep vein thrombosis. Mice lacking TAFI had more thrombus degradation and higher PE burden than WT mice. These results confirm the role of TAFI in venous thrombosis.
Collapse
Affiliation(s)
- James E Chessum
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Shana A Shaya
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dana Rajab
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Ali Aftabjahani
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ji Zhou
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Peter L Gross
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Paul Y Kim
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
27
|
Dalla C, Jaric I, Pavlidi P, Hodes GE, Kokras N, Bespalov A, Kas MJ, Steckler T, Kabbaj M, Würbel H, Marrocco J, Tollkuhn J, Shansky R, Bangasser D, Becker JB, McCarthy M, Ferland-Beckham C. Practical solutions for including sex as a biological variable (SABV) in preclinical neuropsychopharmacological research. J Neurosci Methods 2024; 401:110003. [PMID: 37918446 PMCID: PMC10842858 DOI: 10.1016/j.jneumeth.2023.110003] [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: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Recently, many funding agencies have released guidelines on the importance of considering sex as a biological variable (SABV) as an experimental factor, aiming to address sex differences and avoid possible sex biases to enhance the reproducibility and translational relevance of preclinical research. In neuroscience and pharmacology, the female sex is often omitted from experimental designs, with researchers generalizing male-driven outcomes to both sexes, risking a biased or limited understanding of disease mechanisms and thus potentially ineffective therapeutics. Herein, we describe key methodological aspects that should be considered when sex is factored into in vitro and in vivo experiments and provide practical knowledge for researchers to incorporate SABV into preclinical research. Both age and sex significantly influence biological and behavioral processes due to critical changes at different timepoints of development for males and females and due to hormonal fluctuations across the rodent lifespan. We show that including both sexes does not require larger sample sizes, and even if sex is included as an independent variable in the study design, a moderate increase in sample size is sufficient. Moreover, the importance of tracking hormone levels in both sexes and the differentiation between sex differences and sex-related strategy in behaviors are explained. Finally, the lack of robust data on how biological sex influences the pharmacokinetic (PK), pharmacodynamic (PD), or toxicological effects of various preclinically administered drugs to animals due to the exclusion of female animals is discussed, and methodological strategies to enhance the rigor and translational relevance of preclinical research are proposed.
Collapse
Affiliation(s)
- Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Ivana Jaric
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Pavlina Pavlidi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Georgia E Hodes
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24060, USA
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece; First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Anton Bespalov
- Partnership for Assessment and Accreditation of Scientific Practice (PAASP GmbH), Heidelberg, Germany
| | - Martien J Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | | | - Mohamed Kabbaj
- Department of Biomedical Sciences & Neurosciences, College of Medicine, Florida State University, USA
| | - Hanno Würbel
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jordan Marrocco
- Department of Biology, Touro University, New York, NY 10027, USA
| | | | - Rebecca Shansky
- Department of Psychology, Northeastern University, Boston, MA 02128, USA
| | - Debra Bangasser
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Jill B Becker
- Department of Psychology and Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret McCarthy
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore MD, USA
| | | |
Collapse
|
28
|
Kumari R, Ponte ME, Franczak E, Prom JC, O'Neil MF, Sardiu ME, Lutkewitte AJ, Shankar K, Morris EM, Thyfault JP. VCD-induced menopause mouse model reveals reprogramming of hepatic metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571644. [PMID: 38168213 PMCID: PMC10760158 DOI: 10.1101/2023.12.14.571644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Menopause adversely impacts systemic energy metabolism and increases the risk of metabolic disease(s) including hepatic steatosis, but the mechanisms are largely unknown. Dosing female mice with vinyl cyclohexene dioxide (VCD) selectively causes follicular atresia in ovaries, leading to a murine menopause-like phenotype. In this study, we treated female C57BL6/J mice with VCD (160mg/kg i.p. for 20 consecutive days followed by verification of the lack of estrous cycling) to investigate changes in body composition, energy expenditure (EE), hepatic mitochondrial function, and hepatic steatosis across different dietary conditions. VCD treatment induced ovarian follicular loss and increased follicle-stimulating hormone (FSH) levels in female mice, mimicking a menopause-like phenotype. VCD treatment did not affect body composition, or EE in mice on a low-fat diet or in response to a short-term (1-week) high-fat, high sucrose diet (HFHS). However, the transition to a HFHS lowered cage activity in VCD mice. A chronic HFHS diet (16 weeks) significantly increased weight gain, fat mass, and hepatic steatosis in VCD-treated mice compared to HFHS-fed controls. In the liver, VCD mice showed suppressed hepatic mitochondrial respiration on LFD, while chronic HFHS diet resulted in compensatory increases in hepatic mitochondrial respiration. Also, liver RNA sequencing revealed that VCD promoted global upregulation of hepatic lipid/cholesterol synthesis pathways. Our findings suggest that the VCD- induced menopause model compromises hepatic mitochondrial function and lipid/cholesterol homeostasis that sets the stage for HFHS diet-induced steatosis while also increasing susceptibility to obesity.
Collapse
|
29
|
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
|
30
|
Gilmer G, Hettinger ZR, Tuakli-Wosornu Y, Skidmore E, Silver JK, Thurston RC, Lowe DA, Ambrosio F. Female aging: when translational models don't translate. NATURE AGING 2023; 3:1500-1508. [PMID: 38052933 PMCID: PMC11099540 DOI: 10.1038/s43587-023-00509-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/25/2023] [Indexed: 12/07/2023]
Abstract
For many pathologies associated with aging, female patients present with higher morbidity and more frequent adverse events from treatments compared to male patients. While preclinical models are the foundation of our mechanistic understanding of age-related diseases, the most common models fail to recapitulate archetypical female aging trajectories. For example, while over 70% of the top age-related diseases are influenced by the systemic effects of reproductive senescence, we found that preclinical studies that include menopausal phenotypes modeling those seen in humans make up <1% of published aging biology research. The long-term impacts of pregnancy, birthing and breastfeeding are also typically omitted from preclinical work. In this Perspective, we summarize limitations in the most commonly used aging models, and we provide recommendations for better incorporating menopause, pregnancy and other considerations of sex in vivo and in vitro. Lastly, we outline action items for aging biology researchers, journals, funding agencies and animal providers to address this gap.
