1
|
Xing Z, Schocken DD, Zgibor JC, Alman AC. BMI, waist circumference, and waist-to-hip trajectories and all-cause, CVD, and cancer mortality by sex in people without diabetes. Int J Obes (Lond) 2025:10.1038/s41366-025-01778-6. [PMID: 40204962 DOI: 10.1038/s41366-025-01778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVES We examined the associations of BMI, waist circumference, and waist-to-hip ratio trajectories with mortality in people without diabetes. METHODS We analyzed 7601 people without diabetes from the Atherosclerosis Risk in Communities Study. We used latent class analysis to identify trajectory patterns for BMI, waist circumference, and waist-to-hip. We employed propensity score matching to enhance the balance of covariates and used Cox proportional hazards regression models to examine the associations. RESULTS In females, the high trajectory of BMI was associated with higher cancer mortality risks than the low group, with the hazard ratio and 95% confidence interval of 1.76 (1.14-2.73). The high waist circumference trajectory was related to increased all-cause, CVD, and cancer mortality risks in males. The moderate and high waist-to-hip ratio trajectories were associated with elevated all-cause and CVD mortality risks in females, and the high trajectory was associated with high all-cause mortality risks in males. The mean lifespan of deceased females did not significantly differ across the trajectories. However, the mean lifespan of males in the waist circumference high group (73.0 years) was shorter than the low group (75.3 years). CONCLUSIONS Sex differences were observed in the long-term impact of high BMI, waist circumference, and waist-to-hip ratio on mortality risks and lifespan in people without diabetes.
Collapse
Affiliation(s)
- Zailing Xing
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Douglas D Schocken
- College of Public Health, University of South Florida, Tampa, FL, USA
- School of Medicine, Duke University, Durham, NC, USA
| | - Janice C Zgibor
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Amy C Alman
- College of Public Health, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
2
|
Prodel E, Moreira M, Gondim M, Rocha H, Mira P, Nobrega A. Relationship between resting augmentation index and spontaneous cardiac baroreflex sensitivity during handgrip exercise in postmenopausal women. Braz J Med Biol Res 2025; 58:e14152. [PMID: 39907405 PMCID: PMC11793149 DOI: 10.1590/1414-431x2024e14152] [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: 05/14/2024] [Accepted: 11/22/2024] [Indexed: 02/06/2025] Open
Abstract
The stiffening of the conductance arteries is a hallmark of ageing and increases drastically after menopause. Therefore, the augmentation index (AIx), a surrogate for arterial stiffness, could be related to the decline in baroreflex sensitivity. We sought to investigate the relationship between resting AIx and spontaneous cardiac baroreflex sensitivity (cBRS) during handgrip exercise in ageing women. Thirteen young women (YW: 24±5 years; 24±2 kg/m2) and nine postmenopausal women (PMW: 60±5 years; 26±3 kg/m2) underwent the protocol, which consisted of 10 min of supine resting followed by 3 min of static handgrip exercise at 40% of the maximal voluntary force. The AIx was provided by the aortic pressure waveform and cBRS was calculated using the sequence technique, and vagal activity was accessed via heart rate variability using the root mean square of successive differences (RMSSD) index. Resting AIx was higher in PMW compared to YW (YW: 8±10%; PMW: 23±8%; P<0.01), while the cBRS (YW: 16±12 ms/mmHg; PMW: 10±5 ms/mmHg; P=0.08) and RMSSD (YW: 46±35 ms; PMW: 34±12 ms; P=0.26) were similar in YW and PMW. At rest, there was no significant (P>0.05) relationship between the AIx and cBRS in YW and PMW. However, in PMW, a negative (slope=-0.22) and strong (r=-0.70; P=0.03) relationship was observed between AIx and cBRS for the increment of blood pressure during the handgrip exercise. The stiffening of the arterial tree is one possible mechanism to explain the decrease of spontaneous cardiac baroreflex sensitivity during exercise in postmenopausal women.
Collapse
Affiliation(s)
- E. Prodel
- Laboratório de Ciências do Exercício, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, RJ, Brasil
| | - M.F.S.C. Moreira
- Departamento de Educação Física, Universidade Federal Fluminense, Niterói, RJ, Brasil
| | - M.L. Gondim
- Laboratório de Ciências do Exercício, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, RJ, Brasil
| | - H.N.M. Rocha
- Laboratório de Ciências do Exercício, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, RJ, Brasil
- Laboratório de Cardiometabologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, RJ, Brasil
| | - P.A.C. Mira
- Laboratório de Ciências do Exercício, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, RJ, Brasil
| | - A.C.L. Nobrega
- Laboratório de Ciências do Exercício, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, RJ, Brasil
| |
Collapse
|
3
|
Battipaglia C, Genazzani AD, Nappi RE, La Marca A. Insights on estetrol, the native estrogen: from contraception to hormone replacement therapy. Minerva Obstet Gynecol 2024; 76:590-603. [PMID: 39283289 DOI: 10.23736/s2724-606x.24.05594-5] [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: 01/21/2025]
Abstract
Estetrol (E4) is a natural estrogen that has recently emerged as new option for contraception and hormone replacement therapy (HRT). Unlike other estrogens, E4 primarily stimulates nuclear estrogen receptor alpha (ERα) and does not activate membrane ERα. For this reason, this novel estrogen has tissue-specific effects across various organs such as liver, vascular endothelium, mammary glands, brain, vagina, and uterus. The selective activation of the nuclear ERα results in distinct pharmacological properties that contribute to its unique therapeutic profile. Moreover, E4 shows minimal interaction with the hepatic cytochrome P450 enzyme system, leading to a favorable pharmacokinetic profile and a reduced potential for drug-drug interactions. Currently, E4 is commercially available in combination with drospirenone as a combined oral contraceptive and its application in HRT is undergoing late-stage clinical development. Many studies have demonstrated that E4 has a lower impact on hemostatic and metabolic parameters compared to other estrogens, potentially reducing the risk of adverse effects commonly associated with hormonal therapies such as thromboembolic events or dyslipidemia. Beyond its role in contraception and HRT, E4 shows promising therapeutic potential in other medical fields, including neuroprotection in neonatal hypoxic-ischemic encephalopathy, enhancement of hematopoietic stem cell transplantation outcomes and prostate cancer management. This review synthesizes the latest evidence on E4 primarily focusing on its pharmacological characteristics and clinical applications. The findings suggest that E4 versatility and peculiar mechanism of action may represent an important therapeutic option for a broad spectrum of medical conditions.
Collapse
Affiliation(s)
- Christian Battipaglia
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy -
| | - Alessandro D Genazzani
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS S. Matteo Foundation, Pavia, Italy
| | - Antonio La Marca
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
4
|
Yousef A, Sosnowski DK, Fang L, Legaspi RJ, Korodimas J, Lee A, Magor KE, Seubert JM. Cardioprotective response and senescence in aged sEH null female mice exposed to LPS. Am J Physiol Heart Circ Physiol 2024; 326:H1366-H1385. [PMID: 38578240 DOI: 10.1152/ajpheart.00706.2023] [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: 11/08/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Deterioration of physiological systems, like the cardiovascular system, occurs progressively with age impacting an individual's health and increasing susceptibility to injury and disease. Cellular senescence has an underlying role in age-related alterations and can be triggered by natural aging or prematurely by stressors such as the bacterial toxin lipopolysaccharide (LPS). The metabolism of polyunsaturated fatty acids by CYP450 enzymes produces numerous bioactive lipid mediators that can be further metabolized by soluble epoxide hydrolase (sEH) into diol metabolites, often with reduced biological effects. In our study, we observed age-related cardiac differences in female mice, where young mice demonstrated resistance to LPS injury, and genetic deletion or pharmacological inhibition of sEH using trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid attenuated LPS-induced cardiac dysfunction in aged female mice. Bulk RNA-sequencing analyses revealed transcriptomics differences in aged female hearts. The confirmatory analysis demonstrated changes to inflammatory and senescence gene markers such as Il-6, Mcp1, Il-1β, Nlrp3, p21, p16, SA-β-gal, and Gdf15 were attenuated in the hearts of aged female mice where sEH was deleted or inhibited. Collectively, these findings highlight the role of sEH in modulating the aging process of the heart, whereby targeting sEH is cardioprotective.NEW & NOTEWORTHY Soluble epoxide hydrolase (sEH) is an essential enzyme for converting epoxy fatty acids to their less bioactive diols. Our study suggests deletion or inhibition of sEH impacts the aging process in the hearts of female mice resulting in cardioprotection. Data indicate targeting sEH limits inflammation, preserves mitochondria, and alters cellular senescence in the aged female heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Renald James Legaspi
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jacob Korodimas
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andy Lee
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Katharine E Magor
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Drinković N, Beus M, Barbir R, Debeljak Ž, Tariba Lovaković B, Kalčec N, Ćurlin M, Bekavac A, Gorup D, Mamić I, Mandić D, Micek V, Turčić P, Günday-Türeli N, Türeli E, Vinković Vrček I. Novel PLGA-based nanoformulation decreases doxorubicin-induced cardiotoxicity. NANOSCALE 2024; 16:9412-9425. [PMID: 38650478 DOI: 10.1039/d3nr06269d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Nanotechnology has the potential to provide formulations of antitumor agents with increased selectivity towards cancer tissue thereby decreasing systemic toxicity. This in vivo study evaluated the potential of novel nanoformulation based on poly(lactic-co-glycolic acid) (PLGA) to reduce the cardiotoxic potential of doxorubicin (DOX). In vivo toxicity of PLGADOX was compared with clinically approved non-PEGylated, liposomal nanoformulation of DOX (LipoDOX) and conventional DOX form (ConvDOX). The study was performed using Wistar Han rats of both sexes that were treated intravenously for 28 days with 5 doses of tested substances at intervals of 5 days. Histopathological analyses of heart tissues showed the presence of myofiber necrosis, degeneration processes, myocytolysis, and hemorrhage after treatment with ConvDOX, whereas only myofiber degeneration and hemorrhage were present after the treatment with nanoformulations. All DOX formulations caused an increase in the troponin T with the greatest increase caused by convDOX. qPCR analyses revealed an increase in the expression of inflammatory markers IL-6 and IL-8 after ConvDOX and an increase in IL-8 expression after lipoDOX treatments. The mass spectra imaging (MSI) of heart tissue indicates numerous metabolic and lipidomic changes caused by ConvDOX, while less severe cardiac damages were found after treatment with nanoformulations. In the case of LipoDOX, autophagy and apoptosis were still detectable, whereas PLGADOX induced only detectable mitochondrial toxicity. Cardiotoxic effects were frequently sex-related with the greater risk of cardiotoxicity observed mostly in male rats.
