1
|
Lazaridis A, Malliora A, Gkaliagkousi E. The Particularities of Arterial Hypertension in Female Sex: From Pathophysiology to Therapeutic Management. J Clin Med 2025; 14:3137. [PMID: 40364167 PMCID: PMC12072934 DOI: 10.3390/jcm14093137] [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: 04/05/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Arterial hypertension is the most important modifiable cardiovascular risk factor and a major cause of cardiovascular mortality worldwide. In daily clinical practice, the hypertensive patient is often treated in a uniform way, thus ignoring the significant effects of sex on several aspects of hypertension, including its prevalence, pathophysiology, response to antihypertensive treatment, and outcomes. Along with the immune response and several cardiometabolic risk factors that frequently coexist, the substantial hormonal changes during a woman's life cycle are among the main pathophysiological mechanisms driving hypertension in women. Concurrently, women exhibit increased cardiovascular risk at lower blood pressure (BP) levels compared to age-matched men and present certain disparities in the incidence of cardiovascular events and subsequent hypertension-related cardiovascular prognosis. In addition, women respond differently to antihypertensive treatment, experience more drug-related side effects, and exhibit lower rates of BP control compared to men. Currently, international guidelines propose the same targets and the same therapeutic algorithms for the treatment of hypertension in both sexes without taking into account the sex differences that exist. In this review, we aim to describe certain particularities of arterial hypertension in the female sex, moving from pathophysiological aspects to clinical and therapeutical management.
Collapse
Affiliation(s)
- Antonios Lazaridis
- 1st Department of Internal Medicine, Papageorgiou General Hospital, 56429 Thessaloniki, Greece;
| | - Anastasia Malliora
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece;
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56403 Thessaloniki, Greece;
| |
Collapse
|
2
|
Satari S, Mota INR, Silva ACL, Brito HO, Oliveira PA, Gil da Costa RM, Medeiros R. Hallmarks of Cancer Cachexia: Sexual Dimorphism in Related Pathways. Int J Mol Sci 2025; 26:3952. [PMID: 40362192 PMCID: PMC12071346 DOI: 10.3390/ijms26093952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cancer-associated cachexia (CAC), also known as wasting syndrome, is a systemic condition that affects multiple tissues and organs via a variety of metabolic pathways. Systemic inflammation, progressive weight loss, depletion of adipose tissue, and skeletal muscle impairment are some of the hallmark features of cachexia. Despite various studies on the clinical features of CAC, the complexity of the syndrome continues to pose significant challenges in clinical practice, leading to late diagnoses and the absence of a standardised treatment. Men and women respond differently to CAC, which may be prompted by the pre-existing physiologic sex differences. This review presents the sexual dimorphism associated with the hallmark pathways involved in CAC. A comprehensive understanding of sexual dimorphism in these pathways could drive research on cachexia to prioritise the inclusion of more females in related studies in order to achieve personalised sex-based therapeutic approaches and, consequently, enhance treatment efficacy and better patient outcomes.
Collapse
Affiliation(s)
- Setareh Satari
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- The Institute of Public Health, University of Porto (ISPUP), Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Inês N. R. Mota
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Ana Carolina Leão Silva
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Haissa Oliveira Brito
- Research Center For Experimental and Clinical Physiology and Pharmacology (NEC)/Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)/Bioanalysis Lab (LaBIO), Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
| | - Rui Miguel Gil da Costa
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Research Center For Experimental and Clinical Physiology and Pharmacology (NEC)/Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)/Bioanalysis Lab (LaBIO), Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Biomedical Research Center, Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal
- ECO-European Cancer Organization, 1040 Brussels, Belgium
- Research Department of the Portuguese League Against Cancer—Regional Nucleus of the North (Liga Portuguesa Contra o Cancro—Núcleo Regional do Norte), 4200-172 Porto, Portugal
| |
Collapse
|
3
|
Wu Y, Xiang M, Zhao Y, Zhang Y, Cheng W, Deng J. The L-shaped link between total antioxidant capacity and phenotypic age acceleration: evidence from NHANES 2003-2010. Biogerontology 2025; 26:85. [PMID: 40175776 PMCID: PMC11965188 DOI: 10.1007/s10522-025-10223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
This study aimed to investigate the relationship between total antioxidant capacity (TAC) and phenotypic age acceleration (PhenoAgeAccel), a measure of accelerated biological aging, using data from the National Health and Nutrition Examination Survey (NHANES). Data from the 2003-2010 NHANES surveys, encompassing 16,395 participants, were analyzed. Principal component analysis (PCA) was used to reduce data dimensionality. Multivariate logistic regression models were employed to evaluate the association between TAC and antioxidant vitamins (α-carotene, β-carotene, β-cryptoxanthin, lycopene, lutein-zeaxanthin, vitamin A, vitamin C, vitamin E) with PhenoAgeAccel, adjusting for demographic, lifestyle, and clinical factors. Smoothed curve fitting and threshold effects analysis were conducted to explore the nonlinear relationship between log-transformed TAC and PhenoAgeAccel. Subgroup analyses were performed to assess potential effect modifiers based on age, gender, race, education, smoking, alcohol use, diabetes, hypertension, and hyperlipidemia. The weak correlations between the original variables prevent PCA from effectively capturing the primary variability within the data. Higher TAC was significantly inversely associated with PhenoAgeAccel in both unadjusted and adjusted models. Participants in the second tertile (T2) of TAC exhibited 11% lower odds of PhenoAgeAccel compared to those in the first tertile (T1) (OR = 0.89, 95% CI: 0.81-0.98, P = 0.0176). Intake of several antioxidant vitamins, including α-carotene, β-carotene, lutein-zeaxanthin, vitamin A, vitamin C, and vitamin E, was also inversely associated with the odds of PhenoAgeAccel. A nonlinear relationship between log-transformed TAC and PhenoAgeAccel was observed, with a significant protective effect within a specific range of TAC. Subgroup analyses revealed no significant effect modification by most factors, except for gender, smoking, and alcohol consumption. TAC is closely associated with PhenoAgeAccel. A nonlinear relationship was observed, with higher TAC exhibiting significant protective effects within a specific range, particularly among males, smokers, and alcohol consumer. These findings underscore the potential value of TAC in mitigating the aging process.
Collapse
Affiliation(s)
- Yukun Wu
- The Affiliated Changsha Central Hospital, Department of Peripheral Vascular Intervention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Mengxiang Xiang
- The Affiliated Changsha Central Hospital, Department of Peripheral Vascular Intervention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yangcheng Zhao
- The Affiliated Changsha Central Hospital, Department of Peripheral Vascular Intervention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yu Zhang
- The Affiliated Changsha Central Hospital, Department of Peripheral Vascular Intervention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Wenxiang Cheng
- The Affiliated Changsha Central Hospital, Department of Peripheral Vascular Intervention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jiangbei Deng
- The Affiliated Changsha Central Hospital, Department of Peripheral Vascular Intervention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
4
|
Yu K, Li X, Shi X, Li R, Zhang M. EEPD1 regulates inflammation and endothelial apoptosis in atherosclerosis through KLF4-EEPD1-ERK axis. Clin Transl Med 2025; 15:e70311. [PMID: 40268512 PMCID: PMC12017893 DOI: 10.1002/ctm2.70311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Inflammation and endothelial apoptosis are implicated in the advancement of atherosclerosis. EEPD1 holds a pivotal position in the repair of DNA damage and contributes to the progression of multiple cancers. However, the role of EEPD1 in cardiovascular diseases needs to be explored further, especially in atherosclerosis. METHODS We constructed EEPD1 and ApoE (apolipoprotein E)-deficient mice to assess how EEPD1 influences endothelial inflammation and apoptosis within atherosclerotic plaques. High-throughput RNA sequencing of human aortic endothelial cell groups treated with siCon+TNFα and siEEPD1+TNFα identified notable disparities in the MAPK pathway between groups. Chromatin immunoprecipitation and luciferase reporter assay confirmed that KLF4 directly regulates EEPD1. RESULTS Further examination of gene expression data revealed elevated EEPD1 concentrations in atherosclerotic plaques of patients, which findings were corroborated in the aortas of ApoE-/- mice. Present study demonstrated that adhesion molecule expression, endothelial apoptosis, aortic root plaques and macrophage accumulation were markedly ameliorated in EEPD1-/-ApoE-/- mice compared to WT ApoE-/- mice. Functional analysis revealed that increase in EEPD1 promotes ERK phosphorylation and significantly increases endothelial apoptosis and inflammation in atherosclerosis, which was abrogated by inhibition of ERK phosphorylation. We found KLF4 to be the transcription repressor of EEPD1 through luciferase assay and chromatin immunoprecipitation, and KLF4 inhibition abrogated the amelioration of endothelial apoptosis and inflammation caused by EEPD1 deletion. CONCLUSIONS Collectively, this study revealed that EEPD1 deletion can lead to amelioration of atherosclerosis through the KLF4-EEPD1-ERK axis. Hence, targeting EEPD1 could be a promising therapeutic strategy for patients with atherosclerosis.
