1
|
Methawasin M, Strom J, Marino VA, Gohlke J, Muldoon J, Herrick SR, van der Piji R, Konhilas JP, Granzier H. An ovary-intact postmenopausal HFpEF mouse model; menopause is more than just estrogen deficiency. Am J Physiol Heart Circ Physiol 2025; 328:H719-H733. [PMID: 39963865 DOI: 10.1152/ajpheart.00575.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/06/2024] [Accepted: 01/10/2025] [Indexed: 03/11/2025]
Abstract
The incidence of heart failure with preserved ejection fraction (HFpEF) in women significantly increases following menopause. This trend cannot solely be attributed to chronological aging, as evidenced by the more gradual increase in prevalence among men, suggesting that menopause is a provocative event for HFpEF. However, the underlying mechanisms remain elusive and challenging to investigate in human subjects; moreover, an attempt to create HFpEF in ovariectomized (OVX) mice was unsuccessful. In this study, we created an animal model that resembles HFpEF in women undergoing natural menopause. We used 4-vinylcyclohexene dioxide (VCD) to induce "ovary-intact" menopause, combined with the 2hit regimen (HFpEF inducing regimen) to model postmenopausal HFpEF. The female-VCD-2hit mice demonstrate diastolic dysfunction. At the left ventricle (LV) levels, the increased stiffness coefficient of end-diastolic pressure-volume relation (EDPVR), elevated LV end-diastolic pressure, and increased relaxation time constant indicate a heightened LV stiffness, delayed relaxation, and elevated LV filling pressure. At the cardiomyocyte level, the female-VCD-2hit mice exhibit increased cellular diastolic stiffness and delayed relaxation, suggesting that the observed LV dysfunction is derived from the cardiomyocytes. In addition, plasma N-terminal pro-β-type natriuretic peptide (NT-pro-BNP) levels were elevated, whereas Xbp1s transcript levels were reduced, further supporting the existence of HFpEF. Plasma-free testosterone was increased in VCD mice compared with premenopausal and OVX models. Further studies are required to determine whether the relative increase in testosterone is the factor driving HFpEF susceptibility in VCD mice. Ovary-intact postmenopausal status makes female mice vulnerable to HFpEF development. The VCD-2hit model develops a robust HFpEF-like phenotype and is suitable for studying female HFpEF.NEW & NOTEWORTHY Although ovariectomized mice were observed to be resistant to developing HFpEF, ovary-intact postmenopausal mice exhibited an HFpEF-like phenotype under metabolic stress conditions. The increased susceptibility of ovary-intact postmenopausal mice may be due to relative androgen excess conditions, as postmenopausal ovaries retain the ability to secrete androgens. Menopause should be viewed as the imbalance of estrogen and androgens rather than merely an estrogen deficiency, and the role of female androgens in postmenopausal HFpEF warrants further investigation.
Collapse
Affiliation(s)
- Mei Methawasin
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Joshua Strom
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Vito A Marino
- Department of Physiology and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Julia Muldoon
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Shelby R Herrick
- Department of Physiology and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Robbert van der Piji
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - John P Konhilas
- Department of Physiology and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
2
|
Pyle WG. Tackling change: menopause as a cardiovascular disease risk factor and the path to health equity. Cardiovasc Res 2024; 120:e63-e64. [PMID: 39495205 PMCID: PMC11646100 DOI: 10.1093/cvr/cvae232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/15/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Affiliation(s)
- W Glen Pyle
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Dalhousie University, 100 Tucker Park Road, Saint John, New Brunswick, Canada E2K 5E2
- Women’s Health Research Institute, B.C. Women’s Hospital Health Centre, H214 4500 Oak Street, Box 42B, Vancouver, British Columbia, Canada V6H 3N1