Collapse
Affiliation(s)
| | - Maja Beus
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Rinea Barbir
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Željko Debeljak
- JJ Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia
- University Hospital Osijek, Osijek, Croatia
| | | | - Nikolina Kalčec
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | | | - Ana Bekavac
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Dunja Gorup
- Department of Neuroradiology, Klinik für Neuroradiology, Universitätspital Zürich Universitätsspital Zürich, 8006 Zürich, Switzerland
| | - Ivan Mamić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | | | - Vedran Micek
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Petra Turčić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | | | | | - Ivana Vinković Vrček
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
- University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
6
|
Karolczak K, Watala C. Estradiol as the Trigger of Sirtuin-1-Dependent Cell Signaling with a Potential Utility in Anti-Aging Therapies. Int J Mol Sci 2023; 24:13753. [PMID: 37762053 PMCID: PMC10530977 DOI: 10.3390/ijms241813753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Aging entails the inevitable loss of the structural and functional integrity of cells and tissues during the lifetime. It is a highly hormone-dependent process; although, the exact mechanism of hormone involvement, including sex hormones, is unclear. The marked suppression of estradiol synthesis during menopause suggests that the hormone may be crucial in maintaining cell lifespan and viability in women. Recent studies also indicate that the same may be true for men. Similar anti-aging features are attributed to sirtuin 1 (SIRT1), which may possibly be linked at the molecular level with estradiol. This finding may be valuable for understanding the aging process, its regulation, and possible prevention against unhealthy aging. The following article summarizes the initial studies published in this field with a focus on age-associated diseases, like cancer, cardiovascular disease and atherogenic metabolic shift, osteoarthritis, osteoporosis, and muscle damage, as well as neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Kamil Karolczak
- Department of Haemostatic Disorders, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland;
| | | |
Collapse
|
7
|
LaPenna KB, Li Z, Doiron JE, Sharp TE, Xia H, Moles K, Koul K, Wang JS, Polhemus DJ, Goodchild TT, Patel RB, Shah SJ, Lefer DJ. Combination Sodium Nitrite and Hydralazine Therapy Attenuates Heart Failure With Preserved Ejection Fraction Severity in a "2-Hit" Murine Model. J Am Heart Assoc 2023; 12:e028480. [PMID: 36752224 PMCID: PMC10111505 DOI: 10.1161/jaha.122.028480] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2023]
Abstract
Background Recent studies have suggested that cardiac nitrosative stress mediated by pathological overproduction of nitric oxide (NO) via inducible NO synthase (iNOS) contributes to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). Other studies have suggested that endothelial NO synthase (eNOS) dysfunction and attenuated NO bioavailability contribute to HFpEF morbidity and mortality. We sought to further investigate dysregulated NO signaling and to examine the effects of a NO-based dual therapy (sodium nitrite+hydralazine) following the onset of HFpEF using a "2-hit" murine model. Methods and Results Nine-week-old male C57BL/6 N mice (n=15 per group) were treated concurrently with high-fat diet and N(ω)-nitro-L-arginine methyl ester (L-NAME) (0.5 g/L per day) via drinking water for 10 weeks. At week 5, mice were randomized into either vehicle (normal saline) or combination treatment with sodium nitrite (75 mg/L in the drinking water) and hydralazine (2.0 mg/kg IP, BID). Cardiac structure and function were monitored with echocardiography and invasive hemodynamic measurements. Cardiac mitochondrial respiration, aortic vascular function, and exercise performance were also evaluated. Circulating and myocardial nitrite were measured to determine the bioavailability of NO. Circulating markers of oxidative or nitrosative stress as well as systemic inflammation were also determined. Severe HFpEF was evident by significantly elevated E/E', LVEDP, and Tau in mice treated with L-NAME and HFD, which was associated with impaired NO bioavailability, mitochondrial respiration, aortic vascular function, and exercise capacity. Treatment with sodium nitrite and hydralazine restored NO bioavailability, reduced oxidative and nitrosative stress, preserved endothelial function and mitochondrial respiration, limited the fibrotic response, and improved exercise capacity, ultimately attenuating the severity of "two-hit" HFpEF. Conclusions Our data demonstrate that nitrite, a well-established biomarker of NO bioavailability and a physiological source of NO, is significantly reduced in the heart and circulation in the "2-hit" mouse HFpEF model. Furthermore, sodium nitrite+hydralazine combined therapy significantly attenuated the severity of HFpEF in the "2-hit" cardiometabolic HFpEF. These data suggest that supplementing NO-based therapeutics with a potent antioxidant and vasodilator agent may result in synergistic benefits for the treatment of HFpEF.
Collapse
Affiliation(s)
- Kyle B. LaPenna
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA
| | - Zhen Li
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Jake E. Doiron
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA
| | - Thomas E. Sharp
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Medicine, Section of CardiologyLouisiana State University Health Sciences CenterNew OrleansLA
| | - Huijing Xia
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - Karl Moles
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - Kashyap Koul
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - John S. Wang
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | | | - Traci T. Goodchild
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Ravi B. Patel
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoIL
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoIL
| | - David J. Lefer
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| |
Collapse
|
8
|
Sheshadri D, Onkar A, Ganesh S. Alterations in brain glycogen levels influence life-history traits and reduce the lifespan in female Drosophila melanogaster. Biol Open 2021; 10:273730. [PMID: 34817590 PMCID: PMC8689487 DOI: 10.1242/bio.059055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
Abstract
Sexual dimorphism in lifespan, wherein females outlive males, is evident across all animal taxa. The longevity difference between sexes is controlled by multiple physiological processes with complex relationships to one another. In recent years, glycogen, the storage form of glucose, has been shown to cause rapid aging upon forced synthesis in healthy neurons. Glycogen in the form of corpora amylacea in the aging brain is also widely reported. While these studies did suggest a novel role for glycogen in aging, most of them have focused on pooled samples, and have not looked at sex-specific effects, if any. Given the widespread occurrence of sex-biased expression of genes and the underlying physiology, it is important to look at the sex-specific effects of metabolic processes. In the present study, using transgenic fly lines for the human glycogen synthase, we investigated the sex-specific effects of glycogen on stress resistance, fitness, and survival. We demonstrate that Drosophila melanogaster females with altered levels of glycogen in the brain display a shortened lifespan, increased resistance to starvation, and higher oxidative stress than male flies. The present study thus provides a novel insight into the sex-specific effect of glycogen in survival and aging and how differences in metabolic processes could contribute to sex-specific traits.
Collapse
Affiliation(s)
- Deepashree Sheshadri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Akanksha Onkar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
9
|
Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex Differences in Molecular Mechanisms of Cardiovascular Aging. FRONTIERS IN AGING 2021; 2:725884. [PMID: 35822017 PMCID: PMC9261391 DOI: 10.3389/fragi.2021.725884] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of illness and death in the Western world. Cardiovascular aging is a progressive modification occurring in cardiac and vascular morphology and physiology where increased endothelial dysfunction and arterial stiffness are observed, generally accompanied by increased systolic blood pressure and augmented pulse pressure. The effects of biological sex on cardiovascular pathophysiology have long been known. The incidence of hypertension is higher in men, and it increases in postmenopausal women. Premenopausal women are protected from CVD compared with age-matched men and this protective effect is lost with menopause, suggesting that sex-hormones influence blood pressure regulation. In parallel, the heart progressively remodels over the course of life and the pattern of cardiac remodeling also differs between the sexes. Lower autonomic tone, reduced baroreceptor response, and greater vascular function are observed in premenopausal women than men of similar age. However, postmenopausal women have stiffer arteries than their male counterparts. The biological mechanisms responsible for sex-related differences observed in cardiovascular aging are being unraveled over the last several decades. This review focuses on molecular mechanisms underlying the sex-differences of CVD in aging.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Jennifer C. Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fernanda R. Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - R. Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
10
|
Age and sex modify cellular proliferation responses to oxidative stress and glucocorticoid challenges in baboon cells. GeroScience 2021; 43:2067-2085. [PMID: 34089175 DOI: 10.1007/s11357-021-00395-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022] Open
Abstract
Aging is associated with progressive loss of cellular homeostasis resulting from intrinsic and extrinsic challenges. Lack of a carefully designed, well-characterized, precise, translational experimental model is a major limitation to understanding the cellular perturbations that characterize aging. Here, we tested the feasibility of primary fibroblasts isolated from nonhuman primates (baboons) as a model of cellular resilience in response to homeostatic challenge. Using a real-time live-cell imaging system, we precisely defined a protocol for testing effects of prooxidant compounds (e.g., hydrogen peroxide (H2O2), paraquat), thapsigargin, dexamethasone, and a low glucose environment on cell proliferation in fibroblasts derived from baboons across the life course (n = 11/sex). Linear regression analysis indicated that donor age significantly reduced the ability of cells to proliferate following exposure to H2O2 (50 and 100 µM) and paraquat (100 and 200 µM) challenges in cells from males (6.4-21.3 years; average lifespan 21 years) but not cells from females (4.3-15.9 years). Inhibitory effects of thapsigargin on cell proliferation were dependent on challenge duration (2 vs 24 h) and concentration (0.1 and 1 µM). Cells from older females (14.4-15.9 years) exhibited greater resilience to thapsigargin (1 µM; 24 h) and dexamethasone (500 µM) challenges than did those from younger females (4.3-6.7 years). The cell proliferation response to low glucose (1 mM) was reduced with age in both sexes. These data indicate that donor's chronological age and sex are important variables in determining fibroblast responses to metabolite and other challenges.
Collapse
|
11
|
Teixeira VP, Miranda K, Scalzo S, Rocha-Resende C, Silva MM, Tezini GCSV, Melo MB, Souza-Neto FP, Silva KSC, Jesus ICG, Santos AK, de Oliveira M, Szawka RE, Salgado HC, Prado MAM, Poletini MO, Guatimosim S. Increased cholinergic activity under conditions of low estrogen leads to adverse cardiac remodeling. Am J Physiol Cell Physiol 2021; 320:C602-C612. [PMID: 33296286 DOI: 10.1152/ajpcell.00142.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Cholinesterase inhibitors are used in postmenopausal women for the treatment of neurodegenerative diseases. Despite their widespread use in the clinical practice, little is known about the impact of augmented cholinergic signaling on cardiac function under reduced estrogen conditions. To address this gap, we subjected a genetically engineered murine model of systemic vesicular acetylcholine transporter overexpression (Chat-ChR2) to ovariectomy and evaluated cardiac parameters. Left-ventricular function was similar between Chat-ChR2 and wild-type (WT) mice. Following ovariectomy, WT mice showed signs of cardiac hypertrophy. Conversely, ovariectomized (OVX) Chat-ChR2 mice evolved to cardiac dilation and failure. Transcript levels for cardiac stress markers atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) were similarly upregulated in WT/OVX and Chat-ChR2/OVX mice. 17β-Estradiol (E2) treatment normalized cardiac parameters in Chat-ChR2/OVX to the Chat-ChR2/SHAM levels, providing a link between E2 status and the aggravated cardiac response in this model. To investigate the cellular basis underlying the cardiac alterations, ventricular myocytes were isolated and their cellular area and contractility were assessed. Myocytes from WT/OVX mice were wider than WT/SHAM, an indicative of concentric hypertrophy, but their fractional shortening was similar. Conversely, Chat-ChR2/OVX myocytes were elongated and presented contractile dysfunction. E2 treatment again prevented the structural and functional changes in Chat-ChR2/OVX myocytes. We conclude that hypercholinergic mice under reduced estrogen conditions do not develop concentric hypertrophy, a critical compensatory adaptation, evolving toward cardiac dilation and failure. This study emphasizes the importance of understanding the consequences of cholinesterase inhibition, used clinically to treat dementia, for cardiac function in postmenopausal women.