Collapse
Affiliation(s)
- Kaiwen Yu
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Xiang Li
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Xin Shi
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Ruogu Li
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Min Zhang
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| |
Collapse
|
5
|
Pinheiro VDS, Proctor DN, Nogueira Soares R, Alvares TS. Effect of 12-wk dietary nitrate supplementation on carotid arterial stiffness in postmenopausal females. Am J Physiol Heart Circ Physiol 2025; 328:H937-H944. [PMID: 40066813 DOI: 10.1152/ajpheart.00065.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Menopause is associated with reduced nitric oxide (NO) bioavailability, a key contributor to increased arterial stiffness and, consequently, greater risk of cardiovascular disease-related mortality in postmenopausal females. Even though dietary nitrate has been shown to increase NO bioavailability in postmenopausal females acutely, previous studies showed no impact of dietary nitrate supplementation on arterial stiffness in postmenopausal females. Their findings were likely limited by the acute and/or short-term design. Thus, this study aimed to determine whether 12 wk of dietary nitrate supplementation via beetroot extract improves carotid artery stiffness in postmenopausal females. A randomized, double-blind, placebo-controlled, and parallel-design trial was conducted with 20 postmenopausal females (60-85 yr). Participants received nitrate-rich (NR-BEETx, 8.8 mmol/day) or nitrate-depleted (ND-BEETx) beetroot extract. Carotid stiffness parameters-pulse wave velocity (PWVβ), β stiffness, pressure-strain elastic modulus, augmentation index (AIx), and arterial compliance-were measured at baseline and weeks 4, 8, and 12. Serum nitrate and nitrite concentrations and blood pressure were also assessed. Compared with ND-BEETx, NR-BEETx supplementation significantly reduced PWVβ, β stiffness, elastic modulus, and AIx at weeks 4, 8, and 12, whereas arterial compliance increased by week 12. Serum nitrate and nitrite concentrations were elevated five- to sixfold and 1.5- to 2-fold, respectively, in the NR-BEETx group, with peak concentrations occurring at week 8 and showing a plateau or slight decrease at week 12. Blood pressure remained unchanged in both groups. Twelve weeks of nitrate-rich beetroot extract supplementation improved carotid artery stiffness and increased NO bioavailability without altering blood pressure. These findings suggest that beetroot extract supplementation can be recommended as an alternative nutritional strategy to mitigate carotid artery stiffening in postmenopausal females.NEW & NOTEWORTHY Postmenopausal females experience reduced nitric oxide (NO) bioavailability and elevated carotid artery stiffness, a well-established independent risk factor for end-organ damage and all-cause mortality. In this study, we demonstrate that 12 wk of dietary nitrate supplementation through beetroot extract significantly increased NO bioavailability and improved carotid artery stiffness in postmenopausal females.
Collapse
Affiliation(s)
- Vivian Dos Santos Pinheiro
- Food and Nutrition Institute, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - David N Proctor
- Integrative Vascular Physiology Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Rogerio Nogueira Soares
- Division of Kinesiology, Health, and Sports Studies, Wayne State University, Detroit, Michigan, United States
| | - Thiago Silveira Alvares
- Food and Nutrition Institute, Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Xia Z, Xu G, Zhao M, Li Y, Ye P, Liu Y, Gaisano HY, He Y. Total bilirubin modified the association between diabetes and stroke: a cross-sectional study from NHANES 2011-2016. J Neurol Neurosurg Psychiatry 2025; 96:406-414. [PMID: 39231583 DOI: 10.1136/jnnp-2024-334408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Total bilirubin (TBIL) has antioxidant and anti-inflammatory properties. This study aimed to determine whether elevated TBIL could modify the association between diabetes and stroke. METHOD Data were obtained from the National Health and Nutrition Examination Survey 2011-2016. TBIL was stratified by median (10.3 µmol/L). The association between diabetes and stroke was quantified using multivariable logistic regression models. The cut-off concentration for the presence of TBIL modification effects was identified by Johnson-Neyman analyses. Mediation analyses were performed to determine the influence of TBIL on mediating factors that mediate the relationship between diabetes and stroke. RESULTS This cross-sectional study included 16 130 participants, with the mean age of 46.8±0.4 years and 48.5% of men. Diabetes was associated with the presence of stroke at TBIL <10.3 µmol/L (OR=2.19, 95% CI 1.58 to 3.05) but not at TBIL ≥10.3 µmol/L (OR=1.27, 95% CI 0.85 to 1.88) after adjustment for confounders. Above associations were significantly different between the two TBIL concentrations (P for interaction=0.03). Moreover, the modification effect of TBIL specifically occurred in men (P for interaction=0.02) rather than in women (P for interaction=0.08). The cut-off concentration for the presence of TBIL modification effects was 17.05 µmol/L. Additionally, the TBIL of ≥10.3 µmol/L inhibited mediating effects of hypersensitive C reactive protein (mediating effect=0.03, 95% CI -0.15 to 0.22, P=0.72) and systemic immune-inflammation index (mediating effect=0.01, 95% CI -0.01 to 0.04, P=0.29) as compared with the TBIL of <10.3 µmol/L. CONCLUSIONS Elevated TBIL modified the association between diabetes and stroke through inhibiting mediating effects of inflammatory factors.
Collapse
Affiliation(s)
- Zhang Xia
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Guozheng Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Mingyang Zhao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yuhao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Peiyu Ye
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Center for Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yijian Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Herbert Y Gaisano
- Department of Medicine and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yan He
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
7
|
Gu S, Kopecky BJ, Peña B, Vagnozzi RJ, Lahm T. Sex-dependent Pathophysiology and Therapeutic Considerations in Right Heart Disease. Can J Cardiol 2025:S0828-282X(25)00178-3. [PMID: 40054579 DOI: 10.1016/j.cjca.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Right ventricular (RV) adaptation to the increased afterload in the setting of pulmonary hypertension (PH) and other cardiac and pulmonary vascular conditions is a major determinant of survival. Although the RV remains understudied and less well understood than the left ventricle, recent advances have been made in understanding the function and biology of the RV in health and in disease, particularly in PH. RV adaptation in PH exhibits significant sexual dimorphisms in pathophysiology, adaptation, and outcomes. Despite a higher incidence of PH, women consistently demonstrate better RV adaptation and survival rates in the setting of increased RV afterload compared with men. Sexual dimorphisms extend to therapy responsiveness, with women benefiting more from certain pulmonary vasodilators and exhibiting superior RV recovery. In this review we discuss the current literature on sexual dimorphisms in RV structure, function, and molecular pathways in health and disease, as well as in RV-specific clinical manifestations, treatments, and outcomes in PH. Sex steroid-mediated effects as well as emerging studies on sex steroid-independent effects are reviewed. In general, sex steroids such as 17β-estradiol and dehydroepiandrosterone exert RV-protective effects. In contrast, testosterone negatively impacts RV structure and function. Emerging evidence highlights the influence of nonhormonal genetic determinants, such as BMPR1A and DMRT2 loci, which are associated with better RV function in women. A better understanding of the interplay between sex hormones, genetic factors, and RV biology is crucial for advancing and developing RV-directed therapies for patients of either sex.