Collapse
Affiliation(s)
- Gabrielle Gilmer
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding Rehabilitation, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Cellular and Molecular Pathology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary R Hettinger
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding Rehabilitation, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
- Department of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yetsa Tuakli-Wosornu
- Department of Social and Behavioral Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elizabeth Skidmore
- Department of Occupational Therapy, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julie K Silver
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Massachusetts General Hospital, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Brigham and Women's Hospital, Boston, MA, USA
| | - Rebecca C Thurston
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding Rehabilitation, Boston, MA, USA.
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, MA, USA.
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
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
|
32
|
Crișan S, Pop AL, Lacatusu I, Badea N, Mustaciosu C, Radu M, Varlas VN, Peneş ON, Ciobanu AM, Ghica M, Voicu SN, Udeanu DI. Safety of Innovative Nanotechnology Oral Formulations Loaded with Bioactive Menopause Molecules: Influence of Genotoxicity and Biochemical Parameters on a Menopausal Rat Model. Nutrients 2023; 15:4951. [PMID: 38068809 PMCID: PMC10708031 DOI: 10.3390/nu15234951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
In recent years, nanoparticles have gained significant importance due to their unique properties, such as pharmacological, electrical, optical, and magnetic abilities, contributing to the growth of the science and technology sector. Particular naturally derived biomolecules with beneficial effects on menopause disorder have been the subject of studies of pharmaceutical formulation to obtain alternative pharmaceutical forms with increased bioavailability and without side effects, as in nanostructured lipid carriers (NLCs) loaded with such active ingredients. In the present study, one stage of a broader project, we have performed pharmacotoxicology studies for six combinatory innovative nanocapsule pharmaceutical forms containing active natural biomolecules before considering them as oral formulas for (1) in vitro toxicity studies on culture cells and (2) in vivo preclinical studies on a surgically induced menopause model of Wistar female rats, and the influence of the NLCs on key biochemical parameters: lipid profile (TG, Chol, HDL), glycemic markers (Gli), bone markers (Pac, Palc, Ca, phosphorus), renal markers (Crea, urea, URAC), inflammation (TNF), oxidative stress (GSH, MDA), and estrogen-progesterone hormonal profile. The micronucleus test did not reveal the genotoxicity of the tested compounds; the menopause model showed no significant safety concerns for the six tested formulas evaluated using the blood biochemical parameters; and the results showed the potential hypoglycemic, hypolipidemic, hypouricemic, and antioxidant potential of one of the tested formulas containing nano diosgenin and glycyrrhizic acid.
Collapse
Affiliation(s)
- Simona Crișan
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania; (S.C.); (A.L.P.); (A.M.C.); (M.G.); (D.I.U.)
- R&D Center, AC HELCOR, Victor Babes St., 430082 Baia Mare, Romania
| | - Anca Lucia Pop
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania; (S.C.); (A.L.P.); (A.M.C.); (M.G.); (D.I.U.)
| | - Ioana Lacatusu
- Faculty of Applied Chemistry and Materials Science, The Polytechnic University of Bucharest, Polizu No 1, 011061 Bucharest, Romania; (I.L.); (N.B.)
| | - Nicoleta Badea
- Faculty of Applied Chemistry and Materials Science, The Polytechnic University of Bucharest, Polizu No 1, 011061 Bucharest, Romania; (I.L.); (N.B.)
| | - Cosmin Mustaciosu
- Horia Hulubei National Institute for Physics and Nuclear Engineering IFIN-HH, 077125 Bucharest, Romania; (C.M.); (M.R.)
| | - Mihai Radu
- Horia Hulubei National Institute for Physics and Nuclear Engineering IFIN-HH, 077125 Bucharest, Romania; (C.M.); (M.R.)
| | - Valentin Nicolae Varlas
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Ovidiu Nicolae Peneş
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Anne Marie Ciobanu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania; (S.C.); (A.L.P.); (A.M.C.); (M.G.); (D.I.U.)
| | - Manuela Ghica
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania; (S.C.); (A.L.P.); (A.M.C.); (M.G.); (D.I.U.)
| | - Sorina Nicoleta Voicu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania;
| | - Denisa Ioana Udeanu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania; (S.C.); (A.L.P.); (A.M.C.); (M.G.); (D.I.U.)
| |
Collapse
|
33
|
Rowe AA, Issioui Y, Johnny B, Wert KJ. Murine Orchiectomy and Ovariectomy to Reduce Sex Hormone Production. J Vis Exp 2023:10.3791/64379. [PMID: 38047564 PMCID: PMC10868640 DOI: 10.3791/64379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023] Open
Abstract
Sex hormone signaling plays a critical role in multiple organ systems as well as in the progression of various diseases, including neurodegenerative disease. The manipulation of sex hormone levels in the murine model system allows for the study of their impact on organs/tissues and within disease progression. Orchiectomy - the surgical removal of the testes - and ovariectomy - the surgical removal of the ovaries - provide a method to deplete the endogenous sex hormones so that the precise hormone levels can be provided through drug or other delivery methods. Here, we provide rapid and minimally invasive methods for both orchiectomy and ovariectomy in the murine model system for the reduction of sex hormones. This protocol details the surgical preparation and excision of the testes through the scrotal sac, and excision of the ovaries via two incisions in the right and left lateral dorsum.