Collapse
MESH Headings
- Acetylcholine/metabolism
- Animals
- Cholinergic Fibers/metabolism
- Estradiol/pharmacology
- Estrogen Replacement Therapy
- Estrogens/deficiency
- Female
- Heart/innervation
- Heart Rate
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Mice, Inbred C57BL
- Mice, Transgenic
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Ovariectomy
- Signal Transduction
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Vesicular Acetylcholine Transport Proteins/genetics
- Vesicular Acetylcholine Transport Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Vanessa P Teixeira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kiany Miranda
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio Scalzo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cibele Rocha-Resende
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mário Morais Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geisa C S V Tezini
- Ribeirão Preto Medical School, Universidade de São Paulo, Riberão Preto, São Paulo, Brazil
| | - Marcos B Melo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando Pedro Souza-Neto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kaoma S C Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Itamar C G Jesus
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anderson K Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro de Oliveira
- Ribeirão Preto Medical School, Universidade de São Paulo, Riberão Preto, São Paulo, Brazil
| | - Raphael E Szawka
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Helio C Salgado
- Ribeirão Preto Medical School, Universidade de São Paulo, Riberão Preto, São Paulo, Brazil
| | - Marco Antonio Máximo Prado
- Robarts Research Institute, Department of Physiology and Pharmacology and Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Maristela O Poletini
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
12
|
Ward LJ, Nilsson S, Hammar M, Lindh-Åstrand L, Berin E, Lindblom H, Spetz Holm AC, Rubér M, Li W. Resistance training decreases plasma levels of adipokines in postmenopausal women. Sci Rep 2020; 10:19837. [PMID: 33199796 PMCID: PMC7669850 DOI: 10.1038/s41598-020-76901-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/03/2020] [Indexed: 01/04/2023] Open
Abstract
Physical inactivity and the onset of menopause increase the risk of cardiovascular disease amongst postmenopausal women. We aim to investigate the effect of resistance training (RT) on plasma levels of selected cytokines, adipokines, myokines, and sex hormones in postmenopausal women with vasomotor symptoms. This was a sub-study of a randomised controlled trial investigating the effects of RT on vasomotor symptoms in postmenopausal women. Women were randomised to join a 15-week RT program (n = 26) or remain sedentary as control (n = 29). Venous blood samples were taken at week-0 and week-15 for all participants. Enzyme-linked immunosorbent assays and multiple bead assays were used to measure cytokines, adipokines, myokines, and sex hormones in plasma. Plasma measurements of 16 of 33 analytes were within detectable limits. After adjusting for good compliance in the RT group (58% of RT participants), after 15 weeks, significantly lower plasma levels of adiponectin (p < 0.001), lipocalin-2 (p < 0.01) and resistin (p = 0.04) were found. Comparing control and RT women, using change-over-time values, significant increases in median testosterone and sex hormone binding globulin levels were seen in RT women. RT intervention lowers the levels of adipokines, particularly adiponectin, in postmenopausal women with vasomotor symptoms. These results were secondary outcomes of a clinical trial, and further investigations in a larger cohort are essential with the additional control of diet control and body composition analyses. Nevertheless, our study shows RT may be a beneficial intervention in reducing inflammation amongst postmenopausal women.
Collapse
Affiliation(s)
- Liam J Ward
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden. .,Occupational and Environmental Medicine Center in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, 58185, Linköping, Sweden. .,Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186, Stockholm, Sweden.
| | - Sigrid Nilsson
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mats Hammar
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lotta Lindh-Åstrand
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Emilia Berin
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hanna Lindblom
- Department of Health, Medicine and Caring Sciences, Unit of Physiotherapy, Linköping University, Linköping, Sweden
| | - Anna-Clara Spetz Holm
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Marie Rubér
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Wei Li
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
13
|
Ward LJ, Hammar M, Lindh-Åstrand L, Berin E, Lindblom H, Rubér M, Spetz Holm AC, Li W. Does resistance training have an effect on levels of ferritin and atherogenic lipids in postmenopausal women? - A pilot trial. Sci Rep 2020; 10:3838. [PMID: 32123242 PMCID: PMC7052219 DOI: 10.1038/s41598-020-60759-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/10/2020] [Indexed: 12/29/2022] Open
Abstract
The objective of this study was to determine if 15 weeks of resistance training (RT) can alter the levels of blood lipids, body iron status, and oxidative stress in postmenopausal women with vasomotor symptoms. Postmenopausal women enrolled in a randomised controlled trial were allocated to either a sedentary control group (n = 29) or a RT group (n = 26). Blood samples were taken at week-0 and week-15 for all participants. Blood lipids and iron status were measured via routine clinical analyses. Immunoassays were used to measure oxidative stress markers. The RT group, with good compliance, was associated with significant reductions in ferritin, total cholesterol, low-density lipoprotein, and non-high-density lipoprotein cholesterol. Moreover, ferritin was positively correlated with atherogenic lipids while negatively correlated with high-density lipoprotein in RT women. This occurred without alterations in serum iron, transferrin, transferrin-saturation, C-reactive protein and oxidative stress markers. No differences were found in control women. This study suggests that RT in postmenopausal women both reduces levels of ferritin and counteracts atherogenic lipid profiles independent of an apparent oxidative mechanism. RT may be a beneficial intervention in postmenopausal women via an interaction between ferritin and lipids; however, further investigation in a larger cohort is essential.
Collapse
Affiliation(s)
- Liam J Ward
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden. .,Occupational and Environmental Medicine Center in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
| | - Mats Hammar
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lotta Lindh-Åstrand
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Emilia Berin
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hanna Lindblom
- Department of Health, Medicine and Caring Sciences, Unit of Physiotherapy, Linköping University, Linköping, Sweden
| | - Marie Rubér
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna-Clara Spetz Holm
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Wei Li
- Department of Obstetrics and Gynaecology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
14
|
Bassiouni SS, White EM, Karvonen-Gutierrez C, Harlow SD. Lack of food access and food consumption patterns of late midlife women in southeast Michigan. MICHIGAN JOURNAL OF PUBLIC HEALTH 2020; 10:8. [PMID: 38274401 PMCID: PMC10809997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
|
15
|
Januário Costa T, Jiménez-Altayó F, Echem C, Akamine EH, Tostes R, Vila E, Dantas AP, Catelli de Carvalho MH. Late Onset of Estrogen Therapy Impairs Carotid Function of Senescent Females in Association with Altered Prostanoid Balance and Upregulation of the Variant ERα36. Cells 2019; 8:cells8101217. [PMID: 31597326 PMCID: PMC6829869 DOI: 10.3390/cells8101217] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 01/14/2023] Open
Abstract
Recent analysis of clinical trials on estrogen therapy proposes the existence of a therapeutic window of opportunity for the cardiovascular benefits of estrogens, which depend on women's age and the onset of therapy initiation. In this study, we aimed to determine how vascular senescence and the onset of estrogen treatment influence the common carotid artery (CCA) function in senescent and non-senescent females. Ovariectomized female senescence-accelerated (SAMP8) or non-senescent (SAMR1) mice were treated with vehicle (OVX) or 17β-estradiol starting at the day of ovariectomy (early-onset, E2E) or 45 days after surgery (late-onset, E2L). In SAMR1, both treatments, E2E and E2L, reduced constriction to phenylephrine (Phe) in CCA [(AUC) OVX: 193.8 ± 15.5; E2E: 128.1 ± 11.6; E2L: 130.2 ± 15.8, p = 0.004] in association with positive regulation of NO/O2- ratio and increased prostacyclin production. In contrast, E2E treatment did not modify vasoconstrictor responses to Phe in OVX-SAMP8 and, yet, E2L increased Phe vasoconstriction [(AUC) OVX: 165.3 ± 10; E2E: 183.3 ± 11.1; E2L: 256.3 ± 30.4, p = 0.005]. Increased vasoconstriction in E2L-SAMP8 was associated with augmented thromboxane A2 and reduced NO production. Analysis of wild-type receptor alpha (ERα66) expression and its variants revealed an increased expression of ERα36 in E2L-SAMP8 in correlation with unfavorable effects of estrogen in those animals. In conclusion, estrogen exerts beneficial effects in non-senescent CCA, regardless of the initiation of the therapy. In senescent CCA, however, estrogen loses its beneficial action even when administered shortly after ovariectomy and may become detrimental when given late after ovariectomy. Aging and onset of estrogen treatment are two critical factors in the mechanism of action of this hormone in CCA.
Collapse
Affiliation(s)
- Tiago Januário Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil; (T.J.C.); (C.E.); (E.H.A.); (M.H.C.d.C.)
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (F.J.-A.); (E.V.)
- Group of Atherosclerosis and Coronary Disease, Institut Clinic del Torax, Institut d’Investigaciones Biomédiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Pharmacology Department, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo 14049-900, Brazil;
| | - Francesc Jiménez-Altayó
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (F.J.-A.); (E.V.)
| | - Cinthya Echem
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil; (T.J.C.); (C.E.); (E.H.A.); (M.H.C.d.C.)
| | - Eliana Hiromi Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil; (T.J.C.); (C.E.); (E.H.A.); (M.H.C.d.C.)
| | - Rita Tostes
- Pharmacology Department, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo 14049-900, Brazil;
| | - Elisabet Vila
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (F.J.-A.); (E.V.)
| | - Ana Paula Dantas
- Group of Atherosclerosis and Coronary Disease, Institut Clinic del Torax, Institut d’Investigaciones Biomédiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Correspondence:
| | - Maria Helena Catelli de Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil; (T.J.C.); (C.E.); (E.H.A.); (M.H.C.d.C.)
| |
Collapse
|
16
|
Ghimire S, Hill CV, Sy FS, Rodriguez R. Decline in telomere length by age and effect modification by gender, allostatic load and comorbidities in National Health and Nutrition Examination Survey (1999-2002). PLoS One 2019; 14:e0221690. [PMID: 31469870 PMCID: PMC6716670 DOI: 10.1371/journal.pone.0221690] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study aims to assess the decline in telomere length (TL) with age and evaluate effect modification by gender, chronic stress, and comorbidity in a representative sample of the US population. METHODS Cross-sectional data on 7826 adults with a TL measurement, were included from the National Health and Nutrition Examination Survey, years 1999-2002. The population rate of decline in TL across 10-year age categories was estimated using crude and adjusted regression. RESULTS In an adjusted model, the population rate of decline in TL with age was consistent and linear for only three age categories: 20-29 (β = -0.0172, 95% CI: -0.0342, -0.0002), 50-59 (β = -0.0182, 95% CI: -0.0311, -0.0054) and 70-79 (β = -0.0170, 95% CI: -0.0329, -0.0011) years. The population rate of decline in TL with age was significantly greater for males and those with high allostatic load and a history of comorbidities. When the population rate of decline in TL was analyzed by gender in 10-year age bins, a fairly consistent yet statistically non-significant decline for males was observed; however, a trough in the rate was observed for females in the age categories 20-29 years (β = -0.0284, 95% CI: -0.0464, -0.0103) and 50-59 years (β = -0.0211, 95% CI: -0.0391, -0.0032). To further elucidate the gender difference observed in the primary analyses, secondary analyses were conducted with reproductive and hormonal status; a significant inverse association was found between TL and parity, menopause, and age at menopause. CONCLUSIONS TL was shorter with increasing age and this decline was modified by gender, chronic stress and comorbidities; individuals with chronic morbidity and/or chronic stress and females in their twenties and fifties experienced greater decline. Female reproductive factors, i.e., parity and menopause, were associated with TL.