Collapse
Affiliation(s)
- Sue Gu
- Cardio Vascular Pulmonary Research Laboratory, University of Colorado School of Medicine, Aurora, Colorado, USA; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Benjamin J Kopecky
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, College of Engineering, Design and Computing, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ronald J Vagnozzi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, USA; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA.
| |
Collapse
|
8
|
Cui J, Zhao YC, She LZ, Wang TJ. Causal relationship between uterine fibroids and cardiovascular disease: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41713. [PMID: 40020116 PMCID: PMC11875593 DOI: 10.1097/md.0000000000041713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Previous studies have indicated that patients with uterine fibroids (UF) may have an elevated risk of cardiovascular disease (CVD), although the causal relationship between UF and CVD remains unclear. In this Mendelian randomization (MR) study, we aimed to investigate the causal association between genetic susceptibility to UF and the risk of developing CVD. We extracted summary statistics for single nucleotide polymorphisms associated with UF and 5 CVDs from multiple databases for further analysis. First, we used linkage disequilibrium score regression to assess the genetic correlation across the genome. Next, we performed univariate MR (UVMR), and to ensure the robustness of our results, we conducted sensitivity analyses using several methods. Additionally, we applied multivariable MR (MVMR) to adjust for potential confounders. The linkage disequilibrium score regression results showed that there was no genetic correlation between UF and coronary heart disease, myocardial infarction (MI), atrial fibrillation, heart failure, cardioembolic stroke (CES). The UVMR revealed a significant association between UF and CES (OR = 1.113, 95% confidence interval [CI]: 1.018-1.218, P = .019, PFDR = .047) and a suggestive causal relationship between UF and MI (OR = 0.943, 95% CI: 0.899-0.989, P = .015, PFDR = .075). In the MVMR analysis, after adjusting for a range of potential confounders, the causal relationships between UF and both CES (OR = 1.104, 95% CI = 1.012-1.205, P = .027) and MI (OR = 0.935, 95% CI = 0.882-0.992, P = .025) remained significant. Our study found that UF increase the risk of CES but decrease the risk of MI, providing a theoretical basis for further research into the underlying mechanisms.
Collapse
Affiliation(s)
- Jie Cui
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yue-Chen Zhao
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Li-Zhen She
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Tie-Jun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
9
|
Masenga SK, Wandira N, Cattivelli-Murdoch G, Saleem M, Beasley H, Hinton A, Ertuglu LA, Mwesigwa N, Kleyman TR, Kirabo A. Salt sensitivity of blood pressure: mechanisms and sex-specific differences. Nat Rev Cardiol 2025:10.1038/s41569-025-01135-0. [PMID: 39984695 DOI: 10.1038/s41569-025-01135-0] [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] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular disease in individuals with or without hypertension. However, the mechanisms and management of SSBP remain unclear, mainly because the diagnosis of this condition relies on salt loading-depletion protocols that are not feasible in the clinic. The prevalence of hypertension is lower in premenopausal women than in men, but this sex-specific difference is reversed after menopause. Whether excessive SSBP in women at any age contributes to this reversal is unknown, but many clinical studies that have rigorously assessed for SSBP using salt loading-depletion protocols have confirmed that SSBP is more prevalent in women than in men, including during premenopausal age. In this Review, we discuss sex-specific mechanisms of SSBP. We describe sex-related differences in renal transporters, hypertensive pregnancy, SSBP in autoimmune disorders and mitogen-activated protein kinase signalling pathways, and highlight limitations and lessons learned from Dahl salt-sensitive rat models.
Collapse
Affiliation(s)
- Sepiso K Masenga
- HAND research Group, Department of Pathology and Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia.
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Nelson Wandira
- Vanderbilt Mater of Public Health Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lale A Ertuglu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naome Mwesigwa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, Department of Cell Biology, and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
10
|
Yao C, Dong Y, Zhou H, Zou X, Alhaskawi A, Ezzi SHA, Wang Z, Lai J, Kota VG, Abdulla MHAH, Liu Z, Abdalbary SA, Alenikova O, Lu H. COVID-19 and acute limb ischemia: latest hypotheses of pathophysiology and molecular mechanisms. J Zhejiang Univ Sci B 2025; 26:333-352. [PMID: 40274383 PMCID: PMC12021539 DOI: 10.1631/jzus.b2300512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/01/2024] [Indexed: 04/26/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is a multi-system disease that can lead to various severe complications. Acute limb ischemia (ALI) has been increasingly recognized as a COVID-19-associated complication that often predicts a poor prognosis. However, the pathophysiology and molecular mechanisms underlying COVID-19-associated ALI remain poorly understood. Hypercoagulability and thrombosis are considered important mechanisms, but we also emphasize the roles of vasospasm, hypoxia, and acidosis in the pathogenesis of the disease. The angiotensin-converting enzyme 2 (ACE2) pathway, inflammation, and platelet activation may be important molecular mechanisms underlying these pathological changes induced by COVID-19. Furthermore, we discuss the hypotheses of risk factors for COVID-19-associated ALI from genetic, age, and gender perspectives based on our analysis of molecular mechanisms. Additionally, we summarize therapeutic approaches such as use of the interleukin-6 (IL-6) blocker tocilizumab, calcium channel blockers, and angiotensin-converting enzyme inhibitors, providing insights for the future treatment of coronavirus-associated limb ischemic diseases.
Collapse
Affiliation(s)
- Chengjun Yao
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yanzhao Dong
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaodi Zou
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| | - Ahmad Alhaskawi
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sohaib Hasan Abdullah Ezzi
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zewei Wang
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingtian Lai
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Vishnu Goutham Kota
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | | | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sahar Ahmed Abdalbary
- Department of Orthopaedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef 2711860, Egypt
| | - Olga Alenikova
- Republic Scientific Practical Center of Neurology and Neurosurgery, Ministry of Health of the Republic of Belarus, Minsk 220004, Belarus
| | - Hui Lu
- Department of Orthopaedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
11
|
Saijo Y, Okushi Y, Gillinov AM, Pettersson GB, Unai S, Grimm RA, Griffin BP, Xu B. Sex-related differences in outcomes and prognosis of severe calcific mitral stenosis due to mitral annular calcification: A propensity-score matched cohort study. Int J Cardiol 2025; 421:132893. [PMID: 39647783 DOI: 10.1016/j.ijcard.2024.132893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/02/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND The prevalence of severe calcific mitral stenosis (MS) is higher in female patients. The aim of the study was to assess the sex-related differences in clinical characteristics and prognosis in patients with severe calcific MS. METHODS Among 7154 adult patients with MS due to mitral annular calcification who underwent echocardiography between October 2010 and August 2020, 287 patients with severe MS were retrospectively analyzed. The endpoint was all-cause mortality. We calculated a propensity score matched analysis with 22 potential confounding covariates including clinical characteristics and medication usage. RESULTS In the entire cohort, there was a predominance of female patients (66 %), and the mean age was 72 ± 11 years. While 97 patients (34 %) underwent MV intervention, 190 patients (66 %) were conservatively managed. During a median follow-up of 12 months (25th -75th percentile: 3-29 months), 102 patients (36 %) died. The cumulative survival rate of female patients was lower compared with male patients in conservative treatment group (p = 0.012), while the cumulative survival rate was comparable between the sexes in MV intervention group (p = 0.63). Even after propensity score matching in 170 patients (85 females and 85 males), similar results were obtained (p = 0.012 for conservative treatment group, p = 0.61 for MV intervention group). CONCLUSIONS Female sex predominated in patients with severe calcific MS. Female patients with severe calcific MS had worse prognosis than male patients when treated conservatively, while in patients undergoing MV intervention, prognosis was similar between the sexes.
Collapse
Affiliation(s)
- Yoshihito Saijo
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuichiro Okushi
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - A Marc Gillinov
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Gösta B Pettersson
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Shinya Unai
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Richard A Grimm
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Griffin
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bo Xu
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
12
|
Liu X, Zhao Y, Feng Y, Wang S, Luo A, Zhang J. Ovarian Aging: The Silent Catalyst of Age-Related Disorders in Female Body. Aging Dis 2025:AD.2024.1468. [PMID: 39965250 DOI: 10.14336/ad.2024.1468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
Age-related diseases have emerged as a global concern as the population ages. Consequently, understanding the underlying causes of aging and exploring potential anti-aging interventions is imperative. In females, the ovaries serve as the principal organs responsible for ovulation and the production of female hormones. The aging ovaries are related to infertility, menopause, and associated menopausal syndromes, with menopause representing the culmination of ovarian aging. Current evidence indicates that ovarian aging may contribute to dysfunction across multiple organ systems, including, but not limited to, cognitive impairment, osteoporosis, and cardiovascular disease. Nevertheless, due to the widespread distribution of sex hormone receptors throughout the body, ovarian aging affects not only these specific organs but also influences a broader spectrum of age-related diseases in women. Despite this, the impact of ovarian aging on overall age-related diseases has been largely neglected. This review provides a thorough summary of the impact of ovarian aging on age-related diseases, encompassing the nervous, circulatory, locomotor, urinary, digestive, respiratory, and endocrine systems. Additionally, we have outlined prospective therapeutic approaches for addressing both ovarian aging and age-related diseases, with the aim of mitigating their impacts and preserving women's fertility, physical health, and psychological well-being.