Collapse
Affiliation(s)
- Ashley A Rowe
- Department of Ophthalmology, UT Southwestern Medical Center
| | - Yacine Issioui
- Department of Ophthalmology, UT Southwestern Medical Center
| | | | - Katherine J Wert
- Department of Ophthalmology, UT Southwestern Medical Center; Department of Molecular Biology, UT Southwestern Medical Center; Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center;
| |
Collapse
|
34
|
Ince LM, Darling JS, Sanchez K, Bell KS, Melbourne JK, Davis LK, Nixon K, Gaudet AD, Fonken LK. Sex differences in microglia function in aged rats underlie vulnerability to cognitive decline. Brain Behav Immun 2023; 114:438-452. [PMID: 37709153 PMCID: PMC10790303 DOI: 10.1016/j.bbi.2023.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/07/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
Aging is associated with a significant shift in immune system reactivity ("inflammaging"), as basal inflammation increases but protective responses to infection are compromised. The immune system exhibits considerable sex differences, which may influence the process of inflammaging, including immune cell activation and behavioral consequences of immune signaling (i.e., impaired memory). Here, we test the hypothesis that sex differences in immune aging may mediate sex differences in cognitive decline. Aged male and female rats received peripheral immune stimulation using lipopolysaccharide (LPS), then molecular, cellular, and behavioral outcomes were assessed. We observed that LPS-treated aged male rats showed cognitive impairment and increased neuroinflammatory responses relative to adult males. In contrast, aged female rats did not display these aging-related deficits. Using transcriptomic and flow cytometry analyses, we further observed significant age- and sex- dependent changes in immune cell populations in the brain parenchyma and meninges, indicating a broad shift in the neuroinflammatory environment that may potentiate these behavioral effects. Ovariectomized aged female rats were also resistant to inflammation-induced memory deficits, indicating that ovarian hormones are not required for the attenuated neuroinflammation in aged females. Overall, our results indicate that males have amplified inflammatory priming with age, which contributes to age-associated cognitive decline. Our findings highlight sexual dimorphism in mechanisms of aging, and suggest that sex is a crucial consideration for identifying therapies for aging and neuroinflammation.
Collapse
Affiliation(s)
- Louise M Ince
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jeffrey S Darling
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kevin Sanchez
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kiersten S Bell
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer K Melbourne
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Lourdes K Davis
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Kimberly Nixon
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Andrew D Gaudet
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA; Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
35
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
36
|
Metcalf CA, Duffy KA, Page CE, Novick AM. Cognitive Problems in Perimenopause: A Review of Recent Evidence. Curr Psychiatry Rep 2023; 25:501-511. [PMID: 37755656 PMCID: PMC10842974 DOI: 10.1007/s11920-023-01447-3] [Citation(s) in RCA: 2] [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] [Accepted: 08/08/2023] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW To review recent research regarding cognitive problems during perimenopause, including which menopause-related symptoms, demographic variables, stress exposures, and neural biomarkers are associated with cognitive problems and which interventions demonstrate efficacy at improving cognitive performance. RECENT FINDINGS Cognitive problems are common during perimenopause and have a significant impact on a substantial proportion of women. Evidence continues to indicate that verbal learning and verbal memory are the cognitive functions that are most negatively affected during perimenopause, and new research suggests that perimenopause may also be associated with deficits in processing speed, attention, and working memory. Recent research suggests that the cognitive profiles of women transitioning through perimenopause are heterogenous - with some showing strengths and others demonstrating weaknesses in particular cognitive domains. Depression, sleep problems, and vasomotor symptoms in perimenopause may be associated with cognitive difficulties. Recent neuroimaging studies are identifying changes in activity patterns within brain regions that correlate with cognitive performance in perimenopause, but future causal studies are needed to understand the neural mechanisms of cognitive problems during this time. Although clinical treatment studies for cognitive concerns have historically focused on postmenopause, some small trials in perimenopausal samples have been conducted recently but are frequently underpowered. Current guidelines from the North American Menopause Society do not support the use of hormone therapy at any age for cognitive problems. Animal research demonstrates that estradiol and levonorgestrel combined may alleviate working memory problems. Much progress has been made in understanding how perimenopause impacts cognition, and more research is needed to better identify who is at highest risk and how to meaningfully prevent and alleviate cognitive problems during this reproductive stage. Larger-scale randomized intervention trials specifically during perimenopause are urgently needed to address cognitive concerns in this population of women. More consistent reproductive staging, inclusion of covariates, and analyses examining perimenopause specifically would improve study quality and the ability to draw clear conclusions from this research.
Collapse
Affiliation(s)
- Christina A Metcalf
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, 1890 N. Revere Court MS F546, Aurora, CO, 80045, USA.
| | - Korrina A Duffy
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, 1890 N. Revere Court MS F546, Aurora, CO, 80045, USA
| | - Chloe E Page
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, 1890 N. Revere Court MS F546, Aurora, CO, 80045, USA
| | - Andrew M Novick
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, 1890 N. Revere Court MS F546, Aurora, CO, 80045, USA
| |
Collapse
|
37
|
Jiménez-Herrera R, Contreras A, Djebari S, Mulero-Franco J, Iborra-Lázaro G, Jeremic D, Navarro-López J, Jiménez-Díaz L. Systematic characterization of a non-transgenic Aβ 1-42 amyloidosis model: synaptic plasticity and memory deficits in female and male mice. Biol Sex Differ 2023; 14:59. [PMID: 37716988 PMCID: PMC10504764 DOI: 10.1186/s13293-023-00545-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND The amyloid-β (Aβ) cascade is one of the most studied theories linked to AD. In multiple models, Aβ accumulation and dyshomeostasis have shown a key role in AD onset, leading to excitatory/inhibitory imbalance, the impairments of synaptic plasticity and oscillatory activity, and memory deficits. Despite the higher prevalence of Alzheimer's disease (AD) in women compared to men, the possible sex difference is scarcely explored and the information from amyloidosis transgenic mice models is contradictory. Thus, given the lack of data regarding the early stages of amyloidosis in female mice, the aim of this study was to systematically characterize the effect of an intracerebroventricular (icv.) injection of Aβ1-42 on hippocampal-dependent memory, and on associated activity-dependent synaptic plasticity in the hippocampal CA1-CA3 synapse, in both male and female mice. METHODS To do so, we evaluated long term potentiation (LTP) with ex vivo electrophysiological recordings as well as encoding and retrieval of spatial (working, short- and long-term) and exploratory habituation memories using Barnes maze and object location, or open field habituation tasks, respectively. RESULTS Aβ1-42 administration impaired all forms of memory evaluated in this work, regardless of sex. This effect was displayed in a long-lasting manner (up to 17 days post-injection). LTP was inhibited at a postsynaptic level, both in males and females, and a long-term depression (LTD) was induced for the same prolonged period, which could underlie memory deficits. CONCLUSIONS In conclusion, our results provide further evidence on the shifting of LTP/LTD threshold due to a single icv. Aβ1-42 injection, which underly cognitive deficits in the early stages of AD. These long-lasting cognitive and functional alterations in males and females validate this model for the study of early amyloidosis in both sexes, thus offering a solid alternative to the inconsistence of amyloidosis transgenic mice models.