Collapse
Affiliation(s)
- Saruna Ghimire
- Department of Sociology and Gerontology, Miami University, Oxford, OH, United States of America
- School of Public Health, University of Nevada Las Vegas, Las Vegas, NV, United States of America
- * E-mail:
| | - Carl V. Hill
- Office of Special Populations, National Institute of Aging, Bethesda, MD, United States of America
| | - Francisco S. Sy
- School of Public Health, University of Nevada Las Vegas, Las Vegas, NV, United States of America
| | | |
Collapse
|
17
|
Wu CH, Wang SE, Hsu CH, Hsu YT, Lu CW, Chuang WC, Lee MC. Reproductive Regulation and Oxidative Stress Alleviation of Chinese Herbal Medicine Therapy in Ovariectomised Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:5346518. [PMID: 31360207 PMCID: PMC6642789 DOI: 10.1155/2019/5346518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/27/2019] [Accepted: 06/18/2019] [Indexed: 11/17/2022]
Abstract
In Taiwan, the herbal formula B401 is considered as a health supplement for middle-aged women that can alleviate sweating, anxiety, and sleep disorders. However, the relevant mechanisms are still unclear. In this study, we evaluated the beneficial effects of the herbal formula B401 therapy in the reproductive regulation of ovariectomised mice. Female ICR mice were randomised into four groups: wild-type (WT) mice with sham treatment, wild-type mice treated with the herbal formula B401, bilateral ovariectomised (OVX) mice with sham treatment, and bilateral ovariectomised mice treated with the herbal formula B401. Mice were orally given the herbal formula B401 at a dose of 30 mg/kg bw/day for 2 weeks. At the end of oral treatment with sham or the herbal formula B401, levels of reactive oxygen species (ROS), calcium, phosphorus, and estradiol-17β in the blood; uterine weight and endometrial thickness; and expressions of estrogen receptor α (ERα), estrogen receptor β (ERβ), progesterone receptor (PR), vascular endothelial growth factor (VEGF), and superoxide dismutase 2 (SOD2) in the uterine tissue were examined and then compared among the four groups of mice. We found that OVX mice decreased levels of calcium, phosphorus, and estradiol-17β in the blood, decreased uterine weight and endometrial thickness, and decreased expressions of ERα, ERβ, PR, and SOD2 in the uterine tissue but increased blood ROS levels compared with those of WT mice. In addition, OVX mice with the herbal formula B401 therapy can increase levels of calcium, phosphorus, and estradiol-17β in the blood, increase uterine weight and endometrial thickness, and increase expressions of ERα, ERβ, PR, VEGF, and SOD2 in the uterine tissue but decrease blood ROS levels. Our results may provide reasonable explanation for the reproductive regulation of the herbal formula B401 therapy.
Collapse
Affiliation(s)
- Chung-Hsin Wu
- School of Life Science, National Taiwan Normal University, Taipei City, Taiwan
| | - Sheue-Er Wang
- Pathological Department, Saint Paul's Hospital, Taoyuan City, Taiwan
| | - Chih-Hsiang Hsu
- School of Life Science, National Taiwan Normal University, Taipei City, Taiwan
| | - Yu-Tsen Hsu
- School of Life Science, National Taiwan Normal University, Taipei City, Taiwan
| | - Chen-Wen Lu
- School of Life Science, National Taiwan Normal University, Taipei City, Taiwan
| | - Wu-Chang Chuang
- Sun Ten Pharmaceutical Co. Ltd., New Taipei City 23143, Taiwan
| | - Ming-Chung Lee
- Brion Research Institute of Taiwan, New Taipei City 23143, Taiwan
| |
Collapse
|
18
|
Ward LJ, Olausson P, Li W, Yuan XM. Proteomics and multivariate modelling reveal sex-specific alterations in distinct regions of human carotid atheroma. Biol Sex Differ 2018; 9:54. [PMID: 30594242 PMCID: PMC6311011 DOI: 10.1186/s13293-018-0217-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/12/2018] [Indexed: 01/05/2023] Open
Abstract
Background Atherosclerotic lesions are comprised of distinct regions with different proteomic profiles. Men and women develop differences in lesion phenotype, with lesions from women generally being more stable and less prone to rupture. We aimed to investigate the differences in proteomic profiles between sexes, including distinct lesion regions, to identify altered proteins that contribute to these differences observed clinically. Methods Carotid endarterectomy samples (ten men/ten women) were obtained, and intraplaque biopsies from three distinct regions (internal control, fatty streak and plaque) were analysed by tandem-mass spectrometry. Multivariate statistical modelling, using orthogonal partial least square-discriminant analysis, was used to discriminate the proteomes between men and women. Results Multivariate discriminant modelling revealed proteins from 16 functional groups that displayed sex-specific associations. Additional statistics revealed ten proteins that display region-specific alterations when comparing sexes, including proteins related to inflammatory response, response to reactive oxygen species, complement activation, transport and blood coagulation. Transport protein afamin and blood coagulation proteins antithrombin-III and coagulation factor XII were significantly increased in plaque region from women. Inflammatory response proteins lysozyme C and phospholipase A2 membrane-associated were significantly increased in plaque region from men. Limitations with this study are the small sample size, limited patient information and lack of complementary histology to control for cell type differences between sexes. Conclusions This pilot study, for the first time, utilises a multivariate proteomic approach to investigate sexual dimorphism in human atherosclerotic tissue, and provides an essential proteomic platform for further investigations to help understand sexual dimorphism and plaque vulnerability in atherosclerosis. Electronic supplementary material The online version of this article (10.1186/s13293-018-0217-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liam J Ward
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden. .,Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Patrik Olausson
- Pain and Rehabilitation Centre, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Wei Li
- Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Linköping University, SE-581 85, Linköping, Sweden
| | - Xi-Ming Yuan
- Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
19
|
Marais GA, Gaillard JM, Vieira C, Plotton I, Sanlaville D, Gueyffier F, Lemaitre JF. Sex gap in aging and longevity: can sex chromosomes play a role? Biol Sex Differ 2018; 9:33. [PMID: 30016998 PMCID: PMC6050741 DOI: 10.1186/s13293-018-0181-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 05/27/2018] [Indexed: 12/15/2022] Open
Abstract
It is well known that women live longer than men. This gap is observed in most human populations and can even reach 10-15 years. In addition, most of the known super centenarians (i.e., humans who lived for > 110 years) are women. The differences in life expectancy between men and women are often attributed to cultural differences in common thinking. However, sex hormones seem to influence differences in the prevalence of diseases, in the magnitude of aging, and in the longevity between men and women. Moreover, far from being human specific, the sex gap in longevity is extremely common in non-human animals, especially in mammals. Biological factors clearly contribute to such a sex gap in aging and longevity. Different hypotheses have been proposed to explain why males and females age and die differently. The cost of sexual selection and sexual dimorphism has long been considered the best explanation for the observed sex gap in aging/longevity. However, the way mitochondria are transmitted (i.e., through females in most species) could have an effect, called the mother's curse. Recent data suggest that sex chromosomes may also contribute to the sex gap in aging/longevity through several potential mechanisms, including the unguarded X/Z, the toxic Y/W and the loss of Y/W. We discuss future research directions to test these ideas.
Collapse
Affiliation(s)
- Gabriel A.B. Marais
- Laboratoire “Biométrie et Biologie Evolutive”- UMR 5558, CNRS / Université Lyon 1, Villeurbanne, France
| | - Jean-Michel Gaillard
- Laboratoire “Biométrie et Biologie Evolutive”- UMR 5558, CNRS / Université Lyon 1, Villeurbanne, France
| | - Cristina Vieira
- Laboratoire “Biométrie et Biologie Evolutive”- UMR 5558, CNRS / Université Lyon 1, Villeurbanne, France
| | - Ingrid Plotton
- Service d’Endocrinologie Moléculaire et Maladies Rares, Hospices Civils de Lyon, Lyon, France
| | - Damien Sanlaville
- Service de Génétique, Hospices Civils de Lyon, CRNL, GENDEV team, INSERM U1028, CNRS UMR5292, Université Lyon 1, Lyon, France
| | - François Gueyffier
- Laboratoire “Biométrie et Biologie Evolutive”- UMR 5558, CNRS / Université Lyon 1, Villeurbanne, France
| | - Jean-Francois Lemaitre
- Laboratoire “Biométrie et Biologie Evolutive”- UMR 5558, CNRS / Université Lyon 1, Villeurbanne, France
| |
Collapse
|
20
|
Miragem AA, Homem de Bittencourt PI. Nitric oxide-heat shock protein axis in menopausal hot flushes: neglected metabolic issues of chronic inflammatory diseases associated with deranged heat shock response. Hum Reprod Update 2018; 23:600-628. [PMID: 28903474 DOI: 10.1093/humupd/dmx020] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.
Collapse
Affiliation(s)
- Antônio Azambuja Miragem
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil.,Federal Institute of Education, Science and Technology 'Farroupilha', Rua Uruguai 1675, Santa Rosa, RS 98900-000, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|
21
|
Litvinov AA, Volotova EV, Kurkin DV, Logvinova EO, Darmanyan AP, Tyurenkov IN. [Focal cerebral ischemia in rats with estrogen deficiency and endothelial dysfunction]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:56-62. [PMID: 28980582 DOI: 10.17116/jnevro20171178256-62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM To assess an effect of ovariectomy (OE) on the cerebral blood flow, endothelium-dependent vasodilation, neurological, cognitive and locomotor deficit as markers of brain damage after focal ischemia in rats. MATERIAL AND METHODS The study was conducted in 48 female Wistar rats. Ovariectomy was performed with ovaries and uterine body extirpation, cerebral ischemia was performed by middle cerebral artery occlusion (MCAO) in rats. To assess brain damage, Combs and Garcia scores, 'open field' test (OFT), 'extrapolatory escape test' (EET), 'passive avoidance test' (PAT), 'beam-walking test' were used. Cerebral blood flow was measured using ultrasonic flowmetry. RESULTS AND CONCLUSION After 7 days of MCAO, the cerebral blood flow in ovarioectomized animals was reduced by 20% compared to sham-ovariectomized animals. Ovariectomized animals with MCAO showed a three-fold endothelium-dependent vasodilation reduction (the reaction of cerebral vessels to the introduction of acetylcholine and N-L-arginine), indicating the presence of severe endothelial dysfunction. In ovarioectomized animals, the cerebral blood flow was reduced by 34% compared to sham-operated animals. MCAO and OE taken together resulted in more than 2-fold increase in neurological, motor disturbances, 3-fold decrease in motor activity of the animals in the OP test. Focal ischemia in ovarioectomized animals with endothelial dysfunction led to memory decrease by 1/5 fold in PAT and by 2-fold in EET.