Collapse
Affiliation(s)
- Xingyu Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanqu Zhao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanzhi Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Aiyue Luo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
13
|
Doughan M, Chehab O, Doughan B, Lima JAC, Michos ED. Association of endogenous sex hormone levels with tooth loss due to periodontitis in men and post-menopausal women: The multi-ethnic study of atherosclerosis. J Periodontal Res 2025; 60:166-176. [PMID: 38895935 DOI: 10.1111/jre.13312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
AIM To investigate the association between endogenous sex hormone levels and history of tooth loss related to periodontitis in healthy middle-aged to older men and post-menopausal women. METHODS This cross-sectional study included 5649 participants aged 45-84 (mean age, 63 ± 10 years) from the Multi-Ethnic Study of Atherosclerosis cohort who had sex hormone levels measured and answered a questionnaire regarding perceived periodontal status at exam 1. Multivariable logistic regression was used to examine the association of sex hormones (exposure) with history of tooth loss (outcome), stratified by sex. RESULTS Among post-menopausal women, higher free testosterone (per 1SD) was associated with a greater prevalence of tooth loss [OR 1.49 (95% CI, 1.08-2.05)], whereas higher sex hormone binding globulin (SHBG) was associated with a lower prevalence of tooth loss [OR 0.74 (0.58-0.94)], after adjustment for cardiometabolic risk factors and reproductive factors. In men, higher free testosterone and lower SHBG were associated with a lower prevalent probability of tooth loss in unadjusted analysis, but these associations lost significance after covariate adjustment. CONCLUSION A higher androgenic sex hormone profile in post-menopausal women (i.e., increased free testosterone, lower SHBG) was associated with an increased prevalence of tooth loss, after adjusting cardiometabolic risk factors. No such association was found in men. These findings suggest that sex hormones may influence or serve as a marker for periodontal health.
Collapse
Grants
- R01 HL074406 NHLBI NIH HHS
- R01 HL074338 NHLBI NIH HHS
- 946222 American Heart Association
- HHSN268201500003I NHLBI NIH HHS
- N01-HC-95159 National Heart, Lung and Blood Institute
- N01-HC-95160 National Heart, Lung and Blood Institute
- N01-HC-95161 National Heart, Lung and Blood Institute
- N01-HC-95162 National Heart, Lung and Blood Institute
- N01-HC-95163 National Heart, Lung and Blood Institute
- N01-HC-95164 National Heart, Lung and Blood Institute
- N01-HC-95165 National Heart, Lung and Blood Institute
- N01-HC-95166 National Heart, Lung and Blood Institute
- N01-HC-95167 National Heart, Lung and Blood Institute
- N01-HC-95168 National Heart, Lung and Blood Institute
- N01-HC-95169 National Heart, Lung and Blood Institute
- Johns Hopkins University: Amato Fund in Women's Cardiovascular Health Research
Collapse
Affiliation(s)
- Maria Doughan
- School of Dentistry, University of Maryland, Baltimore, Maryland, USA
| | - Omar Chehab
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bassel Doughan
- Faculty of Dental Surgery, Côte d'Azur University, Nice, France
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Miller KB, Moir ME, Fico BG. Vascular health and exercise in females throughout the lifespan: Exploring puberty, pregnancy and menopause. Exp Physiol 2025. [PMID: 39887530 DOI: 10.1113/ep092170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025]
Abstract
This narrative review highlights the impact of exercise on vascular health in females over the lifespan with an emphasis on puberty, pregnancy and menopause. These events encompass substantial changes in sex hormone levels, particularly oestrogens and progesterone. They are also accompanied by distinct adaptations of the central, peripheral and cerebral vasculature. Regular exercise is an effective mechanism to reduce vascular risk in females of all ages, especially for those at higher risk for vascular disorders. However, there are large variabilities in the vascular adaptations to exercise in females that may be related to circulating sex hormone levels. In addition, exogenous hormones, such as oral contraceptives taken after puberty or hormonal replacement therapy taken to mitigate symptoms of menopause, may interact with exercise-induced changes in vascular function. We highlight how more research is needed to understand the optimal exercise interventions to promote vascular health in females across the lifespan, especially during times of hormonal transition.
Collapse
Affiliation(s)
- Kathleen B Miller
- Department of Health and Exercise Science, Morrison Family College of Health, University of St. Thomas, Saint Paul, Minnesota, USA
| | - M Erin Moir
- Bruno Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Brandon G Fico
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, Florida, USA
| |
Collapse
|
15
|
Levy MV, Fandl HK, Hijmans JG, Stockelman KA, Ruzzene ST, Reiakvam WR, Goldthwaite ZA, Greiner JJ, DeSouza CA, Garcia VP. Effect of 17β-Estradiol on Endothelial Cell Expression of Inflammation- Related MicroRNA. Microrna 2025; 14:3-8. [PMID: 39069709 DOI: 10.2174/0122115366320085240716180112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Estrogen plays a protective role in vascular health due, in part, to its regulation of endothelial inflammation. However, the mechanism(s) by which estrogen negatively regulates inflammatory signaling pathways is not completely understood. MicroRNAs (miRNAs) are recognized as sensitive and selective regulators of cardiovascular function, inflammation, and disease, yet the effects of 17β-estradiol on the endothelial miRNA profile are largely unknown. OBJECTIVE The aim of this study was to determine the effect of 17β-estradiol on the expression of inflammation-associated miRNAs in endothelial cells in vitro. METHODS Human Umbilical Vein Endothelial cells (HUVECs) were treated with media in the absence (control) and presence of 17β-estradiol (100 nM) for 24 hr. Thereafter, endothelial cell release of cytokines (IL-6 and IL-8), the intracellular expression of the central protein inflammatory mediator NF-κB, and the levels of inflammatory-associated miRNAs: miR-126, miR-146a, miR-181b, miR-204, and miR-Let-7a, were determined. RESULTS 17β-estradiol-treated cells released significantly lower levels of IL-6 (47.6±1.5 pg/mL vs. 59.3±4.9 pg/mL) and IL-8 (36.3±2.3 pg/mL vs. 44.0±2.0 pg/mL). Cellular expression of total NF-κB (26.0±2.8 AU vs. 21.2±3.1 AU) was not different between groups; however, activated NF-κB (Ser536) (12.9±1.7 AU vs. 20.2±2.2 AU) was markedly reduced in 17β-estradiol-treated cells as compared to untreated cells. Furthermore, cellular expressions of miR-126 (1.8±0.3 fold), miR-146a (1.7±0.3 fold), miR-181b (2.1±0.4 fold), miR-204 (1.9±0.4 fold), and miR-Let-7a (1.8±0.3 fold) were markedly increased in response to 17β-estradiol treatment. CONCLUSION These data suggest that the anti-inflammatory effect of 17β-estradiol in endothelial cells may be mediated by miRNAs.
Collapse
Affiliation(s)
- Ma'ayan V Levy
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Hannah K Fandl
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Jamie G Hijmans
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Kelly A Stockelman
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Samuel T Ruzzene
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Whitney R Reiakvam
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Zoe A Goldthwaite
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Jared J Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Christopher A DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| | - Vinicius P Garcia
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado Boulder Boulder, CO 80309, United States
| |
Collapse
|
16
|
Markova I, Hüttl M, Gayova N, Miklankova D, Cerna K, Kavanova M, Skaroupkova P, Cacanyiova S, Malinska H. Visceral Adipose Tissue Inflammation and Vascular Complications in a Rat Model with Severe Dyslipidemia: Sex Differences and PAI-1 Tissue Involvement. Biomolecules 2024; 15:19. [PMID: 39858414 PMCID: PMC11763299 DOI: 10.3390/biom15010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
We investigated the sex-dependent effects of inflammatory responses in visceral adipose tissue (VAT) and perivascular adipose tissue (PVAT), as well as hematological status, in relation to cardiovascular disorders associated with prediabetes. Using male and female hereditary hypertriglyceridemic (HHTg) rats-a nonobese prediabetic model featuring dyslipidemia, hepatic steatosis, and insulin resistance-we found that HHTg females exhibited more pronounced hypertriglyceridemia than males, while HHTg males had higher non-fasting glucose levels. Additionally, HHTg females had higher platelet counts, larger platelet volumes, and lower antithrombin inhibitory activity. Regarding low-grade chronic inflammation, HHTg males exhibited increased serum leptin and leukocyte levels, while females had increased serum interleukin-6 (IL-6). Both sexes had increased circulating plasminogen activator inhibitor-1 (PAI-1), higher PAI-1 gene expression in VAT and PVAT, and elevated intercellular adhesion molecule-1 (ICAM-1) gene expression in the aorta, contributing to endothelial dysfunction in the HHTg strain. However, HHTg females had lower tumor necrosis factor alpha (TNFα) gene expression in the aorta. Severe dyslipidemia in this prediabetic model was associated with hypercoagulation and low-grade chronic inflammation. The increase in PAI-1 expression in both VAT and PVAT seems to indicate a link between inflammation and vascular dysfunction. Despite the more pronounced dyslipidemia and procoagulation status in females, their milder inflammatory response may reflect an association between reduced cardiovascular damage and prediabetes.