| |
Collapse
|
3
|
Gerges SH, El-Kadi AOS. Changes in cardiovascular arachidonic acid metabolism in experimental models of menopause and implications on postmenopausal cardiac hypertrophy. Prostaglandins Other Lipid Mediat 2024; 173:106851. [PMID: 38740361 DOI: 10.1016/j.prostaglandins.2024.106851] [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: 12/12/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Menopause is a normal stage in the human female aging process characterized by the cessation of menstruation and the ovarian production of estrogen and progesterone hormones. Menopause is associated with an increased risk of several different diseases. Cardiovascular diseases are generally less common in females than in age-matched males. However, this female advantage is lost after menopause. Cardiac hypertrophy is a disease characterized by increased cardiac size that develops as a response to chronic overload or stress. Similar to other cardiovascular diseases, the risk of cardiac hypertrophy significantly increases after menopause. However, the exact underlying mechanisms are not yet fully elucidated. Several studies have shown that surgical or chemical induction of menopause in experimental animals is associated with cardiac hypertrophy, or aggravates cardiac hypertrophy induced by other stressors. Arachidonic acid (AA) released from the myocardial phospholipids is metabolized by cardiac cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) enzymes to produce several eicosanoids. AA-metabolizing enzymes and their respective metabolites play an important role in the pathogenesis of cardiac hypertrophy. Menopause is associated with changes in the cardiovascular levels of CYP, COX, and LOX enzymes and the levels of their metabolites. It is possible that these changes might play a role in the increased risk of cardiac hypertrophy after menopause.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
4
|
Barry C, Rouhana S, Braun JL, Geromella MS, Fajardo VA, Pyle WG. Perimenopause Decreases SERCA2a Activity in the Hearts of a Mouse Model of Ovarian Failure. Biomolecules 2024; 14:675. [PMID: 38927078 PMCID: PMC11201532 DOI: 10.3390/biom14060675] [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: 05/12/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Risk of cardiovascular disease mortality rises in women after menopause. While increased cardiovascular risk is largely attributed to postmenopausal declines in estrogens, the molecular changes in the heart that contribute to risk are poorly understood. Disruptions in intracellular calcium handling develop in ovariectomized mice and have been implicated in cardiac dysfunction. Using a mouse model of menopause in which ovarian failure occurs over 120 days, we sought to determine if perimenopause impacted calcium removal mechanisms in the heart and identify the molecular mechanisms. Mice were injected with 4-vinylcyclohexene diepoxide (VCD) to induce ovarian failure over 120 days, mimicking perimenopause. Hearts were removed at 60 and 120 days after VCD injections, representing the middle and end of perimenopause. SERCA2a function was significantly diminished at the end of perimenopause. Neither SERCA2a nor phospholamban expression changed at either time point, but phospholamban phosphorylation at S16 and T17 was dynamically altered. Intrinsic SERCA inhibitors sarcolipin and myoregulin increased >4-fold at day 60, as did the native activator DWORF. At the end of perimenopause, sarcolipin and myoregulin returned to baseline levels while DWORF was significantly reduced below controls. Sodium-calcium exchanger expression was significantly increased at the end of perimenopause. These results show that the foundation for increased cardiovascular disease mortality develops in the heart during perimenopause and that regulators of calcium handling exhibit significant fluctuations over time. Understanding the temporal development of cardiovascular risk associated with menopause and the underlying mechanisms is critical to developing interventions that mitigate the rise in cardiovascular mortality that arises after menopause.