Collapse
Affiliation(s)
- A A Litvinov
- Volgograd State Medical university, Volgograd, Russia
| | - E V Volotova
- Volgograd State Medical university, Volgograd, Russia
| | - D V Kurkin
- Volgograd State Medical university, Volgograd, Russia
| | - E O Logvinova
- Volgograd State Medical university, Volgograd, Russia
| | - A P Darmanyan
- Volgograd State Agrarian university, Volgograd, Russia
| | - I N Tyurenkov
- Volgograd State Medical university, Volgograd, Russia
| |
Collapse
|
22
|
Morselli E, Santos RS, Criollo A, Nelson MD, Palmer BF, Clegg DJ. The effects of oestrogens and their receptors on cardiometabolic health. Nat Rev Endocrinol 2017; 13:352-364. [PMID: 28304393 DOI: 10.1038/nrendo.2017.12] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of mortality in developed countries. The incidence of CVD is sexually dimorphic, and research has focused on the contribution of sex steroids to the development and progression of the cardiometabolic syndrome, which is defined as a clustering of interrelated risk factors that promote the development of atherosclerosis (which can lead to CVD) and type 2 diabetes mellitus. Data are inconclusive as to how sex steroids and their respective receptors increase or suppress the risk of developing the cardiometabolic syndrome and thus CVD. In this Review, we discuss the potential role, or roles, of sex hormones in cardiometabolic health by first focusing on the influence of oestrogens and their receptors on the risk of developing cardiometabolic syndrome and CVD. We also highlight what is known about testosterone and its potential role in protecting against the development of the cardiometabolic syndrome and CVD. Given the inconclusive nature of the data regarding the direct effects of each sex hormone, we advocate and highlight the importance of studying the relative levels and the ratio of sex hormones to each other, as well as the use of cross sex hormone therapy and its effect on cardiometabolic health.
Collapse
Affiliation(s)
- Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Roberta S Santos
- Obesity and Comorbidities Research Center, Institute of Biology, State University of Campinas, Campinas 1, 3083-864, Brazil
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research, Los Angeles, California 90048, USA
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Santiago 8380000, Chile
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago 8380492, Chile
| | - Michael D Nelson
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, University of Texas at Arlington, Texas 76019, USA
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Deborah J Clegg
- Cedars-Sinai Diabetes and Obesity Research Institute, Department of Biomedical Research, Los Angeles, California 90048, USA
| |
Collapse
|
23
|
Abstract
Sex differences in longevity can provide insights into novel mechanisms of aging, yet they have been little studied. Surprisingly, sex-specific longevity patterns are best known in wild animals. Evolutionary hypotheses accounting for longevity patterns in natural populations include differential vulnerability to environmental hazards, differential intensity of sexual selection, and distinct patterns of parental care. Mechanistic hypotheses focus on hormones, asymmetric inheritance of sex chromosomes and mitochondria. Virtually all intensively studied species show conditional sex differences in longevity. Humans are the only species in which one sex is known to have a ubiquitous survival advantage. Paradoxically, although women live longer, they suffer greater morbidity particularly late in life. This mortality-morbidity paradox may be a consequence of greater connective tissue responsiveness to sex hormones in women. Human females' longevity advantage may result from hormonal influences on inflammatory and immunological responses, or greater resistance to oxidative damage; current support for these mechanisms is weak.
Collapse
Affiliation(s)
- Steven N Austad
- Department of Biology and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294-1170, USA.
| | - Kathleen E Fischer
- Department of Biology and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294-1170, USA
| |
Collapse
|
24
|
Zhong Q, Xi S, Liang J, Shi G, Huang Y, Zhang Y, Levy D, Zhong S. The significance of Brf1 overexpression in human hepatocellular carcinoma. Oncotarget 2016; 7:6243-54. [PMID: 26701855 PMCID: PMC4868753 DOI: 10.18632/oncotarget.6668] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/07/2015] [Indexed: 02/05/2023] Open
Abstract
Brf1 (TFIIB-related factor 1) plays a crucial role in cell transformation and tumorigenesis. However, the significance of Brf1 expression in human HCC (hepatocellular carcinoma) cases remains to be addressed. In this study, biopsies of human HCC, liver tumor samples of mice and cell lines of normal and tumor liver were utilized to determine the alteration of Brf1 expression using cytological and molecular biological approaches. Brf1 expression is increased in human HCC cases, which is correlated with shorter survival times. Levels of Brf1 and Pol III (RNA polymerase III-dependent) gene transcription in HCC patients with alcohol consumption are higher than the cases of non-HCC with or without alcohol intake. Induction of Brf1 and Pol III genes by ethanol in hepatoma cells is higher than in non-tumor cells. Ethanol increases the rate of cell transformation. Repression of Brf1 inhibits alcohol-promoted cell transformation. Alcohol consumption enhances Brf1 expression to promote cell transformation. These studies demonstrate that Brf1 is a new biomarker of HCC.
Collapse
Affiliation(s)
- Qian Zhong
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shaoyan Xi
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianzhong Liang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ganggang Shi
- Shantou University Medical College, Shantou, Guangdong, China
| | - Yi Huang
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yanmei Zhang
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel Levy
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shuping Zhong
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Curcumin Supplementation Decreases Intestinal Adiposity Accumulation, Serum Cholesterol Alterations, and Oxidative Stress in Ovariectomized Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5719291. [PMID: 26640615 PMCID: PMC4658407 DOI: 10.1155/2016/5719291] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/21/2015] [Accepted: 01/29/2015] [Indexed: 12/17/2022]
Abstract
The aim of this study was to investigate the potential of curcumin oral supplementation (50 and 100 mg/Kg/day, for 30 days) in circumventing menopause-associated oxidative stress and lipid profile dysfunctions in a rat ovariectomy (OVX) model. Female Wistar rats were operated and randomly divided into either sham-operated or OVX groups. Sham-operated group (n = 8) and one OVX group (n = 11) were treated with vehicle (refined olive oil), and the other two OVX groups received curcumin at 50 or 100 mg/Kg/day doses (n = 8/group). OVX vehicle-treated animals presented a higher deposition of intestinal adipose tissue as well as increased serum levels of IL-6, LDL, and total cholesterol when compared to sham-operated rats. In addition, several oxidative stress markers in serum, blood, and liver (such as TBARS, carbonyl, reduced-sulphydryl, and nonenzymatic antioxidant defenses) were altered toward a prooxidant status by OVX. Interestingly, curcumin supplementation attenuated most of these parameters to sham comparable values. Thus, the herein presented results show that curcumin may be useful to ameliorate lipid metabolism alterations and oxidative damage associated with hormone deprivation in menopause.
Collapse
|
26
|
Wang T, Xu M, Xu Y, Lu J, Li M, Chen Y, Wang W, Lai S, Bi Y, Ning G. Association of Bisphenol A Exposure With Hypertension and Early Macrovascular Diseases in Chinese Adults: A Cross-Sectional Study. Medicine (Baltimore) 2015; 94:e1814. [PMID: 26512580 PMCID: PMC4985394 DOI: 10.1097/md.0000000000001814] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Bisphenol A (BPA) is an environmental endocrine disruptor that has been associated with cardiovascular outcomes in previous observational studies. We aimed to examine the relationships of urinary BPA levels with hypertension and early macrovascular diseases.This is a cross-sectional study. From June through August 2009, 3246 participants ages 40 years or older were enrolled from Baoshan District, Shanghai, China. Logistic regression modes were used to estimate the odds ratios (ORs) for prevalent risk of hypertension, elevated carotid intima-media thickness (CIMT), arterial stiffness, and peripheral artery disease (PAD) with multivariable adjustment. We also performed stratification analysis by age and sex.The median (interquartile range) for BPA was 0.81 (0.48, 1.45) ng/mL, which is notably lower than previously reported in the United States and other Western countries. Urinary BPA concentrations were negatively associated with hypertension (multivariable-adjusted OR for the highest versus lowest BPA quartile = 0.61; 95% confidence interval [CI]: 0.46, 0.80), elevated CIMT (OR = 0.64; 95% CI: 0.47, 0.87), and arterial stiffness (OR = 0.62; 95% CI: 0.44, 0.87). The corresponding OR for PAD (60 cases total) was not significant (OR = 0.89; 95% CI: 0.28, 2.80). The negative associations of BPA with hypertension, elevated CIMT, and arterial stiffness were consistent by age and sex stratifications, and were stronger among participants ≥60 versus <60 years of age, and among women than men.In contrast with previous investigations, our study suggests negative associations of BPA exposure with hypertension and early macrovascular diseases among middle-aged and elderly Chinese. Future investigations are needed to draw more definite conclusions and generalize to other populations.
Collapse
Affiliation(s)
- Tiange Wang
- From the State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, the National Clinical Research Center for Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (TW, MX, YX, JL, ML, YC, WW, YB, GN) and Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA (SL)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Endogenous Estrogen-Mediated Heme Oxygenase Regulation in Experimental Menopause. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:429713. [PMID: 26064421 PMCID: PMC4438186 DOI: 10.1155/2015/429713] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/04/2014] [Indexed: 12/27/2022]
Abstract
Estrogen deficiency is one of the main causes of age-associated diseases in the cardiovascular system. Female Wistar rats were divided into four experimental groups: pharmacologically ovariectomized, surgically ovariectomized, and 24-month-old intact aging animals were compared with a control group. The activity and expression of heme oxygenases (HO) in the cardiac left ventricle, the concentrations of cardiac interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), the myeloperoxidase (MPO) activity in the cardiac left ventricle, and the effects of heme oxygenase blockade (by 24-hour and 1-hour pretreatment with tin-protoporphyrin IX, SnPP) on the epinephrine and phentolamine-induced electrocardiogram ST segment changes in vivo were investigated. The cardiac HO activity and the expression of HO-1 and HO-2 were significantly decreased in the aged rats and after ovariectomy. Estrogen depletion was accompanied by significant increases in the expression of IL-6 and TNF-α. The aged and ovariectomized animals exhibited a significantly elevated MPO activity and a significant ST segment depression. After pretreatment with SnPP augmented ST segment changes were determined. These findings demonstrate that the sensitivity to cardiac ischemia in estrogen depletion models is associated with suppression of the activity and expression of the HO system and increases in the secretion of proinflammatory cytokines and biomarkers.