Collapse
Affiliation(s)
- Irena Markova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| | - Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| | - Natalie Gayova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| | - Denisa Miklankova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| | - Kristyna Cerna
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| | - Martina Kavanova
- Department of Laboratory Methods, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic;
| | - Petra Skaroupkova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| | - Sona Cacanyiova
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia;
| | - Hana Malinska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.M.); (M.H.); (N.G.); (D.M.); (K.C.); (P.S.)
| |
Collapse
|
17
|
Piechota-Polanczyk A, Mariwani Z, Fichna J, Polanczyk A, Jozkowicz A. Chemical Inhibition of NRF2 Transcriptional Activity Influences Colon Function and Oestrogen Receptor Expression in Mice at Different Ages. Int J Mol Sci 2024; 25:13647. [PMID: 39769410 PMCID: PMC11678879 DOI: 10.3390/ijms252413647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
We aim to investigate whether chemical inhibition of NRF2 transcriptional activity (TA) influences distal colon contractions, particularly in an age-dependent manner in females, and whether it impacts oestrogen receptor signalling in female mice. This study was performed on 3 and 6-month-old female mice treated with ML385 (30 mg/kg) or a vehicle for 7 days (i.p.). The colon functionality was verified with a colon bead expulsion test; serum samples were collected for oestradiol levels, and colon samples were stored for various histological analyses. The results show that the seven-day treatment of ML385 significantly downregulated TA (p < 0.05) and impacted its contractility. Additionally, young females treated with ML385 exhibited an increase in goblet cell number and significantly increased ERα, but not ERβ, especially in older mice. It is worth noting that the basal level of the membrane oestrogen receptor GPR30 was higher in older mice within the epithelial layer, and ML385 treatment led to a downregulation of GPR30 in 6-month-old mice. In summary, ML385 decreases NRF2 TA in the colon and impacts its contractility and goblet cell numbers. Additionally, NRF2 TA influences the expression of oestrogen receptors in the colons of female mice.
Collapse
Affiliation(s)
| | - Zanya Mariwani
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; (Z.M.); (A.J.)
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland;
| | - Andrzej Polanczyk
- The Faculty of Safety Engineering and Civil Protection, Fire University, 01-629 Warsaw, Poland;
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; (Z.M.); (A.J.)
| |
Collapse
|
18
|
Lu Y, Wu S, Zhu S, Shen J, Liu C, Zhao C, Su S, Ma H, Xiang M, Xie Y. Integrated Single-Cell Analysis Revealed Novel Subpopulations of Foamy Macrophages in Human Atherosclerotic Plaques. Biomolecules 2024; 14:1606. [PMID: 39766313 PMCID: PMC11675068 DOI: 10.3390/biom14121606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Foam cell formation is a hallmark of atherosclerosis, yet the cellular complexity within foam cells in human plaques remains unexplored. Here, we integrate published single-cell RNA-sequencing, spatial transcriptomic, and chromatin accessibility sequencing datasets of human atherosclerotic lesions across eight distinct studies. Through this large-scale integration of patient-derived information, we identified foamy macrophages enriched for genes characteristic of the foamy signature. We further re-clustered the foamy macrophages into five unique subsets with distinct potential functions: (i) pro-foamy macrophages, exhibiting relatively high inflammatory and adhesive properties; (ii) phagocytic foamy macrophages, specialized in efferocytosis; (iii) high-efflux foamy macrophages marked by high NR1H3 expression; (iv) mature foamy macrophages prone to programmed cell death; and (v) synthetic subset. Trajectory analysis elucidated a bifurcated differentiation cell fate from pro-foam macrophages toward either the programmed death (iv) or synthetic (v) phenotype. The existence of these foamy macrophage subsets was validated by immunostaining. Moreover, these foamy macrophage subsets exhibited strong potential ligand-receptor interactions. Finally, we conducted Mendelian randomization analyses to identify a possible causal relationship between key regulatory genes along the programmed death pathway in foamy macrophages and atherosclerotic diseases. This study provides a high-resolution map of foam cell diversity and a set of potential key regulatory genes in atherosclerotic plaques, offering novel insights into the multifaceted pathophysiology underlying human atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Meixiang Xiang
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (Y.L.); (S.W.); (S.Z.); (J.S.); (C.L.); (C.Z.); (S.S.); (H.M.)
| | - Yao Xie
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (Y.L.); (S.W.); (S.Z.); (J.S.); (C.L.); (C.Z.); (S.S.); (H.M.)
| |
Collapse
|
19
|
Liu Q, Yu F, Zhou Q, Zhou H, Lu H, Luo J, Gan S. Chinese Visceral Adipose Index is Associated with Arterial Stiffness in Type 2 Diabetes Patients: A Cross-Sectional Study. Int J Gen Med 2024; 17:5681-5696. [PMID: 39650791 PMCID: PMC11622680 DOI: 10.2147/ijgm.s489853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/21/2024] [Indexed: 12/11/2024] Open
Abstract
Purpose The purpose of this study is to investigate the association between Chinese Visceral Adipose Index(CVAI) and brachial-ankle pulse wave velocity (baPWV) in type 2 diabetes (T2D) patients, in order to provide scientific evidence for the prevention and treatment of macrovascular complications in T2D. Patients and Methods This research adopts the cross-sectional study design. Anthropometric assessment, baPWV assessment and biochemical assessment were performed in 2906 T2D patients. CVAI was calculated from the combination of triglycerides, age, high-density lipoprotein cholesterol, waist circumference, body mass index. Multivariate regression analysis and generalized additive model were used to analyze the association between bapwv and CVAI. Subgroup analysis and interaction analysis were used to analyze the influencing factors. Results After adjustment for covariates, baPWV tended to increase in the quartiles of CVAI. In males, As CVAI increases by a unit, baPWV was increased by 0.28 cm/s (95% CI: -0.05, 0.61 P=0.0934). In females, As CVAI increases by a unit, baPWV was increased by 1.60 cm/s (95% CI: 1.07, 2.14). A non-linear connection in males and a linear connection in females between the CVAI and baPWV was revealed. In males, we found that CVAI interacted with baPWV in different duration of diabetes (P interaction = 0.0052), alcohol consumption status (P interaction = 0.0375). In females, CVAI interacted with baPWV in glycated hemoglobin (P interaction = 0.0003), systolic blood pressure (P interaction = 0.0001), diastolic blood pressure (P interaction<0.0001), duration of diabetes (P interaction = 0.0014), the use or non-use of glucose-lowering drugs (P interaction = 0.0006), the use or non-use antihypertensive drugs (P interaction = 0.0004), females' menopausal status (P interaction = 0.0012). Conclusion The relationship between CVAI and baPWV in T2D patients is positively non-linear in males and linear in females. In all subjects, this relationship was influenced by diabetes duration. In males, drinking status affected this relationship, and in females, It is influenced by blood pressure, glycemic control, menopausal status, and the use of glucose-lowering and hypotensive drug, highlighting the complex interplay between visceral fat and arterial stiffness in the T2D population.
Collapse
Affiliation(s)
- Qin Liu
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Fang Yu
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Quan Zhou
- Department of Science and Education, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Haifeng Zhou
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Huilin Lu
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Jian Luo
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Shenglian Gan
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| |
Collapse
|
20
|
Bowie DC, Low KA, Rubenstein SL, Islam SS, Zimmerman B, Camacho PB, Sutton BP, Gratton G, Fabiani M. Neurovascular mechanisms of cognitive aging: Sex-related differences in the average progression of arteriosclerosis, white matter atrophy, and cognitive decline. Neurobiol Dis 2024; 201:106653. [PMID: 39214337 DOI: 10.1016/j.nbd.2024.106653] [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: 03/21/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Arterial stiffness (arteriosclerosis) has been linked to heightened risks for cognitive decline, and ultimately for Alzheimer's disease and other forms of dementia. Importantly, neurovascular outcomes generally vary according to one's biological sex. Here, capitalizing on a large sample of participants with neuroimaging and behavioral data (N = 203, age range = 18-87 years), we aimed to provide support for a hierarchical model of neurocognitive aging, which links age-related declines in cerebrovascular health to the rate of cognitive decline via a series of intervening variables, such as white matter integrity. By applying a novel piecewise regression approach to our cross-sectional sample to support Granger-like temporal inferences, we show that, on average, a precipitous decline in cerebral arterial elasticity (measured with diffuse optical imaging of the cerebral arterial pulse; pulse-DOT) precedes an acceleration in the development of white matter lesions by nearly a decade, with women protected from these deleterious effects until approximately age 50, the average onset of menopause. By employing multiple-mediator path analyses while controlling for sex, we show that age may impair cognition via the sequential indirect effects of arteriosclerosis and white matter atrophy on fluid, but not crystallized, abilities. Importantly, we replicate these results using pulse pressure, an independent index of arterial health, thereby providing converging evidence for the central role of arteriosclerosis as an accelerating factor in normal and pathological aging and identifying robust sex-related differences in the progression of cerebral arteriosclerosis and white matter degradation.