Collapse
Affiliation(s)
- Ciara Barry
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
| | - Sarah Rouhana
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
| | - Jessica L. Braun
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Mia S. Geromella
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Val A. Fajardo
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - W. Glen Pyle
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
- Women’s Health Research Institute at BC Women’s Hospital + Health Centre, Vancouver, BC V6H 2N9, Canada
| |
Collapse
|
5
|
Sun H, Qi Q, Pan X, Zhou J, Wang J, Li L, Li D, Wang L. Bu-Shen-Ning-Xin decoction inhibits macrophage activation to ameliorate premature ovarian insufficiency-related osteoimmune disorder via FSH/FSHR pathway. Drug Discov Ther 2024; 18:106-116. [PMID: 38631868 DOI: 10.5582/ddt.2024.01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Limited studies are associated with premature ovarian insufficiency (POI)-related osteoimmune disorder currently. Bu-Shen-Ning-Xin decoction (BSNXD) displayed a favorable role in treating postmenopausal osteoporosis. However, its impact on the POI-related osteoimmune disorder remains unclear. The study primarily utilized animal experiments and network pharmacology to investigate the effects and underlying mechanisms of BSNXD on the POI-related osteoimmune disorder. First, a 4-vinylcyclohexene dioxide (VCD)-induced POI murine model was conducted to explore the therapeutical action of BSNXD. Second, we analyzed the active compounds of BSNXD and predicted their potential mechanisms for POI-related osteoimmune disorder via network pharmacology, further confirmed by molecular biology experiments. The results demonstrated that VCD exposure led to elevated follicle-stimulating hormone (FSH) levels, a 50% reduction in the primordial follicles, bone microstructure changes, and macrophage activation, indicating an osteoimmune disorder. BSNXD inhibited macrophage activation and osteoclast differentiation but did not affect serum FSH and estradiol levels in the VCD-induced POI model. Network pharmacology predicted the potential mechanisms of BSNXD against the POI-related osteoimmune disorder involving tumor necrosis factor α and MAPK signaling pathways, highlighting BSNXD regulated inflammation, hormone, and osteoclast differentiation. Further experiments identified BSNXD treatment suppressed macrophage activation via downregulating FSH receptor (FSHR) expression and inhibiting the phosphorylation of ERK and CCAAT enhancer binding proteins β. In conclusion, BSNXD regulated POI-related osteoimmune disorder by suppressing the FSH/FSHR pathway to reduce macrophage activation and further inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Hongmei Sun
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
- Hexi University, Zhangye, Gansu, China
| | - Qing Qi
- Wuhan Business University, Wuhan, Hubei, China
| | - Xinyao Pan
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Lisha Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Dajing Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
6
|
Troy AM, Normukhamedova D, Grothe D, Momen A, Zhou YQ, McFadden M, Hussain M, Billia F, Cheng HLM. Impact of ovary-intact menopause in a mouse model of heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2024; 326:H522-H537. [PMID: 38180450 PMCID: PMC11221814 DOI: 10.1152/ajpheart.00733.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) afflicts over half of all patients with heart failure and is a debilitating and fatal syndrome affecting postmenopausal women more than any other demographic. This bias toward older females calls into question the significance of menopause in the development of HFpEF, but this question has not been probed in detail. In this study, we report the first investigation into the impact of ovary-intact menopause in the context of HFpEF. To replicate the human condition as faithfully as possible, vinylcyclohexene dioxide (VCD) was used to accelerate ovarian failure (AOF) in female mice while leaving the ovaries intact. HFpEF was established with a mouse model that involves two stressors typical in humans: a high-fat diet and hypertension induced from the nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester (l-NAME). In young female mice, AOF or HFpEF-associated stressors independently induced abnormal myocardial strain indicative of early subclinical systolic and diastolic cardiac dysfunction. HFpEF but not AOF was associated with elevations in systolic blood pressure. Increased myocyte size and reduced myocardial microvascular density were not observed in any group. Also, a broad panel of measurements that included echocardiography, invasive pressure measurements, histology, and serum hormones revealed no interaction between AOF and HFpEF. Interestingly, AOF did evoke a higher density of infiltrating cardiac immune cells in both healthy and HFpEF mice, suggestive of proinflammatory effects. In contrast to young mice, middle-aged "old" mice did not exhibit cardiac dysfunction from estrogen deprivation alone or from HFpEF-related stressors.NEW & NOTEWORTHY This is the first preclinical study to examine the impact of ovary-intact menopause [accelerated ovarian failure (AOF)] on HFpEF. Echocardiography of young female mice revealed early evidence of diastolic and systolic cardiac dysfunction apparent only on strain imaging in HFpEF only, AOF only, or the combination. Surprisingly, AOF did not exacerbate the HFpEF phenotype. Results in middle-aged "old" females also showed no interaction between HFpEF and AOF and, importantly, no cardiovascular impact from HFpEF or AOF.