Collapse
|
28
|
Wang N, Sun LY, Zhang SC, Wei R, Xie F, Liu J, Yan Y, Duan MJ, Sun LL, Sun YH, Niu HF, Zhang R, Ai J. MicroRNA-23a participates in estrogen deficiency induced gap junction remodeling of rats by targeting GJA1. Int J Biol Sci 2015; 11:390-403. [PMID: 25798059 PMCID: PMC4366638 DOI: 10.7150/ijbs.10930] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/21/2015] [Indexed: 01/07/2023] Open
Abstract
Increased incidence of arrhythmias in women after menopause has been widely documented, which is considered to be related to estrogen (E2) deficiency induced cardiac electrophysiological abnormalities. However, its molecular mechanism remains incompletely clear. In the present study, we found cardiac conduction blockage in post-menopausal rats. Thereafter, the results showed that cardiac gap junctions were impaired and Connexin43 (Cx43) expression was reduced in the myocardium of post-menopausal rats. The phenomenon was also observed in ovariectomized (OVX) rats, which was attenuated by E2 supplement. Further study displayed that microRNA-23a (miR-23a) level was significantly increased in both post-menopausal and OVX rats, which was reversed by daily E2 treatment after OVX. Importantly, forced overexpression of miR-23a led to gap junction impairment and Cx43 downregulation in cultured cardiomyocytes, which was rescued by suppressing miR-23a by transfection of miR-23a specific inhibitory oligonucleotide (AMO-23a). GJA1 was identified as the target gene of miR-23a by luciferase assay and miRNA-masking antisense ODN (miR-Mask) assay. We also found that E2 supplement could reverse cardiac conduction blockage, Cx43 downregulation, gap junction remodeling and miR-23a upregulation in post-menopausal rats. These findings provide the evidence that miR-23a mediated repression of Cx43 participates in estrogen deficiency induced damages of cardiac gap junction, and highlights a new insight into molecular mechanism of post-menopause related arrhythmia at the microRNA level.
Collapse
Affiliation(s)
- Ning Wang
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Lu-Yao Sun
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Shou-Chen Zhang
- 3. Electron Microscopy Center, Harbin Medical University, Harbin, People's Republic of China, 150081
| | - Ran Wei
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Fang Xie
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081 ; 2. Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, People's Republic of China, 150081
| | - Jing Liu
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Yan Yan
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Ming-Jing Duan
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Lin-Lin Sun
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Ying-Hui Sun
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Hui-Fang Niu
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Rong Zhang
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| | - Jing Ai
- 1. Department of Pharmacology, Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, People's Republic of China, 150081
| |
Collapse
|
29
|
Efficacy of female rat models in translational cardiovascular aging research. J Aging Res 2014; 2014:153127. [PMID: 25610649 PMCID: PMC4294461 DOI: 10.1155/2014/153127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in women in the United States. Aging is a primary risk factor for the development of cardiovascular disease as well as cardiovascular-related morbidity and mortality. Aging is a universal process that all humans undergo; however, research in aging is limited by cost and time constraints. Therefore, most research in aging has been done in primates and rodents; however it is unknown how well the effects of aging in rat models translate into humans. To compound the complication of aging gender has also been indicated as a risk factor for various cardiovascular diseases. This review addresses the systemic pathophysiology of the cardiovascular system associated with aging and gender for aging research with regard to the applicability of rat derived data for translational application to human aging.
Collapse
|
30
|
Retarding the senescence of human vascular endothelial cells induced by hydrogen peroxide: effects of 17beta-estradiol (E2) mediated mitochondria protection. Biogerontology 2014; 15:367-75. [DOI: 10.1007/s10522-014-9507-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/30/2014] [Indexed: 01/26/2023]
|
31
|
Li R, Cui J, Shen Y. Brain sex matters: estrogen in cognition and Alzheimer's disease. Mol Cell Endocrinol 2014; 389:13-21. [PMID: 24418360 PMCID: PMC4040318 DOI: 10.1016/j.mce.2013.12.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 12/31/2013] [Accepted: 12/31/2013] [Indexed: 02/06/2023]
Abstract
Estrogens are the primary female sex hormones and play important roles in both reproductive and non-reproductive systems. Estrogens can be synthesized in non-reproductive tissues such as liver, heart, muscle, bone and the brain. During the past decade, increasing evidence suggests that brain estrogen can not only be synthesized by neurons, but also by astrocytes. Brain estrogen also works locally at the site of synthesis in paracrine and/or intracrine fashion to maintain important tissue-specific functions. Here, we will focus on the biology of brain estrogen and its impact on cognitive function and Alzheimer's disease. This comprehensive review provides new insights into brain estrogens by presenting a better understanding of the tissue-specific estrogen effects and their roles in healthy ageing and cognitive function.
Collapse
Affiliation(s)
- Rena Li
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, United States.
| | - Jie Cui
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, United States
| | - Yong Shen
- Center for Advanced Therapeutic Strategies for Brain Disorders (CATSBD), Roskamp Institute, Sarasota, FL 34243, United States
| |
Collapse
|
32
|
Baxter MG, Roberts MT, Gee NA, Lasley BL, Morrison JH, Rapp PR. Multiple clinically relevant hormone therapy regimens fail to improve cognitive function in aged ovariectomized rhesus monkeys. Neurobiol Aging 2013; 34:1882-90. [PMID: 23369546 PMCID: PMC3622837 DOI: 10.1016/j.neurobiolaging.2012.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 12/10/2012] [Accepted: 12/22/2012] [Indexed: 10/27/2022]
Abstract
Preclinical studies in aged, surgically-menopausal rhesus monkeys have revealed powerful benefits of intermittent estrogen injections on prefrontal cortex-dependent working memory, together with corresponding effects on dendritic spine morphology in the prefrontal cortex. This contrasts with the inconsistent effects of hormone therapy (HT) reported in clinical studies in women. Factors contributing to this discrepancy could include differences in the formulation and sequence of HT regimens, resulting in different neurobiological outcomes. The current study evaluated, in aging surgically menopausal rhesus monkeys, the cognitive effects of 4 HT regimens modeled directly on human clinical practice, including continuous estrogen treatment opposed by progesterone. None of the regimens tested produced any cognitive effect, despite yielding physiologically relevant serum hormone levels, as intended. These findings have implications for the design of regimens that might optimize the benefits of hormone treatment for healthy aging, and suggest that common HT protocols used by women may fail to result in substantial cognitive benefit, at least via direct effects on the prefrontal cortex.
Collapse
Affiliation(s)
- Mark G Baxter
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Hypoxia-inducible factor (HIF) is a set of transcription factors that regulate the cellular response to hypoxia. There is a great body of evidence supporting the protective role of HIF-1α in cardiovascular pathophysiology, however, newer studies are hinting at a maladaptive and deleterious role of this transcription factor that merits further investigation. There is a general agreement, however, that HIF-mediated responses appear to differ under conditions of acute and chronic oxygen deprivation. The intensity and sustainability of HIF-1α activation are major determinants of whether the responses are pathological or beneficial. HIF activation is seen to be beneficial in the setting of acute myocardial ischemia and deleterious in chronic conditions. In this review, we will focus on recent insights into the role of HIF-1α in the heart and especially in the setting of ischemic heart disease.
Collapse
|
34
|
Fishpool SJC, Tomkinson A. Patterns of hospital admission with epistaxis for 26,725 patients over an 18-year period in Wales, UK. Ann R Coll Surg Engl 2013; 94:559-62. [PMID: 23131225 PMCID: PMC3954281 DOI: 10.1308/003588412x13373405386691] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Epistaxis is the one of the most common otorhinolaryngology emergencies. This study examined the age and sex distribution of all patients admitted with epistaxis to National Health Service (NHS) hospitals in Wales, UK, over a period of 18 years and 9 months. METHODS The Patient Episode Database for Wales was examined for all patient admissions with a diagnosis of epistaxis between April 1991 and December 2009. The age and sex of these patients was recorded and the proportion of the underlying population affected was calculated by comparing admission rates to the population data derived from the 1991 and 2001 national population censuses for Wales. RESULTS A total of 26,725 patients were admitted to NHS hospitals in Wales with epistaxis over the period studied. The proportion of the population admitted with epistaxis increased from the age of 40 onwards. For all ages except patients in the 10-14 years group, a higher proportion of the male population was admitted with epistaxis than the comparable female population. This discrepancy was most pronounced between the ages of 15 and 49 years, with the female-to-male ratio of hospital admissions with epistaxis being 0.55. These ages (15 and 49 years) approximate the average age of menarche and menopause respectively in the UK. CONCLUSIONS Women of menstrual age have fewer hospital admissions with epistaxis. This may be due to oestrogens providing protection to the nasal vasculature (as they do to other areas of the vascular tree).
Collapse
Affiliation(s)
- S J C Fishpool
- Cardiff and Vale University Health Board, UK, Department of Otolaryngology/Head and Neck Surgery, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK.
| | | |
Collapse
|
35
|
Kim DI, Choi MS, Pak SC, Lee SB, Jeon S. The effects of Sutaehwan-Gami on menopausal symptoms induced by ovariectomy in rats. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 12:227. [PMID: 23173704 PMCID: PMC3561075 DOI: 10.1186/1472-6882-12-227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 10/30/2012] [Indexed: 12/03/2022]
Abstract
Background This study was undertaken to evaluate the beneficial effects of a modified prescription of Sutaehwan named Sutaehwan-Gami (SG), created by adding Rhizoma dioscoreae and Carthami semen to Sutaehwan, on menopausal symptoms. Methods To evaluate the estrogenic effect of SG, we first examined estrogen receptor (ER) activation by SG treatment in breast adenocarcinoma cells and confirmed the estrogenic effect of SG in vivo ovariectomized rats. The animals were randomized into four groups: Sham operated group (Sham), saline treated ovariectomized group (OVX), SG treated group (SG) and raloxifene treated group (RLX). Animals were provided with SG at a dose of 500 mg/kg bw/day and RLX at a dose of 5.4 mg/kg bw/day with standard rat pellets for 3 months. Results SG significantly increased ERα phosphorylation, and its downstream effectors, extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) phosphorylation in breast adenocarcinoma cells. Treatment with SG reversed ovariectomy-induced uterine weight reduction and weight gain. Decreases in the levels of GOT and GPT were observed in the SG group. The significantly reduced E2β level in OVX rats was raised by treatment with SG. Moreover, SG significantly increased the phosphorylation levels of ERK and Akt in the uterus. Conclusion Taken together, these data indicate that SG has phytoestrogen-like properties through ERK and Akt activation, implying that it could be protective and beneficial for the management of menopausal symptoms.
Collapse
|
36
|
Foxa1 and Foxa2 are essential for sexual dimorphism in liver cancer. Cell 2012; 148:72-83. [PMID: 22265403 DOI: 10.1016/j.cell.2011.11.026] [Citation(s) in RCA: 310] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2011] [Revised: 08/26/2011] [Accepted: 11/07/2011] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is sexually dimorphic in both rodents and humans, with significantly higher incidence in males, an effect that is dependent on sex hormones. The molecular mechanisms by which estrogens prevent and androgens promote liver cancer remain unclear. Here, we discover that sexually dimorphic HCC is completely reversed in Foxa1- and Foxa2-deficient mice after diethylnitrosamine-induced hepatocarcinogenesis. Coregulation of target genes by Foxa1/a2 and either the estrogen receptor (ERα) or the androgen receptor (AR) was increased during hepatocarcinogenesis in normal female or male mice, respectively, but was lost in Foxa1/2-deficient mice. Thus, both estrogen-dependent resistance to and androgen-mediated facilitation of HCC depend on Foxa1/2. Strikingly, single nucleotide polymorphisms at FOXA2 binding sites reduce binding of both FOXA2 and ERα to their targets in human liver and correlate with HCC development in women. Thus, Foxa factors and their targets are central for the sexual dimorphism of HCC.