Collapse
Affiliation(s)
- Raquel Jiménez-Herrera
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Ana Contreras
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Souhail Djebari
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Jaime Mulero-Franco
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Guillermo Iborra-Lázaro
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Danko Jeremic
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Juan Navarro-López
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
| | - Lydia Jiménez-Díaz
- Neurophysiology and Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, University of Castilla-La Mancha, 13071, Ciudad Real, Spain.
| |
Collapse
|
38
|
Carvalho V, Colonna I, Curia G, Ferretti MT, Arabia G, Molnar MJ, Lebedeva ER, Moro E, de Visser M, Bui E. Sex steroid hormones and epilepsy: Effects of hormonal replacement therapy on seizure frequency of postmenopausal women with epilepsy-A systematic review. Eur J Neurol 2023; 30:2884-2898. [PMID: 37326003 DOI: 10.1111/ene.15916] [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: 02/15/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND PURPOSE Hormonal replacement therapy (HRT) is used for symptomatic treatment of menopause. Some evidence suggests a proconvulsant effect of estrogen and an anticonvulsant role of progesterone. Thus, the use of exogenous sex steroid hormones might influence the course of epilepsy in peri- and postmenopausal women with epilepsy (WWE). We conducted a systematic review on the impact of HRT on the frequency of seizures of WWE. METHODS PubMed and Scopus were searched for articles published from inception until August 2022. Abstracts from the past 5 years from the European Academy of Neurology and European Epilepsy Congresses were also reviewed. Article reference lists were screened, and relevant articles were retrieved for consultation. Interventional and observational studies on WWE and animal models of estrogen deficiency were included. Critical appraisal was performed using the revised Cochrane risk-of-bias tool for randomized trials and ROBINS-E tool. RESULTS Of 497 articles screened, 13 studies were included, including three human studies. One cross-sectional study showed a decrease in seizure frequency in WWE using combined HRT, a case-control study showed an increase in comparison with controls, and a randomized clinical trial found a dose-dependent increase in seizure frequency in women with focal epilepsy taking combined HRT. Ten studies addressing the impact of HRT in rat models were also included, which showed conflicting results. CONCLUSIONS There is scarce evidence of the impact of HRT in WWE. Further studies should evaluate the harmful potential, and prospective registries are needed for monitoring this population.
Collapse
Affiliation(s)
- Vanessa Carvalho
- Department of Neurosciences and Mental Health, Hospital de Santa Maria, Lisbon, Portugal
- Instituto de Medicina Molecular Professor João Lobo Antunes, Lisbon, Portugal
- Centro de Estudos Egas Moniz, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Isabella Colonna
- Complex Operative Unit of Neurology, "F. Ferrari" Hospital, Casarano, Italy
| | - Giulia Curia
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Gennarina Arabia
- Institute of Neurology, University Magna Graecia, Catanzaro, Italy
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Elena R Lebedeva
- Department of Neurology, Ural State Medical University, International Medical Center "Europe-Asia" Yekaterinburg, Yekaterinburg, Russia
| | - Elena Moro
- CHU Grenoble Alpes, Division of Neurology, Movement Disorders Unit, Grenoble Institute of Neurosciences, Grenoble Alpes University, Grenoble, France
| | - Marianne de Visser
- Department of Neurology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Esther Bui
- Division of Neurology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
39
|
Koebele SV, Bernaud VE, Northup-Smith SN, Willeman MN, Strouse IM, Bulen HL, Schrier AR, Newbern JM, DeNardo DF, Mayer LP, Dyer CA, Bimonte-Nelson HA. Gynecological surgery in adulthood imparts cognitive and brain changes in rats: A focus on hysterectomy at short-, moderate-, and long-term intervals after surgery. Horm Behav 2023; 155:105411. [PMID: 37659358 PMCID: PMC11060888 DOI: 10.1016/j.yhbeh.2023.105411] [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: 04/04/2023] [Revised: 07/11/2023] [Accepted: 08/08/2023] [Indexed: 09/04/2023]
Abstract
Premenopausal hysterectomy is associated with a greater relative risk of dementia. We previously demonstrated cognitive impairments in adult rats six weeks after hysterectomy with ovarian conservation compared with intact sham-controls and other gynecological surgery variations. Here, we investigated whether hysterectomy-induced cognitive impairments are transient or persistent. Adult rats received sham-control, ovariectomy (Ovx), hysterectomy, or Ovx-hysterectomy surgery. Spatial working memory, reference memory, and anxiety-like behavior were tested either six-weeks post-surgery, in adulthood; seven-months post-surgery, in early middle-age; or twelve-months post-surgery, in late middle-age. Hysterectomy in adulthood yielded spatial working memory deficits at short-, moderate-, and long-term post-surgery intervals. Serum hormone levels did not differ between ovary-intact, but differed from Ovx, groups. Hysterectomy had no significant impact on healthy ovarian follicle or corpora lutea counts for any post-surgery timepoint compared with intact sham-controls. Frontal cortex, dorsal hippocampus, and entorhinal cortex were assessed for activity-dependent markers. In entorhinal cortex, there were alterations in FOSB and ΔFOSB expression during the early middle-age timepoint, and phosphorylated ERK1/2 levels at the adult timepoint. Collectively, results suggest a primary role for the uterus in regulating cognition, and that memory-related neural pathways may be modified following gynecological surgery. This is the first preclinical report of long-term effects of hysterectomy with and without ovarian conservation on cognition, endocrine, ovarian, and brain assessments, initiating a comprehensive framework of gynecological surgery effects. Translationally, findings underscore critical needs to decipher how gynecological surgeries, especially those involving the uterus, impact the brain and its functions, the ovaries, and overall aging from a systems perspective.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Victoria E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Steven N Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Mari N Willeman
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Isabel M Strouse
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Haidyn L Bulen
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Ally R Schrier
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Dale F DeNardo
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | | | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA.