Collapse
Affiliation(s)
- Aaron M Troy
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | - Diyora Normukhamedova
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | - Daniela Grothe
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Abdul Momen
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yu-Qing Zhou
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | - Meghan McFadden
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | - Mansoor Hussain
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Filio Billia
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Hai-Ling Margaret Cheng
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Huang F, Cao Y, Liang J, Tang R, Wu S, Zhang P, Chen R. The influence of the gut microbiome on ovarian aging. Gut Microbes 2024; 16:2295394. [PMID: 38170622 PMCID: PMC10766396 DOI: 10.1080/19490976.2023.2295394] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Ovarian aging occurs prior to the aging of other organ systems and acts as the pacemaker of the aging process of multiple organs. As life expectancy has increased, preventing ovarian aging has become an essential goal for promoting extended reproductive function and improving bone and genitourinary conditions related to ovarian aging in women. An improved understanding of ovarian aging may ultimately provide tools for the prediction and mitigation of this process. Recent studies have suggested a connection between ovarian aging and the gut microbiota, and alterations in the composition and functional profile of the gut microbiota have profound consequences on ovarian function. The interaction between the gut microbiota and the ovaries is bidirectional. In this review, we examine current knowledge on ovary-gut microbiota crosstalk and further discuss the potential role of gut microbiota in anti-aging interventions. Microbiota-based manipulation is an appealing approach that may offer new therapeutic strategies to delay or reverse ovarian aging.
Collapse
Affiliation(s)
- Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Ying Cao
- School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Jinghui Liang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Ruiyi Tang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Si Wu
- School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Rare Disease Center, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| |
Collapse
|
8
|
Medzikovic L, Azem T, Sun W, Rejali P, Esdin L, Rahman S, Dehghanitafti A, Aryan L, Eghbali M. Sex Differences in Therapies against Myocardial Ischemia-Reperfusion Injury: From Basic Science to Clinical Perspectives. Cells 2023; 12:2077. [PMID: 37626887 PMCID: PMC10453147 DOI: 10.3390/cells12162077] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Mortality from myocardial infarction (MI) has declined over recent decades, which could be attributed in large part to improved treatment methods. Early reperfusion is the cornerstone of current MI treatment. However, reoxygenation via restored blood flow induces further damage to the myocardium, leading to ischemia-reperfusion injury (IRI). While experimental studies overwhelmingly demonstrate that females experience greater functional recovery from MI and decreased severity in the underlying pathophysiological mechanisms, the outcomes of MI with subsequent reperfusion therapy, which is the clinical correlate of myocardial IRI, are generally poorer for women compared with men. Distressingly, women are also reported to benefit less from current guideline-based therapies compared with men. These seemingly contradicting outcomes between experimental and clinical studies show a need for further investigation of sex-based differences in disease pathophysiology, treatment response, and a sex-specific approach in the development of novel therapeutic methods against myocardial IRI. In this literature review, we summarize the current knowledge on sex differences in the underlying pathophysiological mechanisms of myocardial IRI, including the roles of sex hormones and sex chromosomes. Furthermore, we address sex differences in pharmacokinetics, pharmacodynamics, and pharmacogenetics of current drugs prescribed to limit myocardial IRI. Lastly, we highlight ongoing clinical trials assessing novel pharmacological treatments against myocardial IRI and sex differences that may underlie the efficacy of these new therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mansoureh Eghbali
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Ave, CHS BH-550 CHS, Los Angeles, CA 90095, USA (W.S.)