Collapse
|
37
|
Swindell WR, Johnston A, Sun L, Xing X, Fisher GJ, Bulyk ML, Elder JT, Gudjonsson JE. Meta-profiles of gene expression during aging: limited similarities between mouse and human and an unexpectedly decreased inflammatory signature. PLoS One 2012; 7:e33204. [PMID: 22413003 PMCID: PMC3296693 DOI: 10.1371/journal.pone.0033204] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 02/13/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Skin aging is associated with intrinsic processes that compromise the structure of the extracellular matrix while promoting loss of functional and regenerative capacity. These processes are accompanied by a large-scale shift in gene expression, but underlying mechanisms are not understood and conservation of these mechanisms between humans and mice is uncertain. RESULTS We used genome-wide expression profiling to investigate the aging skin transcriptome. In humans, age-related shifts in gene expression were sex-specific. In females, aging increased expression of transcripts associated with T-cells, B-cells and dendritic cells, and decreased expression of genes in regions with elevated Zeb1, AP-2 and YY1 motif density. In males, however, these effects were contrasting or absent. When age-associated gene expression patterns in human skin were compared to those in tail skin from CB6F1 mice, overall human-mouse correspondence was weak. Moreover, inflammatory gene expression patterns were not induced with aging of mouse tail skin, and well-known aging biomarkers were in fact decreased (e.g., Clec7a, Lyz1 and Lyz2). These unexpected patterns and weak human-mouse correspondence may be due to decreased abundance of antigen presenting cells in mouse tail skin with age. CONCLUSIONS Aging is generally associated with a pro-inflammatory state, but we have identified an exception to this pattern with aging of CB6F1 mouse tail skin. Aging therefore does not uniformly heighten inflammatory status across all mouse tissues. Furthermore, we identified both intercellular and intracellular mechanisms of transcriptome aging, including those that are sex- and species-specific.
Collapse
Affiliation(s)
- William R Swindell
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Pechenino AS, Lee AR, Lin L, Mbai FN, Stallone JN, Knowlton AA. Reply to “Letter to the Editor: ‘Understanding the WHI gap’”. Physiol Genomics 2012. [DOI: 10.1152/physiolgenomics.00188.2011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - Alison R. Lee
- Cellular and Molecular Cardiology, Department of Medicine,
| | - Li Lin
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Fiona N. Mbai
- Cellular and Molecular Cardiology, Department of Medicine,
- Institute of Tropical Medicine and Infectious Diseases, University of Nairobi, Nairobi, Kenya; and
| | - John N. Stallone
- Department of Physiology, and Pharmacology, School of Veterinary Medicine, Texas A&M, College Station, Texas
| | - A. A. Knowlton
- Cellular and Molecular Cardiology, Department of Medicine,
- Department of Medical Pharmacology, University of California, Davis
- VA Northern California Health Care System, Mather, California
| |
Collapse
|
39
|
Etgen AM, Jover-Mengual T, Zukin RS. Neuroprotective actions of estradiol and novel estrogen analogs in ischemia: translational implications. Front Neuroendocrinol 2011; 32:336-52. [PMID: 21163293 PMCID: PMC3080451 DOI: 10.1016/j.yfrne.2010.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/15/2022]
Abstract
This review highlights our investigations into the neuroprotective efficacy of estradiol and other estrogenic agents in a clinically relevant animal model of transient global ischemia, which causes selective, delayed death of hippocampal CA1 neurons and associated cognitive deficits. We find that estradiol rescues a significant number of CA1 pyramidal neurons that would otherwise die in response to global ischemia, and this is true when hormone is provided as a long-term pretreatment at physiological doses or as an acute treatment at the time of reperfusion. In addition to enhancing neuronal survival, both forms of estradiol treatment induce measurable cognitive benefit in young animals. Moreover, estradiol and estrogen analogs that do not bind classical nuclear estrogen receptors retain their neuroprotective efficacy in middle-aged females deprived of ovarian hormones for a prolonged duration (8weeks). Thus, non-feminizing estrogens may represent a new therapeutic approach for treating the neuronal damage associated with global ischemia.
Collapse
Affiliation(s)
- Anne M Etgen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
40
|
Stice JP, Chen L, Kim SC, Jung JS, Tran AL, Liu TT, Knowlton AA. 17β-Estradiol, aging, inflammation, and the stress response in the female heart. Endocrinology 2011; 152:1589-98. [PMID: 21303943 PMCID: PMC3060632 DOI: 10.1210/en.2010-0627] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 12/29/2010] [Indexed: 01/21/2023]
Abstract
Heat shock proteins (HSPs) are a cardioprotective class of proteins induced by stress and regulated by the transcription factor, heat shock factor (HSF)-1. 17β-estradiol (E(2)) indirectly regulates HSP expression through rapid activation of nuclear factor-κB (NF-κB) and HSF-1 and protects against hypoxia. As males experience a loss of protective cellular responses in aging, we hypothesized that aged menopausal (old ovariectomized) rats would have an impaired HSP response, which could be prevented by immediate in vivo E(2) replacement. After measuring cardiac function in vivo, cardiac myocytes were isolated from ovariectomized adult and old rats with and without 9 weeks of E(2) replacement. Myocytes were treated with E(2) in vitro and analyzed for activation of NF-κB, HSF-1, and HSP expression. In addition, we measured inflammatory cytokine expression and susceptibility to hypoxia/reoxygenation injury. Cardiac contractility was reduced in old ovariectomized rats and could prevented by immediate E(2) replacement in vivo. Subsequent investigations in isolated cardiac myocytes found that in vitro E(2) activated NF-κB, HSF-1, and increased HSP 72 expression in adult but not old rats. In response to hypoxia/reoxygenation, myocytes from adult, but not old, rats had increased HSP 72 expression. In addition, expression of the inflammatory cytokines TNF-α and IL-1β, as well as oxidative stress, were increased in myocytes from old ovariectomized rats; only the change in cytokine expression could be attenuated by in vivo E(2) replacement. This study demonstrates that while aging in female rats led to a loss of the cardioprotective HSP response, E(2) retains its protective cellular properties.
Collapse
Affiliation(s)
- James P Stice
- Molecular and Cellular Cardiology, Genomics and Biomedical Sciences Facility, Room 6317, University of California, Davis, 451 Health Sciences Way, Davis, California 95616, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Lin J, Kroenke CH, Epel E, Kenna HA, Wolkowitz OM, Blackburn E, Rasgon NL. Greater endogenous estrogen exposure is associated with longer telomeres in postmenopausal women at risk for cognitive decline. Brain Res 2011; 1379:224-31. [PMID: 20965155 PMCID: PMC3057451 DOI: 10.1016/j.brainres.2010.10.033] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 10/07/2010] [Accepted: 10/11/2010] [Indexed: 01/01/2023]
Abstract
Longer duration of reproductive years of life and thus greater exposure to endogenous estrogen may be associated with a lower risk of age-related diseases in women. The present study examined the relationship between estimated endogenous estrogen exposure and telomere length (TL) and telomerase activity, two biomarkers of cellular aging, in a sample of postmenopausal women at risk for cognitive decline. Telomere length was measured using a quantitative PCR method and telomerase activity by TRAP (Telomere-Repeats Amplification Protocol) assay in peripheral blood mononuclear cells (PBMCs). Study subjects were 53 postmenopausal women (35 with natural and 18 with surgical menopause) receiving hormone therapy (HT) for at least one year or longer. Length of reproductive years of life, computed as the difference between age at menopause and age at menarche, was used as a proxy of duration of exposure to endogenous estrogen. Length of time on HT was the measure used for duration of exogenous estrogen exposure. We found that longer endogenous estrogen exposure was associated with greater TL (standardized β=0.06, Wald χ(2)=3.7, p=0.04) and with lower telomerase activity (standardized β=-0.09, Wald χ(2)=5.0, p=0.03). Length of reproductive years was also inversely associated with the combination of short TL and high telomerase (OR=0.78, 95% CI: 0.63, 0.97, p=0.02). Length of HT use was not associated with TL or telomerase activity in this study. The results suggest that the endogenous estrogens may be associated with deceleration of cellular aging. This is the first study to examine associations between endogenous estrogens, telomere length and telomerase activity.
Collapse
Affiliation(s)
- Jue Lin
- Department of Biochemistry and Biophysics, University of California, San Francisco
| | - Candyce H. Kroenke
- Center for Health and Community, Department of Psychiatry, University of California, San Francisco
| | - Elissa Epel
- Center for Health and Community, Department of Psychiatry, University of California, San Francisco
| | - Heather A. Kenna
- Stanford Center for Neuroscience in Women’s Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, United States
| | | | - Elizabeth Blackburn
- Department of Biochemistry and Biophysics, University of California, San Francisco
| | - Natalie L. Rasgon
- Stanford Center for Neuroscience in Women’s Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, United States
| |
Collapse
|
42
|
Recchia AG, De Francesco EM, Vivacqua A, Sisci D, Panno ML, Andò S, Maggiolini M. The G protein-coupled receptor 30 is up-regulated by hypoxia-inducible factor-1alpha (HIF-1alpha) in breast cancer cells and cardiomyocytes. J Biol Chem 2011; 286:10773-82. [PMID: 21266576 DOI: 10.1074/jbc.m110.172247] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
GPR30, also known as GPER, has been suggested to mediate rapid effects induced by estrogens in diverse normal and cancer tissues. Hypoxia is a common feature of solid tumors involved in apoptosis, cell survival, and proliferation. The response to low oxygen environment is mainly mediated by the hypoxia-inducible factor named HIF-1α, which activates signaling pathways leading to adaptive mechanisms in tumor cells. Here, we demonstrate that the hypoxia induces HIF-1α expression, which in turn mediates the up-regulation of GPER and its downstream target CTGF in estrogen receptor-negative SkBr3 breast cancer cells and in HL-1 cardiomyocytes. Moreover, we show that HIF-1α-responsive elements located within the promoter region of GPER are involved in hypoxia-dependent transcription of GPER, which requires the ROS-induced activation of EGFR/ERK signaling in both SkBr3 and HL-1 and cells. Interestingly, the apoptotic response to hypoxia was prevented by estrogens through GPER in SkBr3 cells. Taken together, our data suggest that the hypoxia-induced expression of GPER may be included among the mechanisms involved in the anti-apoptotic effects elicited by estrogens, particularly in a low oxygen microenvironment.