| |
Collapse
|
40
|
Lambi AG, Harris MY, Amin M, Joiner PG, Hilliard BA, Assari S, Popoff SN, Barbe MF. Blocking CCN2 Reduces Established Bone Loss Induced by Prolonged Intense Loading by Increasing Osteoblast Activity in Rats. JBMR Plus 2023; 7:e10783. [PMID: 37701153 PMCID: PMC10494513 DOI: 10.1002/jbm4.10783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 09/14/2023] Open
Abstract
We have an operant model of reaching and grasping in which detrimental bone remodeling is observed rather than beneficial adaptation when rats perform a high-repetition, high-force (HRHF) task long term. Here, adult female Sprague-Dawley rats performed an intense HRHF task for 18 weeks, which we have shown induces radial trabecular bone osteopenia. One cohort was euthanized at this point (to assay the bone changes post task; HRHF-Untreated). Two other cohorts were placed on 6 weeks of rest while being simultaneously treated with either an anti-CCN2 (FG-3019, 40 mg/kg body weight, ip; twice per week; HRHF-Rest/anti-CCN2), or a control IgG (HRHF-Rest/IgG), with the purpose of determining which might improve the trabecular bone decline. Results were compared with food-restricted control rats (FRC). MicroCT analysis of distal metaphysis of radii showed decreased trabecular bone volume fraction (BV/TV) and thickness in HRHF-Untreated rats compared with FRCs; responses improved with HRHF-Rest/anti-CCN2. Rest/IgG also improved trabecular thickness but not BV/TV. Histomorphometry showed that rest with either treatment improved osteoid volume and task-induced increases in osteoclasts. Only the HRHF-Rest/anti-CCN2 treatment improved osteoblast numbers, osteoid width, mineralization, and bone formation rate compared with HRHF-Untreated rats (as well as the latter three attributes compared with HRHF-Rest/IgG rats). Serum ELISA results were in support, showing increased osteocalcin and decreased CTX-1 in HRHF-Rest/anti-CCN2 rats compared with both HRHF-Untreated and HRHF-Rest/IgG rats. These results are highly encouraging for use of anti-CCN2 for therapeutic treatment of bone loss, such as that induced by chronic overuse. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alex G Lambi
- Department of Orthopedics and RehabilitationUniversity of New MexicoAlbuquerqueNMUSA
| | - Michele Y Harris
- Center for Translational Medicine, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Mamta Amin
- Center for Translational Medicine, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Patrice G Joiner
- Center for Translational Medicine, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Brendan A Hilliard
- Center for Translational Medicine, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | | | - Steven N Popoff
- Department of Biomedical Education and Data Science, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Mary F Barbe
- Center for Translational Medicine, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| |
Collapse
|
41
|
Soytürk H, Bozat BG, Pehlivan Karakas F, Coskun H, Firat T. Neuroprotective effects of goji berry (Lycium barbarum L.) polysaccharides on depression-like behavior in ovariectomized rats: behavioral and biochemical evidence. Croat Med J 2023; 64:231-242. [PMID: 37654035 PMCID: PMC10509687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/10/2023] [Indexed: 09/02/2023] Open
Abstract
AIM To assess the protective effects of goji berry (Lycium barbarum L.) polysaccharides (LBP) on depression-like behavior in ovariectomized rats and to elucidate the mechanisms underlying these effects. METHODS One hundred female Wistar albino rats (three months old) were randomly assigned either to ovariectomy (n=50) or sham surgery (n=50). After a 14-day recovery period, the groups were divided into five treatment subgroups (10 per group): high-dose LBP (200 mg/kg), low-dose LBP (20 mg/kg), imipramine (IMP, 2.5 mg/kg), 17-beta estradiol (E2, 1 mg/kg), and distilled water. Then, rats underwent a forced swimming test. We also determined the levels of serum antioxidant enzymes (superoxide dismutase, catalase, glutathione peroxidase, and malondialdehyde), E2 levels, hippocampal brain-derived neurotrophic factor (BDNF), 5HT2A receptor, and transferase dUTP nick end labeling (TUNEL)-positive cells. RESULTS Both low-dose LBP and imipramine decreased depression-like behavior by increasing serum superoxide dismutase activity and by decreasing serum malondialdehyde level. Furthermore, low-dose LPB, high-dose LBP, and imipramine increased the number of 5-HT2A receptor- and BDNF-positive cells but decreased the number of TUNEL-positive cells in the hippocampus. CONCLUSION This is the first study to show the antidepressant effect of LBP. Although additional research is needed, LBP may be considered a potential new antidepressant.