| |
Collapse
|
9
|
Yu S, Zhang L, Wang Y, Yan J, Wang Q, Bian H, Huang L. Mood, hormone levels, metabolic and sleep across the menopausal transition in VCD-induced ICR mice. Physiol Behav 2023; 265:114178. [PMID: 37001841 DOI: 10.1016/j.physbeh.2023.114178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/27/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
AIMS Menopausal transition is the transitional period before menopause in women, often accompanied by abnormal fluctuations in hormone levels that increase the risk of aging-related diseases. 4-vinylcyclohexene dioxide (VCD) is a chemical agent that induces gradual depletion of ovarian follicles, which can mimic the natural human process of transition from menopausal transition to post-menopause. Previous studies have shown that the onset of menopausal transition or menopause in VCD-injected mice is associated with a specific strain, even in inbred animals. Institute of Cancer Research (ICR) mice constitute general purpose outbred population, which has not been well-characterized in the VCD-induced model. Thus, the current study aimed to explore the characteristic features, including sleep, mood, and metabolism, of the model by examining the effect of timing of VCD injection in ICR mice to extend the applications of this model. MATERIALS AND METHODS ICR mice were randomly divided into six groups: 20d VCD and 20d Control, 35d VCD and 35d Control, 52d VCD and 52d Control. VCD mice were intraperitoneally injected with VCD (160 mg/kg), while Control mice were injected intraperitoneally with sesame oil for 4 consecutive weeks, five times a week daily. A vaginal smear was used to observe the estrous cycle of the mice. On the 20th, 35th, and 52nd day after VCD or sesame oil injection, the ovarian morphology, the number of atretic cells, hormone levels, anxiety, depression-like behaviors, sleep phase, and energy metabolism were observed. KEY FINDINGS The menopausal transition model was successfully replicated by injecting VCD into ICR mice. On the specific days after VCD treatment, the number of atretic follicles increased, the level of E2 decreased and FSH increased, the depressive- and anxiety-like behavior increased, the time of REM and NREM sleep time decreased, and energy metabolism was reduced. SIGNIFICANCE These results suggested that the ICR mice model has human-like characteristics during the menopause transition. Moreover, the ICR model has a long menopausal transition duration.
Collapse
Affiliation(s)
- Shuang Yu
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Lixin Zhang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Yanyan Wang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Jinming Yan
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Qi Wang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Hongsheng Bian
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China
| | - Lili Huang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Harbin 150040, China.
| |
Collapse
|
10
|
Sui K, Yasrebi A, Longoria CR, MacDonell AT, Jaffri ZH, Martinez SA, Fisher SE, Malonza N, Jung K, Tveter KM, Wiersielis KR, Uzumcu M, Shapses SA, Campbell SC, Roepke TA, Roopchand DE. Coconut Oil Saturated Fatty Acids Improved Energy Homeostasis but not Blood Pressure or Cognition in VCD-Treated Female Mice. Endocrinology 2023; 164:bqad001. [PMID: 36626144 PMCID: PMC11009791 DOI: 10.1210/endocr/bqad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
Obesity, cardiometabolic disease, cognitive decline, and osteoporosis are symptoms of postmenopause, which can be modeled using 4-vinylcyclohexene diepoxide (VCD)-treated mice to induce ovarian failure and estrogen deficiency combined with high-fat diet (HFD) feeding. The trend of replacing saturated fatty acids (SFAs), for example coconut oil, with seed oils that are high in polyunsaturated fatty acids, specifically linoleic acid (LA), may induce inflammation and gut dysbiosis, and worsen symptoms of estrogen deficiency. To investigate this hypothesis, vehicle (Veh)- or VCD-treated C57BL/6J mice were fed a HFD (45% kcal fat) with a high LA:SFA ratio (22.5%: 8%), referred to as the 22.5% LA diet, or a HFD with a low LA:SFA ratio (1%: 31%), referred to as 1% LA diet, for a period of 23 to 25 weeks. Compared with VCD-treated mice fed the 22.5% LA diet, VCD-treated mice fed the 1% LA diet showed lower weight gain and improved glucose tolerance. However, VCD-treated mice fed the 1% LA diet had higher blood pressure and showed evidence of spatial cognitive impairment. Mice fed the 1% LA or 22.5% LA diets showed gut microbial taxa changes that have been associated with a mix of both beneficial and unfavorable cognitive and metabolic phenotypes. Overall, these data suggest that consuming different types of dietary fat from a variety of sources, without overemphasis on any particular type, is the optimal approach for promoting metabolic health regardless of estrogen status.