Collapse
Affiliation(s)
- Anna Grazia Recchia
- Department of Pharmaco-Biology, University of Calabria, 87030 Rende (Cosenza), Italy
| | | | | | | | | | | | | |
Collapse
|
43
|
Behr GA, Schnorr CE, Moreira JCF. Increased blood oxidative stress in experimental menopause rat model: the effects of vitamin A low-dose supplementation upon antioxidant status in bilateral ovariectomized rats. Fundam Clin Pharmacol 2011; 26:235-49. [PMID: 21226757 DOI: 10.1111/j.1472-8206.2010.00923.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Menopause has been reported to be associated with increased oxidative stress and metabolic disorders among women worldwide. Disarrangements in the redox state similar to those observed in women during the decline of ovarian hormonal activity can be obtained experimentally through rat bilateral ovariectomy. The search for alternative treatments to improve life quality in postmenopausal woman is really important. The aim of this study was to evaluate biochemical and oxidative stress parameters that distinguish sham-operated female rats from Wistar rats bilaterally ovariectomized (OVX). Additionally, we have also investigated the effects of retinol palmitate (a vitamin A supplement) low-dose supplementation (500 or 1500 IU/kg/day, during 30 days) upon blood and plasma antioxidant status in OVX rats. Ovariectomy caused an increase in body weight gain, pronounced uterine atrophy, decreased plasma triglycerides and increased total cholesterol levels, and reduced acid uric content. Moreover, we found increased blood peroxidase activities (catalase and glutathione peroxidase), decreased plasma non-enzymatic antioxidant defenses total reactive antioxidant potential and total antioxidant reactivity, decreased protein and non-protein SH levels, accompanied by increased protein oxidative damage (carbonyl). In addition, vitamin A low-dose supplementation was capable to ameliorate antioxidant status in OVX rats, restoring both enzymatic and non-enzymatic defenses, promoting reduction in plasma SH content, and decreasing protein oxidative damage levels. This is the first work in the literature showing that vitamin A at low dose may be beneficial in the treatment of menopause symptoms. Further studies will be made to better understand the effects of vitamin A supplementation in menopause rat model.
Collapse
Affiliation(s)
- Guilherme Antônio Behr
- Center of Oxidative Stress Research, Professor Tuiskon Dick Department of Biochemistry, Institute of Health Basic Sciences, Federal University of Rio Grande do Sul, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil.
| | | | | |
Collapse
|
44
|
Habauzit D, Flouriot G, Pakdel F, Saligaut C. Effects of estrogens and endocrine-disrupting chemicals on cell differentiation-survival-proliferation in brain: contributions of neuronal cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2011; 14:300-327. [PMID: 21790314 DOI: 10.1080/10937404.2011.578554] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Estrogens and estrogen receptors (ER) are key actors in the control of differentiation and survival and act on extrareproductive tissues such as brain. Thus, estrogens may display neuritogenic effects during development and neuroprotective effects in the pathophysiological context of brain ischemia and neurodegenerative pathologies like Alzheimer's disease or Parkinson's disease. Some of these effects require classical transcriptional "genomic" mechanisms through ER, whereas other effects appear to rely clearly on "membrane-initiated mechanisms" through cytoplasmic signal transduction pathways. Disturbances of these mechanisms by endocrine-disrupting chemicals (EDC) may exert adverse effects on brain. Some EDC may act via ER-independent mechanisms but might cross-react with endogenous estrogen. Other EDC may act through ER-dependent mechanisms and display agonistic/antagonistic estrogenic properties. Because of these potential effects of EDC, it is necessary to establish sensitive cell-based assays to determine EDC effects on brain. In the present review, some effects of estrogens and EDC are described with focus on ER-mediated effects in neuronal cells. Particular attention is given to PC12 cells, an interesting model to study the mechanisms underlying ER-mediated differentiating and neuroprotective effects of estrogens.
Collapse
Affiliation(s)
- Denis Habauzit
- UMR CNRS 6026 (Interactions Cellulaires et Moléculaires, Equipe RED), Université de Rennes 1, Rennes, France
| | | | | | | |
Collapse
|
45
|
Ovariectomy in aged versus young rats augments matrix metalloproteinase-mediated vasoconstriction in mesenteric arteries. Menopause 2010; 17:516-23. [PMID: 20142791 DOI: 10.1097/gme.0b013e3181c91f04] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Ovarian deficiency is known to undermine vasoprotective mechanisms and accelerate cardiovascular disease in postmenopausal women. In a rat model of menopause (aged ovariectomized [Ovx] rats), we recently revealed a vasoconstrictor pathway mediated by matrix metalloproteinases (MMPs) via cleavage of big endothelin-1 (ET-1). However, the specific impact of aging and/or Ovx on this pathway remains unknown. We hypothesized that aging exacerbates MMP-mediated vasoconstriction in an ovary-deficient state. METHODS Young and aged female Sprague-Dawley rats, either intact or Ovx, were assessed for MMP-dependent vasoreactivity. Dose responses to big ET-1 in the absence or presence of an MMP inhibitor (GM6001) were tested on small mesenteric arteries using a pressure myograph system. MMP levels in the vascular tissue were measured by gelatin zymography. RESULTS Both young Ovx and aged Ovx animals demonstrated a similar increase in the vasoconstriction to big ET-1 compared with the age-matched intact groups. MMP inhibition attenuated big ET-1 response in both Ovx groups and aged controls, but this effect was more pronounced in aged Ovx arteries (area under the curve reduction, 3.8 +/- 0.6 units in aged Ovx rats vs 1.5 +/- 0.5 units in young Ovx rats or 1.8 +/- 0.6 units in aged intact rats; P < 0.05). MMP-2 activity in the vascular tissue increased with age and was further augmented by Ovx. CONCLUSIONS Regardless of age, ovarian loss increases vascular reactivity to big ET-1, which is mediated, in part, by MMP. Superimposed with advancing age, ovarian deficiency further increases the proconstrictor role of MMP, which corresponds with higher MMP-2 levels in the aging vessel wall. MMP-mediated vasoconstriction may be a mechanism contributing to vascular dysfunction in postmenopausal women.
Collapse
|
46
|
Chen L, Knowlton AA. Mitochondria and heart failure: new insights into an energetic problem. Minerva Cardioangiol 2010; 58:213-229. [PMID: 20440251 PMCID: PMC3786553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Cardiac mitochondria are powerful organelles supplying energy to support the high adenosine triphosphate (ATP) consumption of the beating heart. The progression of HF (HF) is characterized by diminished energy metabolism, calcium mishandling, reactive oxygen species (ROS) generation and apoptotic cell death. Although the etiologies of HF are multifactoral, many of the changes of HF are associated with cardiac mitochondrial dysfunction either directly or indirectly. A number of studies have established the role of calcium mishandling and reduced ATP production in mitochondrial dysfunction in HF. More recent work has contributed to our understanding of the role of ROS and proapoptotic protein release by the mitochondria in HF. New interest has been generated in mitochondria by the relatively recent identification of the processes of fusion and fission, which are critical to the maintenance of healthy mitochondria. Fission and fusion also have significant roles in apoptosis. Other studies have shown that estrogen has important functions in the mitochondria, including regulation of mitochondrial gene expression. Aging alone contributes to the development of HF through multiple mechanisms. These new insights into HF have implications for our understanding of this important disease, and will be reviewed here.
Collapse
Affiliation(s)
- L. Chen
- Molecular and Cellular Cardiology, University of California, Davis, Davis, CA, USA
| | - A. A. Knowlton
- Molecular and Cellular Cardiology, University of California, Davis, Davis, CA, USA
- V.A. Medical Center, Sacramento, CA, USA
| |
Collapse
|
47
|
Rasgon NL, Kenna HA, Wroolie TE, Kelley R, Silverman D, Brooks J, Williams KE, Powers BN, Hallmayer J, Reiss A. Insulin resistance and hippocampal volume in women at risk for Alzheimer's disease. Neurobiol Aging 2009; 32:1942-8. [PMID: 20031276 DOI: 10.1016/j.neurobiolaging.2009.12.005] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 09/14/2009] [Accepted: 12/08/2009] [Indexed: 11/27/2022]
Abstract
Insulin resistance (IR) is the main pathological condition underlying vascular disorders, such as diabetes and cardiovascular disease, which are well established risk factors for cognitive decline and Alzheimer disease (AD). Hippocampal atrophy has been associated with cognitive decline, but little is known about the influence of IR on hippocampus integrity in non-diabetic, cognitively intact individuals. Herein, 50 women ages 50-65, current users of hormone therapy, underwent magnetic resonance imaging, cognitive testing, and homeostatic assessment of insulin resistance (HOMA-IR), as part of a longitudinal study examining brain structure and function in postmenopausal women at risk for AD. Results demonstrated a significant negative relationship between HOMA-IR and right and total hippocampal volume, overall cognitive performance, and selective tests of verbal and non-verbal memory. The main effect of HOMA-IR on brain structure and cognition was not altered by the presence of APOE-ε4 allele or by reproductive history, such as duration of endogenous and exogenous estrogen exposure. These results suggest that IR in middle-aged individuals at risk for AD may be biomarker for dementia risk.
Collapse
Affiliation(s)
- Natalie L Rasgon
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stan-ford, CA 94305-5723, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Song JY, Kim MJ, Jo HH, Hwang SJ, Chae B, Chung JE, Kwon DJ, Lew YO, Lim YT, Kim JH, Kim JH, Kim MR. Antioxidant effect of estrogen on bovine aortic endothelial cells. J Steroid Biochem Mol Biol 2009; 117:74-80. [PMID: 19635556 DOI: 10.1016/j.jsbmb.2009.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Revised: 07/15/2009] [Accepted: 07/17/2009] [Indexed: 11/17/2022]
Abstract
OBJECTIVE This study discussed the role of estrogen as an antioxidant in the damage of vascular endothelial cells. DESIGN We treated bovine aortic endothelial cells (bAEC) either with 1mM of H(2)O(2) alone or with 1 microM of 17beta-estradiol (E(2)) for 24h followed by 1mM of H(2)O(2) for 3h. The cell survival was evaluated by MTT assay, cellular apoptosis by fluorescence activated cell sorter (FACS) and Hoechst 33342 staining, oxidative stress by intracellular reactive oxygen species (ROS) and apoptosis after oxidative stress by western blotting for phospho-p38, p38, and Bcl-2. RESULTS MTT assay showed that bAEC viability was reduced to 55.7+/-3.0% and 39.1+/-3.7% after 30 and 60 min of H(2)O(2) treatment, respectively. E(2) and H(2)O(2) treated cells did not show significant decrease in the cell survival. Similarly the FACS analysis and Hoechst 33342 stain showed that the latter decreased cellular apoptosis induced by H(2)O(2). Intracellular ROS increased by 181.6+/-68.9% in the former and by 37.0+/-3.9% in the latter (P<0.05). The expression of phospho-p38 mitogen-activated protein kinase (MAPK) was higher in the latter. CONCLUSIONS E(2) mediates antioxidant effects on the oxidative stress induced by H(2)O(2). This antioxidant effect on bAEC may elucidate the scientific basis of hormone therapy for maintaining cardiovascular integrity in postmenopausal women.
Collapse
Affiliation(s)
- Jae-Yen Song
- Department of Obstetrics and Gynecology, The Catholic University of Korea, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|