Collapse
Affiliation(s)
- Daniel C Bowie
- Department of Psychology, University of Illinois Urbana-Champaign, 603 E Daniel St., Champaign, IL 61820, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America.
| | - Kathy A Low
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America
| | - Samantha L Rubenstein
- Department of Psychology, University of Illinois Urbana-Champaign, 603 E Daniel St., Champaign, IL 61820, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America
| | - Samia S Islam
- Department of Psychology, University of Illinois Urbana-Champaign, 603 E Daniel St., Champaign, IL 61820, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America
| | - Benjamin Zimmerman
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America; Helfgott Research Institute, National University of Natural Medicine, 2220 SW 1st Ave., Portland, OR 97201, United States of America
| | - Paul B Camacho
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America
| | - Bradley P Sutton
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America; Department of Bioengineering, University of Illinois Urbana-Champaign, 1406 W Green St, Urbana, IL 61801, United States of America
| | - Gabriele Gratton
- Department of Psychology, University of Illinois Urbana-Champaign, 603 E Daniel St., Champaign, IL 61820, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America
| | - Monica Fabiani
- Department of Psychology, University of Illinois Urbana-Champaign, 603 E Daniel St., Champaign, IL 61820, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, IL 61801, United States of America.
| |
Collapse
|
21
|
Wang Y, Chen C, Chou K, Huang S, Ou K. Low-dose estrogen release from silastic capsule enhanced flap wound healing in an animal model. Int Wound J 2024; 21:e70083. [PMID: 39401979 PMCID: PMC11473190 DOI: 10.1111/iwj.70083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/23/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Deep and extensive wounds usually cannot be closed directly by suturing or skin grafting. Flap transplantation is typically used to reconstruct large wounds clinically. The flap survival is based on a stable blood perfusion. It is established that estrogen promotes wound healing and angiogenesis, and regulates the inflammatory response, leading to enhanced flap survival after transplantation. However, estrogen concentrations administered in previous studies were significantly higher than physiological levels, potentially causing systemic side effects. Estrogen-sustained-release silastic capsules can maintain blood serum estrogen closer to physiological levels. This study aimed to investigate whether administering estrogen at a lower concentration, closer to physiological levels, could still enhance flap survival. MATERIALS AND METHODS This study was performed in a random skin flap model in ovariectomized (OVX) mice. Sustained-release estrogen silastic capsules were implanted into OVX mice to determine the functional role of estrogen in wound healing after flap transplantation. Flap blood perfusion was analysed using a colour laser Doppler scanner. Immunohistochemical staining of CD31, hypoxia-inducible factor 1 alpha (HIF-1α), alpha-smooth muscle actin (α-SMA), cleaved caspase 3 and apoptotic terminal dUTP nick end-labelling stain was used to investigate flap angiogenesis, tissue hypoxia, wound healing and cell death in the flap tissue, respectively. RESULTS We observed that administering estrogen at a lower concentration enhanced superficial blood perfusion while reducing the flap's ischemic area and tissue necrosis. HIF-1α expression was significantly decreased in the dermis layer but not in the fascia, whereas cleaved caspase 3 levels decreased in the fascia but remained unchanged in the dermis. Additionally, there was no significant difference in CD31and α-SMA expression between the groups. CONCLUSION In summary, the study showed that an estrogen silastic capsule maintained physiological estrogen levels and improved superficial perfusion, thereby reducing dermal hypoxia, and cell death in a mouse random pattern skin flap model. Although no significant promotion of angiogenesis was observed, the study suggests that appropriate estrogen supplements could enhance flap wound recovery.
Collapse
Affiliation(s)
- Yi‐Wen Wang
- Department and Graduate Institute of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Chun‐Ting Chen
- Division of Gastroenterology and Hepatology, Department of Internal MedicineTri‐Service General Hospital Penghu Branch, National Defense Medical CenterPenghuTaiwan
- Division of Gastroenterology and Hepatology, Department of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Kai‐Chieh Chou
- Graduate Institute of Life Sciences, National Defense Medical CenterTaipeiTaiwan
| | - Shih‐Ming Huang
- Department of BiochemistryNational Defense Medical CenterTaipeiTaiwan
| | - Kuang‐Ling Ou
- Burn Center and Division of Plastic and Reconstructive Surgery, Department of SurgeryTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| |
Collapse
|
22
|
Wu B, Qiu L, Lin Y, Lin Q, Pan Y. The association between the dietary inflammatory index and cardiorespiratory fitness in United States young adults: a cross-sectional study from the National Health and Nutrition Examination Study, 1999-2004. Front Nutr 2024; 11:1442710. [PMID: 39391678 PMCID: PMC11464452 DOI: 10.3389/fnut.2024.1442710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Background Cardiorespiratory fitness (CRF) is a vital indicator of overall health and cardiovascular efficiency. Systemic inflammation significantly impacts CRF, and reducing systemic inflammation may serve as an effective strategy to improve CRF. Diet plays a crucial role in systemic inflammation, but daily dietary intake typically involves multiple elements rather than a single nutrient. The Dietary Inflammatory Index (DII) provides an overall assessment of dietary inflammation on the basis of the anti-inflammatory and pro-inflammatory effects of the nutrients consumed. However, the relationship between DII and CRF is not yet well understood. Aims To examine the association between the DII and CRF. Method This study analyzed 3,087 participants from the National Health and Nutrition Examination Survey (NHANES) between 1999 and 2002. The study subjects were divided into three distinct groups by DII tertile: T1 (n = 1,027), T2 (n = 1,029), and T3 (n = 1,031). The associations between DII levels and CRF were examined via logistic regression analysis and restricted cubic splines (RCSs). Results Elevated DII scores were significantly linked to low CRF levels. Compared with those in the lowest tertile, participants in the highest DII tertile exhibited a greater prevalence of low CRF (T1: 10.85%, T2: 16.32%, T3: 19.31%). In the model with full adjustments, elevated scores on the DII were consistently linked with a heightened likelihood of low CRF (OR: 1.17, 95% CI: 1.07-1.28; P < 0.001). Compared with those in the T1 group, participants with higher DIIs had an increased risk of lower CRF (T2: OR: 1.42, 95% CI: 1.01-2.01, P = 0.046; T3: OR: 1.71, 95% CI: 1.22-2.40, P = 0.003). Additionally, a significant interaction (P = 0.045) between sex and the DII for low CRF was observed within the population. Conclusion A higher DII score is linked to an elevated risk of low CRF. Moreover, sex can impact CRF, with women being more prone to low CRF.