Collapse
Affiliation(s)
- Ke Sui
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ali Yasrebi
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Candace R Longoria
- Department of Kinesiology and Applied Physiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Avery T MacDonell
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Zehra H Jaffri
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Savannah A Martinez
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Samuel E Fisher
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Natasha Malonza
- Department of Kinesiology and Applied Physiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Katie Jung
- Department of Nutritional Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Kevin M Tveter
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Kimberly R Wiersielis
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Mehmet Uzumcu
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Sue A Shapses
- Department of Nutritional Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Sara C Campbell
- Department of Kinesiology and Applied Physiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Troy A Roepke
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research, Center for Human Nutrition, Exercise and Metabolism Center, and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Diana E Roopchand
- Department of Food Science, NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research and Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
11
|
Lindsey ML, Brunt KR, Kirk JA, Kleinbongard P, Calvert JW, de Castro Brás LE, DeLeon-Pennell KY, Del Re DP, Frangogiannis NG, Frantz S, Gumina RJ, Halade GV, Jones SP, Ritchie RH, Spinale FG, Thorp EB, Ripplinger CM, Kassiri Z. Guidelines for in vivo mouse models of myocardial infarction. Am J Physiol Heart Circ Physiol 2021; 321:H1056-H1073. [PMID: 34623181 PMCID: PMC8834230 DOI: 10.1152/ajpheart.00459.2021] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022]
Abstract
Despite significant improvements in reperfusion strategies, acute coronary syndromes all too often culminate in a myocardial infarction (MI). The consequent MI can, in turn, lead to remodeling of the left ventricle (LV), the development of LV dysfunction, and ultimately progression to heart failure (HF). Accordingly, an improved understanding of the underlying mechanisms of MI remodeling and progression to HF is necessary. One common approach to examine MI pathology is with murine models that recapitulate components of the clinical context of acute coronary syndrome and subsequent MI. We evaluated the different approaches used to produce MI in mouse models and identified opportunities to consolidate methods, recognizing that reperfused and nonreperfused MI yield different responses. The overall goal in compiling this consensus statement is to unify best practices regarding mouse MI models to improve interpretation and allow comparative examination across studies and laboratories. These guidelines will help to establish rigor and reproducibility and provide increased potential for clinical translation.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - John W Calvert
- Carlyle Fraser Heart Center of Emory University Hospital Midtown, Atlanta, Georgia
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Nikolaos G Frangogiannis
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Steven P Jones
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Victoria, Australia
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine and the Columbia Veteran Affairs Medical Center, Columbia, South Carolina
| | - Edward B Thorp
- Department of Pathology and Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
Pontifex MG, Martinsen A, Saleh RNM, Harden G, Tejera N, Müller M, Fox C, Vauzour D, Minihane AM. APOE4 genotype exacerbates the impact of menopause on cognition and synaptic plasticity in APOE-TR mice. FASEB J 2021; 35:e21583. [PMID: 33891334 DOI: 10.1096/fj.202002621rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 01/15/2023]
Abstract
The impact of sex and menopausal status in Alzheimer's disease remains understudied despite increasing evidence of greater female risk, particularly in APOE4 carriers. Utilizing female APOE-TR mice maintained on a high-fat diet background we induced ovarian failure through repeated VCD injections, to mimic human menopause. At 12 months of age, recognition memory and spatial memory were assessed using object recognition, Y-maze spontaneous alternation, and Barnes maze. A VCD*genotype interaction reduced the recognition memory (P < .05), with APOE4 VCD-treated animals unable to distinguish between novel and familiar objects. APOE4 mice displayed an additional 37% and 12% reduction in Barnes (P < .01) and Y-maze (P < .01) performance, indicative of genotype-specific spatial memory impairment. Molecular analysis indicated both VCD and genotype-related deficits in synaptic plasticity with BDNF, Akt, mTOR, and ERK signaling compromised. Subsequent reductions in the transcription factors Creb1 and Atf4 were also evident. Furthermore, the VCD*genotype interaction specifically diminished Ephb2 expression, while Fos, and Cnr1 expression reduced as a consequence of APOE4 genotype. Brain DHA levels were 13% lower in VCD-treated animals independent of genotype. Consistent with this, we detected alterations in the expression of the DHA transporters Acsl6 and Fatp4. Our results indicate that the combination of ovarian failure and APOE4 leads to an exacerbation of cognitive and neurological deficits.
Collapse
Affiliation(s)
| | | | | | - Glenn Harden
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Noemi Tejera
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Michael Müller
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Chris Fox
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|