Collapse
Affiliation(s)
| | | | - Fatma Pehlivan Karakas
- Fatma Pehlivan Karakas, Bolu Abant Izzet Baysal University, Department of Biology, 14280 Bolu, Turkey,
| | | | | |
Collapse
|
42
|
McDonald R, Larsen M, Liu Z, Southekal S, Eudy J, Guda C, Kumar TR. RNA-seq analysis identifies age-dependent changes in expression of mRNAs - encoding N-glycosylation pathway enzymes in mouse gonadotropes. Mol Cell Endocrinol 2023; 574:111971. [PMID: 37301504 PMCID: PMC10528389 DOI: 10.1016/j.mce.2023.111971] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/01/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
Follicle-stimulating hormone (FSH) is a glycoprotein that is assembled as a heterodimer of α/β subunits in gonadotropes. Each subunit contains two N-glycan chains. Our previous in vivo genetic studies identified that at least one N-glycan chain must be present on the FSHβ subunit for efficient FSH dimer assembly and secretion. Moreover, macroheterogeneity observed uniquely on human FSHβ results in ratiometric changes in age-specific FSH glycoforms, particularly during menopausal transition. Despite the recognition of many prominent roles of sugars on FSH including dimer assembly and secretion, serum half-life, receptor binding and signal transduction, the N-glycosylation machinery in gonadotropes has never been defined. Here, we used a mouse model in which gonadotropes are GFP-labeled in vivo and achieved rapid purification of GFP+ gonadotropes from pituitaries of female mice at reproductively young, middle, and old ages. We identified by RNA-seq analysis 52 mRNAs encoding N-glycosylation pathway enzymes expressed in 3- and 8-10-month-old mouse gonadotropes. We hierarchically mapped and localized the enzymes to distinct subcellular organelles within the N-glycosylation biosynthetic pathway. Of the 52 mRNAs, we found 27 mRNAs are differentially expressed between the 3- and 8-10-month old mice. We subsequently selected 8 mRNAs which showed varying changes in expression for confirmation of abundance in vivo via qPCR analysis, using more expanded aging time points with distinct 8-month and 14-month age groups. Real time qPCR analysis indicated dynamic changes in expression of N-glycosylation pathway enzyme-encoding mRNAs across the life span. Notably, computational analysis predicted the promoters of genes encoding these 8 mRNAs contain multiple high probability binding sites for estrogen receptor-1 and progesterone receptor. Collectively, our studies define the N-glycome and identify age-specific dynamic changes in mRNAs encoding N-glycosylation pathway enzymes in mouse gonadotropes. Our studies suggest the age-related decline in ovarian steroids may regulate expression of N-glycosylation enzymes in mouse gonadotropes and explain the age-related N-glycosylation shift previously observed on human FSHβ subunit in pituitaries of women.
Collapse
Affiliation(s)
- Rosemary McDonald
- Garduate Program in Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mark Larsen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Zhenghui Liu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Siddesh Southekal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - James Eudy
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - T Rajendra Kumar
- Garduate Program in Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
43
|
Lee HJ, Park MJ, Heo JD, Joo BS, Joo JK. Timing of hormone therapy and its association with cardiovascular risk and metabolic parameters in 4-vinylcyclohexene diepoxide-induced primary ovarian insufficiency mouse model. Gynecol Endocrinol 2023; 39:2247094. [PMID: 37599578 DOI: 10.1080/09513590.2023.2247094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/14/2022] [Accepted: 08/07/2023] [Indexed: 08/22/2023] Open
Abstract
OBJECTIVE To evaluate the effects of various initiation time points and durations of hormone therapy (HT) on cardiovascular and metabolic parameters of premenarche, primary ovarian insufficiency (POI) mouse model, induced by 4-vinylcyclohexene diepoxide. METHODS A total of 50 mice at 4 weeks of age were developed into POI mouse model, further randomly categorized into 5 groups: control group without any intervention; no HT group with only high-fat diet (NT); group 1 with delayed estradiol treatment (T1); group 2 with on-time, continuous estradiol treatment (T2); and group 3 with on-time estradiol treatment but early stop (T3). Cardiovascular risk and metabolic parameters were measured. RESULTS Presenting with similar body weights, blood glucose levels of T1, T2, and T3 were all significantly lower than NT (p < .001). Serum total cholesterol and insulin were also significantly lower in all HT groups than in NT, especially in T2 (p < .001). For serum low-density lipoprotein-cholesterol, only T2 resulted in the statically lower level than those of NT, T1, and T3 (p < .001). Aortic thickness was significantly increased with aggravated fibrotic change of the intima in NT, and such consequence was significantly ameliorated in HT groups, mostly lowered in T2 (p < .05). Last, serum pro-inflammatory cytokines were significantly low in the HT groups than in NT, especially in T2 with the lowest level (p < .05). . CONCLUSIONS On-time, continuous E2 treatment immediately after a biologic estrogen deprivation event significantly reduced metabolic and cardiovascular risks in young, pre-menarche female mouse models of POI, confirming decreased serum levels of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Hyun Joo Lee
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Min Jung Park
- The Korea Institute for Public Sperm Bank, Busan, Republic of Korea
| | - Jeong-Doo Heo
- Korea Institute of Toxicology, Gyeongnam Branch Institute, Jinju, Gyeongsangnam-do, Republic of Korea
| | - Bo Sun Joo
- The Korea Institute for Public Sperm Bank, Busan, Republic of Korea
| | - Jong Kil Joo
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
44
|
Cavalcante MB, Sampaio OGM, Câmara FEA, Schneider A, de Ávila BM, Prosczek J, Masternak MM, Campos AR. Ovarian aging in humans: potential strategies for extending reproductive lifespan. GeroScience 2023; 45:2121-2133. [PMID: 36913129 PMCID: PMC10651588 DOI: 10.1007/s11357-023-00768-8] [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: 02/23/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
Ovarian reserve is a term used to estimate the total number of immature follicles present in the ovaries. Between birth and menopause, there is a progressive decrease in the number of ovarian follicles. Ovarian aging is a continuous physiological phenomenon, with menopause being the clinical mark of the end of ovarian function. Genetics, measured as family history for age at the onset of menopause, is the main determinant. However, physical activity, diet, and lifestyle are important factors that can influence the age of menopause. The low estrogen levels after natural or premature menopause increased the risk for several diseases, resulting in increased mortality risk. Besides that, the decreasing ovarian reserve is associated to reduced fertility. In women with infertility undergoing in vitro fertilization, reduced markers of ovarian reserve, including antral follicular count and anti-Mullerian hormone, are the main indicators of reduced chances of becoming pregnant. Therefore, it becomes clear that the ovarian reserve has a central role in women's life, affecting fertility early in life and overall health later in life. Based on this, the ideal strategy for delaying ovarian aging should have the following characteristics: (1) be initiated in the presence of good ovarian reserve; (2) maintained for a long period; (3) have an action on the dynamics of primordial follicles, controlling the rate of activation and atresia; and (4) safe use in pre-conception, pregnancy, and lactation. In this review, we therefore discuss some of these strategies and its feasibility for preventing a decline in the ovarian reserve.