Collapse
Affiliation(s)
- Bo Wu
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Lanlan Qiu
- Department of Cardiology, Longyan People’s Hospital, Longyan, China
| | - Yun Lin
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Qian Lin
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuxiong Pan
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| |
Collapse
|
23
|
Wang X, Han Y, Li H, Wang X, Wang G. Impact of a Precision Intervention for Vascular Health in Middle-Aged and Older Postmenopausal Women Using Polar Heart Rate Sensors: A 24-Week RCT Study Based on the New Compilation of Tai Chi (Bafa Wubu). SENSORS (BASEL, SWITZERLAND) 2024; 24:5832. [PMID: 39275744 PMCID: PMC11398210 DOI: 10.3390/s24175832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024]
Abstract
(1) Background: This study utilized a 24-week intervention incorporating heart rate sensors for real-time monitoring of intervention training, aiming to comprehensively assess the effects of Tai Chi on vascular endothelial function, atherosclerosis progression, and lipid metabolism. The insights gained may inform personalized non-pharmacological interventions to enhance the management of cardiovascular health in this population to provide sustainable benefits and improve quality of life. (2) Methods: Forty postmenopausal middle-aged and elderly women were randomly assigned to an exercise or control group. The exercise group underwent a 24-week Tai Chi (BaFa WuBu) training intervention with real-time heart rate monitoring using Polar sensors. Pre- and post-intervention assessments included body composition, blood pressure, vascularity, and blood parameters measured with the Inbody 720, Vascular Endothelial Function Detector, and Arteriosclerosis. Data were analyzed using SPSS 26.0 and mixed-design ANOVA to assess the effects of time, group, and their interactions on study outcomes. (3) Results: After training through 24 weeks of Tai Chi (BaFa WuBu) intervention, compared with the control group, systolic blood pressure in the exercise group was significantly lower (p < 0.05), and the difference between left and right arm pulse pressure, left and right ankle mean arterial pressure, left and right side baPWV, left and right side ABI, TC, TG, LDL, and blood pressure viscosity were all very significantly lower (p < 0.01), and the diastolic blood pressure was significantly higher (p < 0.05). Compared with baseline values in the exercise group, systolic blood pressure, right and left arm pulse pressure difference, right and left ankle mean arterial pressure, right and left side baPWV, right and left side ABI, TC, TG, LDL, and blood pressure viscosity decreased very significantly (p < 0.01) and diastolic blood pressure and FMD increased very significantly (p < 0.01) in the exercise group after the intervention. (4) Conclusions: In our study, a 24-week Tai Chi (BaFa WuBu) program significantly improved vascular health in middle-aged and older postmenopausal women. This simplified Tai Chi form is gentle and effective, ideal for older adults. Regular practice led to reduced vascular obstruction, improved lipid metabolism, and enhanced vascular endothelial function, crucial for preventing vascular diseases. The real-time heart rate sensors used were pivotal, enabling precise monitoring and adjustment of exercise intensity, thereby enhancing the study's scientific rigor and supporting Tai Chi (BaFa WuBu) as a beneficial therapeutic exercise.
Collapse
Affiliation(s)
- Xiaona Wang
- Chinese WuShu Academy, Beijing Sport University, Beijing 100084, China
| | - Yanli Han
- Chinese WuShu Academy, Beijing Sport University, Beijing 100084, China
| | - Haojie Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200483, China
| | - Xin Wang
- Physical Education College, Zhengzhou University, Zhengzhou 451000, China
| | - Guixian Wang
- Chinese WuShu Academy, Beijing Sport University, Beijing 100084, China
| |
Collapse
|
24
|
Arenaza‐Urquijo EM, Boyle R, Casaletto K, Anstey KJ, Vila‐Castelar C, Colverson A, Palpatzis E, Eissman JM, Kheng Siang Ng T, Raghavan S, Akinci M, Vonk JMJ, Machado LS, Zanwar PP, Shrestha HL, Wagner M, Tamburin S, Sohrabi HR, Loi S, Bartrés‐Faz D, Dubal DB, Vemuri P, Okonkwo O, Hohman TJ, Ewers M, Buckley RF, for the Reserve, Resilience and Protective Factors Professional Interest Area, Sex and Gender Professional Interest area and the ADDRESS! Special Interest Group. Sex and gender differences in cognitive resilience to aging and Alzheimer's disease. Alzheimers Dement 2024; 20:5695-5719. [PMID: 38967222 PMCID: PMC11350140 DOI: 10.1002/alz.13844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 07/06/2024]
Abstract
Sex and gender-biological and social constructs-significantly impact the prevalence of protective and risk factors, influencing the burden of Alzheimer's disease (AD; amyloid beta and tau) and other pathologies (e.g., cerebrovascular disease) which ultimately shape cognitive trajectories. Understanding the interplay of these factors is central to understanding resilience and resistance mechanisms explaining maintained cognitive function and reduced pathology accumulation in aging and AD. In this narrative review, the ADDRESS! Special Interest Group (Alzheimer's Association) adopted a multidisciplinary approach to provide the foundations and recommendations for future research into sex- and gender-specific drivers of resilience, including a sex/gender-oriented review of risk factors, genetics, AD and non-AD pathologies, brain structure and function, and animal research. We urge the field to adopt a sex/gender-aware approach to resilience to advance our understanding of the intricate interplay of biological and social determinants and consider sex/gender-specific resilience throughout disease stages. HIGHLIGHTS: Sex differences in resilience to cognitive decline vary by age and cognitive status. Initial evidence supports sex-specific distinctions in brain pathology. Findings suggest sex differences in the impact of pathology on cognition. There is a sex-specific change in resilience in the transition to clinical stages. Gender and sex factors warrant study: modifiable, immune, inflammatory, and vascular.
Collapse
Affiliation(s)
- Eider M. Arenaza‐Urquijo
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Rory Boyle
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kaitlin Casaletto
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kaarin J. Anstey
- University of New South Wales Ageing Futures InstituteSydneyNew South WalesAustralia
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Psychology, University of New South WalesSidneyNew South WalesAustralia
| | | | - Aaron Colverson
- University of Florida Center for Arts in Medicine Interdisciplinary Research LabUniversity of Florida, Center of Arts in MedicineGainesvilleFloridaUSA
| | - Eleni Palpatzis
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jaclyn M. Eissman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ted Kheng Siang Ng
- Rush Institute for Healthy Aging and Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | | | - Muge Akinci
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jet M. J. Vonk
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Luiza S. Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, FarroupilhaPorto AlegreBrazil
| | - Preeti P. Zanwar
- Jefferson College of Population Health, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- The Network on Life Course and Health Dynamics and Disparities, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Maude Wagner
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicagoIllinoisUSA
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Hamid R. Sohrabi
- Centre for Healthy AgeingHealth Future InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- School of Psychology, Murdoch UniversityMurdochWestern AustraliaAustralia
| | - Samantha Loi
- Neuropsychiatry Centre, Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of PsychiatryUniversity of MelbourneParkvilleVictoriaAustralia
| | - David Bartrés‐Faz
- Department of MedicineFaculty of Medicine and Health Sciences & Institut de NeurociènciesUniversity of BarcelonaBarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques (IDIBAPS)BarcelonaBarcelonaSpain
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la Universitat Autónoma de BarcelonaBadalonaBarcelonaSpain
| | - Dena B. Dubal
- Department of Neurology and Weill Institute of NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Biomedical and Neurosciences Graduate ProgramsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Ozioma Okonkwo
- Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchKlinikum der Universität MünchenLudwig Maximilians Universität (LMU)MunichGermany
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
| | - Rachel F. Buckley
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
25
|
Bartkowiak-Wieczorek J, Jaros A, Gajdzińska A, Wojtyła-Buciora P, Szymański I, Szymaniak J, Janusz W, Walczak I, Jonaszka G, Bienert A. The Dual Faces of Oestrogen: The Impact of Exogenous Oestrogen on the Physiological and Pathophysiological Functions of Tissues and Organs. Int J Mol Sci 2024; 25:8167. [PMID: 39125736 PMCID: PMC11311417 DOI: 10.3390/ijms25158167] [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: 06/09/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Oestrogen plays a crucial physiological role in both women and men. It regulates reproductive functions and maintains various non-reproductive tissues through its receptors, such as oestrogen receptor 1/oestrogen receptor α (ESR1/Erα), oestrogen receptor 2/oestrogen receptor β (ESR2/Erβ), and G protein-coupled oestrogen receptor 1 (GPER). This hormone is essential for the proper functioning of women's ovaries and uterus. Oestrogen supports testicular function and spermatogenesis in men and contributes to bone density, cardiovascular health, and metabolic processes in both sexes. Nuclear receptors Er-α and Er-β belong to the group of transcription activators that stimulate cell proliferation. In the environment, compounds similar in structure to the oestrogens compete with endogenous hormones for binding sites to receptors and to disrupt homeostasis. The lack of balance in oestrogen levels can lead to infertility, cancer, immunological disorders, and other conditions. Exogenous endocrine-active compounds, such as bisphenol A (BPA), phthalates, and organic phosphoric acid esters, can disrupt signalling pathways responsible for cell division and apoptosis processes. The metabolism of oestrogen and its structurally similar compounds can produce carcinogenic substances. It can also stimulate the growth of cancer cells by regulating genes crucial for cell proliferation and cell cycle progression, with long-term elevated levels linked to hormone-dependent cancers such as breast cancer. Oestrogens can also affect markers of immunological activation and contribute to the development of autoimmune diseases. Hormone replacement therapy, oral contraception, in vitro fertilisation stimulation, and hormonal stimulation of transgender people can increase the risk of breast cancer. Cortisol, similar in structure to oestrogen, can serve as a biomarker associated with the risk of developing breast cancer. The aim of this review is to analyse the sources of oestrogens and their effects on the endogenous and exogenous process of homeostasis.