Collapse
Affiliation(s)
- Marcelo Borges Cavalcante
- Postgraduate Program in Medical Sciences, University of Fortaleza (UNIFOR), Fortaleza, CE, 60.811-905, Brazil.
| | - Olga Goiana Martins Sampaio
- Postgraduate Program in Medical Sciences, University of Fortaleza (UNIFOR), Fortaleza, CE, 60.811-905, Brazil
| | | | - Augusto Schneider
- Nutrition College, Federal University of Pelotas (UFPel), Pelotas, RS, 96010-610, Brazil
| | | | - Juliane Prosczek
- Nutrition College, Federal University of Pelotas (UFPel), Pelotas, RS, 96010-610, Brazil
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando FL, USA
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Adriana Rolim Campos
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
45
|
Borodzicz-Jażdżyk S, Kołodzińska A, Czarzasta K, Wojciechowska M, Główczyńska R, Szczepankiewicz B, Puchalska L, Opolski G, Cudnoch-Jędrzejewska A. Inflammatory Forms of Cardiomyocyte Cell Death in the Rat Model of Isoprenaline-Induced Takotsubo Syndrome. Biomedicines 2023; 11:2060. [PMID: 37509699 PMCID: PMC10377582 DOI: 10.3390/biomedicines11072060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Takotsubo syndrome (TTS) is associated with inflammatory response, therefore the aim of the study was to evaluate the presence and dynamics of inflammatory-associated forms of cell death, necroptosis, and pyroptosis in the female rat model of isoprenaline (ISO)-induced TTS. TTS was induced in female Sprague Dawley rats (n = 36) by ISO 150 mg/kg intraperitoneally. Animals were divided into four groups: TTSO (TTS+ovariectomy; n = 10), TTSP (TTS+sham operation; n = 10), CO (0.9% NaCl+ovariectomy; n = 8), CP (0.9% NaCl+sham operation; n = 8). Histopathological analysis, evaluation of plasma concentration, and myocardial expression of pyroptosis- and necroptosis-associated proteins were performed. TTSO and TTSP groups had higher plasma concentrations of interleukin-1β in comparison with the controls. Low myocardial protein expression of mixed lineage kinase domain-like pseudokinase (MLKL), caspase-1 (Casp-1), and calcium/calmodulin-dependent kinase type II isoform delta (CAMKIIδ) was visible 6 and/or 12 h post-ISO. Twenty-four hours post-ISO, high myocardial and vascular protein expression of CAMKIIδ was visible in TTSO but not TTSP rats, while high myocardial expression of MLKL and Casp-1 was visible both in TTSO and TTSP rats. The course of TTS is associated with activation of inflammatory-associated programmed cell death, necroptosis, and pyroptosis, therefore inflammation may be a primary response occurring simultaneously with cardiomyocyte death in TTS.
Collapse
Affiliation(s)
- Sonia Borodzicz-Jażdżyk
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Agnieszka Kołodzińska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Katarzyna Czarzasta
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Małgorzata Wojciechowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Renata Główczyńska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Benedykt Szczepankiewicz
- Department of Pathology, Medical University of Warsaw, 7 Pawińskiego Street, 02-106 Warsaw, Poland
| | - Liana Puchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Grzegorz Opolski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| |
Collapse
|
46
|
Bernaud VE, Koebele SV, Northup-Smith SN, Willeman MN, Barker C, Schatzki-Lumpkin A, Sanchez MV, Bimonte-Nelson HA. Evaluations of memory, anxiety, and the growth factor IGF-1R after post-surgical menopause treatment with a highly selective progestin. Behav Brain Res 2023; 448:114442. [PMID: 37085118 PMCID: PMC11105077 DOI: 10.1016/j.bbr.2023.114442] [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: 08/18/2022] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
Progestogens are a key component of menopausal hormone therapies. While some progestogens can be detrimental to cognition, there is preclinical evidence that progestogens with a strong progesterone-receptor affinity benefit some molecular mechanisms believed to underlie cognitive function. Thus, a progestin that maximizes progesterone-receptor affinity and minimizes affinities to other receptors may be cognitively beneficial. We evaluated segesterone-acetate (SGA), a 19-norprogesterone derivative with a strong progesterone-receptor affinity and no androgenic or estrogenic-receptor activity, hypothesizing that it would enhance cognition. Middle-aged rats underwent Sham or Ovariectomy (Ovx) surgery followed by administration of medroxyprogesterone-acetate (MPA; used as a positive control as we have previously shown MPA-induced cognitive deficits), SGA (low or high dose), or vehicle (one Sham and one Ovx group). Spatial working and reference memory, delayed retention, and anxiety-like behavior were assessed, as were memory- and hormone- related protein assays within the frontal cortex, dorsal hippocampus, and entorhinal cortex. Low-dose SGA impaired spatial working memory, while high-dose SGA had a more extensive detrimental impact, negatively affecting spatial reference memory and delayed retention. Replicating previous findings, MPA impaired spatial reference memory and delayed retention. SGA, but not MPA, alleviated Ovx-induced anxiety-like behaviors. On two working memory measures, IGF-1R expression correlated with better working memory only in rats without hormone manipulation; any hormone manipulation or combination of hormone manipulations used herein altered this relationship. These findings suggest that SGA impairs spatial cognition after surgical menopause, and that surgical menopause with or without progestin administration disrupts relationships between a growth factor critical to neuroplasticity.
Collapse
Affiliation(s)
- Victoria E Bernaud
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Stephanie V Koebele
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Steven N Northup-Smith
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Mari N Willeman
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA; TGen Institute, 445 N 5th St, Phoenix, AZ 85004, USA
| | - Charlotte Barker
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Alex Schatzki-Lumpkin
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Maria Valenzuela Sanchez
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, 950 S. McAllister Ave., Tempe, AZ 85287, USA; Arizona Alzheimer's Consortium, 4745 N 7th St, Phoenix, AZ 85014, USA.
| |
Collapse
|
47
|
Zaki ES, Sayed RH, Saad MA, El-Yamany MF. Roflumilast ameliorates ovariectomy-induced depressive-like behavior in rats via activation of AMPK/mTOR/ULK1-dependent autophagy pathway. Life Sci 2023:121806. [PMID: 37257579 DOI: 10.1016/j.lfs.2023.121806] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/14/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
AIMS Roflumilast, a well-known phosphodiesterase-4 (PDE-4) inhibitor, possess an anti-inflammatory activity with approved indications in chronic obstructive pulmonary disease. This study aimed to evaluate the neuroprotective role of roflumilast in ovariectomy (OVX)-induced depressive-like behavior in female rats and to shed light on a potential autophagy enhancing effect. MAIN METHODS Rats were randomly divided into four groups: sham, OVX, OVX + roflumilast (1 mg/kg, p.o), and OVX + roflumilast + chloroquine (CQ) (50 mg/kg, i.p). Drugs were administered for 4 weeks starting 2 weeks after OVX. KEY FINDINGS Roflumilast improved the depressive-like behaviors observed in OVX rats as evidenced by decreasing both forced swimming and open field immobility times while, increasing % sucrose preference and number of open field crossed squares. Histopathological analysis provides further evidence of roflumilast's beneficial effects, demonstrating that roflumilast ameliorated the neuronal damage caused by OVX. Roflumilast antidepressant potential was mediated via restoring hippocampal cAMP and BDNF levels as well as down-regulating PDE4 expression. Moreover, roflumilast revealed anti-inflammatory and anti-apoptotic effects via hindering TNF-α level and diminishing Bax/Bcl2 ratio. Roflumilast restored the autophagic function via up-regulation of p-AMPK, p-ULK1, Beclin-1 and LC3II/I expression, along with downregulation of P62 level and p-mTOR protein expression. The autophagy inhibitor CQ was used to demonstrate the suggested pathway. SIGNIFICANCE The present study revealed that roflumilast showed an anti-depressant activity in OVX female rats via turning on AMPK/mTOR/ULK1-dependent autophagy pathway; and neurotrophic, anti-inflammatory, and anti-apoptotic activities. Roflumilast could offer a more secure alternative to hormone replacement therapy for postmenopausal depression treatment.
Collapse
Affiliation(s)
- Eman S Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Muhammed A Saad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, 4184, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Muhammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
48
|
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: 10.0] [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
|
49
|
Jasinskyte U, Buisas R, Griskova-Bulanova I, Guzulaitis R. Auditory steady-state responses in the auditory cortex of mice during estrus cycle. Brain Res 2023; 1810:148376. [PMID: 37121427 DOI: 10.1016/j.brainres.2023.148376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
Auditory-steady state responses (ASSRs) disclose brain's potency to oscillate and have been suggested to serve as biomarkers in various psychiatric disorders. GABAergic neurotransmission, a critical component for brain oscillations, is heavily influenced by sex hormones. In line, the severity of symptoms in psychiatric disorders is linked to changes in sex hormones during menstrual cycle. However, how these sex hormones affect ASSRs remain largely unknown. This was addressed by performing chronic recordings of ASSRs in mice while monitoring its estrus cycle. Here, the stability of ASSRs during long term recordings were validated and showed good reliability. 40 Hz ASSRs showed changes throughout estrus cycle where it decreased in metestrus phase compared to diestrus phase. In contrast, other frequency ASSRs did not show significant changes throughout estrus cycle. Taken together, our findings illustrate that the estrus cycle can influence the generation of ASSRs and the phase of the estrus cycle should be taken into consideration when ASSRs are recorded in females.
Collapse
Affiliation(s)
- Urte Jasinskyte
- Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Rokas Buisas
- Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | | | | |
Collapse
|
50
|
Muscat SM, Butler MJ, Mackey-Alfonso SE, Barrientos RM. Young adult and aged female rats are vulnerable to amygdala-dependent, but not hippocampus-dependent, memory impairment following short-term high-fat diet. Brain Res Bull 2023; 195:145-156. [PMID: 36870621 PMCID: PMC10257807 DOI: 10.1016/j.brainresbull.2023.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
Global populations are increasingly consuming diets high in saturated fats and refined carbohydrates, and such diets have been well-associated with heightened inflammation and neurological dysfunction. Notably, older individuals are particularly vulnerable to the impact of unhealthy diet on cognition, even after a single meal, and pre-clinical rodent studies have demonstrated that short-term consumption of high-fat diet (HFD) induces marked increases in neuroinflammation and cognitive impairment. Unfortunately though, to date, most studies on the topic of nutrition and cognition, especially in aging, have been performed only in male rodents. This is especially concerning given that older females are more vulnerable to develop certain memory deficits and/or severe memory-related pathologies than males. Thus, the aim of the present study was to determine the extent to which short-term HFD consumption impacts memory function and neuroinflammation in female rats. Young adult (3 months) and aged (20-22 months) female rats were fed HFD for 3 days. Using contextual fear conditioning, we found that HFD had no effect on long-term contextual memory (hippocampus-dependent) at either age, but impaired long-term auditory-cued memory (amygdala-dependent) regardless of age. Gene expression of Il-1β was markedly dysregulated in the amygdala, but not hippocampus, of both young and aged rats after 3 days of HFD. Interestingly, modulation of IL-1 signaling via central administration of the IL-1 receptor antagonist (which we have previously demonstrated to be protective in males) had no impact on memory function following the HFD in females. Investigation of the memory-associated gene Pacap and its receptor Pac1r revealed differential effects of HFD on their expression in the hippocampus and amygdala. Specifically, HFD induced increased expression of Pacap and Pac1r in the hippocampus, whereas decreased Pacap was observed in the amygdala. Collectively, these data suggest that both young adult and aged female rats are vulnerable to amygdala-dependent (but not hippocampus-dependent) memory impairments following short-term HFD consumption, and identify potential mechanisms related to IL-1β and PACAP signaling in these differential effects. Notably, these findings are strikingly different than those previously reported in male rats using the same diet regimen and behavioral paradigms, and highlight the importance of examining potential sex differences in the context of neuroimmune-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Sabrina E Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|