Collapse
Affiliation(s)
- Joanna Bartkowiak-Wieczorek
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Agnieszka Jaros
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.J.); (A.B.)
| | - Anna Gajdzińska
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Paulina Wojtyła-Buciora
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
- Department of Social Medicine and Public Health, Calisia University, 62-800 Kalisz, Poland
| | - Igor Szymański
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Julian Szymaniak
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Wojciech Janusz
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Iga Walczak
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Gabriela Jonaszka
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Agnieszka Bienert
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.J.); (A.B.)
| |
Collapse
|
26
|
Cignarella A, Bolego C, Barton M. Sex and sex steroids as determinants of cardiovascular risk. Steroids 2024; 206:109423. [PMID: 38631602 DOI: 10.1016/j.steroids.2024.109423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/08/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
There are considerable sex differences regarding the risk of cardiovascular disease (CVD), including arterial hypertension, coronary artery disease (CAD) and stroke, as well as chronic renal disease. Women are largely protected from these conditions prior to menopause, and the risk increases following cessation of endogenous estrogen production or after surgical menopause. Cardiovascular diseases in women generally begin to occur at a later age than in men (on average with a delay of 10 years). Cessation of estrogen production also impacts metabolism, increasing the risk of developing obesity and diabetes. In middle-aged individuals, hypertension develops earlier and faster in women than in men, and smoking increases cardiovascular risk to a greater degree in women than it does in men. It is not only estrogen that affects female cardiovascular health and plays a protective role until menopause: other sex hormones such as progesterone and androgen hormones generate a complex balance that differentiates heart and blood vessel function in women compared to men. Estrogens improve vasodilation of epicardial coronary arteries and the coronary microvasculature by augmenting the release of vasodilating factors such as nitric oxide and prostacyclin, which are mechanisms of coronary vasodilatation that are more pronounced in women compared to men. Estrogens are also powerful inhibitors of inflammation, which in part explains their protective effects on CVD and chronic renal disease. Emerging evidence suggests that sex chromosomes also play a significant role in shaping cardiovascular risk. The cardiovascular protection conferred by endogenous estrogens may be extended by hormone therapy, especially using bioidentical hormones and starting treatment early after menopause.
Collapse
Affiliation(s)
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland; Andreas Grüntzig Foundation, Zürich, Switzerland.
| |
Collapse
|
27
|
Kim HL. Differences in Risk Factors for Coronary Atherosclerosis According to Sex. J Lipid Atheroscler 2024; 13:97-110. [PMID: 38826179 PMCID: PMC11140242 DOI: 10.12997/jla.2024.13.2.97] [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: 10/18/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 06/04/2024] Open
Abstract
Interest in sex differences related to coronary artery disease (CAD) has steadily increased, and the risk factors for CAD show distinct sex differences. For women, cardiovascular risk increases significantly after menopause due to a decrease in estrogen levels. In older individuals, increased arterial stiffness results in a higher pulse pressure, leading to a more common occurrence of isolated systolic hypertension; these changes are more noticeable in women. While the incidence of diabetes is similar in both sexes, women with diabetes face a 50% higher relative risk of fatal coronary heart disease compared to men. Smoking significantly increases the risk of ischemic heart disease in women, particularly those who are younger. The decrease in estrogen in women leads to a redistribution of fat, resulting in increased abdominal obesity and, consequently, an elevated cardiovascular risk. Pregnancy and reproductive factors also have a significant impact on CAD risks in women. Additionally, disparities exist in medical practice. Women are less likely to be prescribed cardioprotective drugs, referred for interventional or surgical treatments, or included in clinical research than men. By increasing awareness of these sex differences and addressing the disparities, we can progress toward more personalized treatment strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Hack-Lyoung Kim
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Bowie DC, Low KA, Rubenstein SL, Islam SS, Zimmerman B, Camacho PB, Sutton BP, Gratton G, Fabiani M. Neurovascular Mechanisms of Cognitive Aging: Sex-Related Differences in the Average Progression of Arteriosclerosis, White Matter Atrophy, and Cognitive Decline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.06.556562. [PMID: 38562861 PMCID: PMC10983862 DOI: 10.1101/2023.09.06.556562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Arterial stiffness (arteriosclerosis) has been linked to heightened risks for cognitive decline, and ultimately for Alzheimer's disease and other forms of dementia. Importantly, neurovascular outcomes generally vary according to one's biological sex. Here, capitalizing on a large sample of participants with neuroimaging and behavioral data ( N = 203, age range = 18-87 years), we aimed to provide support for a hierarchical model of neurocognitive aging, which links age-related declines in cerebrovascular health to the rate of cognitive decline via a series of intervening variables, such as white matter integrity. By applying a novel piecewise regression approach to our cross-sectional sample to support Granger-like causality inferences, we show that, on average, a precipitous decline in cerebral arterial elasticity (measured with diffuse optical imaging of the cerebral arterial pulse; pulse-DOT) temporally precedes an acceleration in the development of white matter lesions by nearly a decade, with women protected from these deleterious effects until approximately age 50, the average onset of menopause. By employing multiple-mediator path analyses while controlling for sex, we show that age may impair cognition via the sequential indirect effects of arteriosclerosis and white matter atrophy on fluid, but not crystallized, abilities. Importantly, we replicate these results using pulse pressure, an independent index of arterial health, thereby providing converging evidence for the central role of arteriosclerosis as an accelerating factor in normal and pathological aging and identifying robust sex-related differences in the progression of cerebral arteriosclerosis and white matter degradation.
Collapse
|
29
|
Agarwal N, St. John J, Van Iterson EH, Laffin LJ. Association of pulse pressure with death, myocardial infarction, and stroke among cardiovascular outcome trial participants. Am J Prev Cardiol 2024; 17:100623. [PMID: 38144432 PMCID: PMC10746405 DOI: 10.1016/j.ajpc.2023.100623] [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: 08/05/2023] [Revised: 11/07/2023] [Accepted: 11/25/2023] [Indexed: 12/26/2023] Open
Abstract
Background Prior evidence demonstrates that pulse pressure (PP), a surrogate marker of arterial stiffness, is an independent risk factor for mortality and major adverse cardiovascular (CV) events. Objectives The study aimed to identify the association of PP with death, myocardial infarction, and stroke among participants enrolled in large CV outcome clinical trials and determine if this association was impacted by pre-existing CV disease, or specific CV risk factors. Methods A total of 65,382 individuals, ages 19 to 98 years, that were enrolled in one of five CV outcome trials were analyzed. Baseline demographics, history, blood pressures, and medications were collected. Univariate and multivariable analyses were conducted to explore temporal patterns, risks, and adjusted survival rates. Results Mean baseline PP was 52 ± 12 mmHg. For every 10 mmHg increase in PP, there was an increased risk of death, stroke, or myocardial infarction (hazard ratio (HR) 1.11, 95 % CI 1.08 to 1.14, p < 0.001). Similarly, a PP ≥ 60 mmHg demonstrated an HR of 1.27 (95 % CI 1.19 to 1.36, p < 0.001) compared with PP < 60 mmHg. A similar association existed for all subgroups analyzed except for participants with a history of stroke where increasing PP did not increase risk (HR 1.02, 95 % CI 0.95 to 1.10, p = 0.53). PP was a better predictor of adverse outcomes when compared to both systolic and diastolic blood pressures using the AIC and C-index. Conclusions Among participants enrolled in CV outcome trials, baseline PP is associated with increased risk of death, myocardial infarction, and stroke for those with pre-existing CV disease and risk factors with the exception of a prior history of stroke.
Collapse
Affiliation(s)
- Neel Agarwal
- Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Julie St. John
- Cleveland Clinic, C5Research, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Erik H. Van Iterson
- Cleveland Clinic, Section of Preventive Cardiology and Rehabilitation, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Luke J. Laffin
- Cleveland Clinic, C5Research, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Cleveland Clinic, Section of Preventive Cardiology and Rehabilitation, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
30
|
Bai X, Wang Y, Luo X, Bao X, Weng X, Chen Y, Zhang S, Lv Y, Dai X, Zeng M, Yang D, Hu S, Li J, Ji Y, Jia H, Yu B. Cigarette tar accelerates atherosclerosis progression via RIPK3-dependent necroptosis mediated by endoplasmic reticulum stress in vascular smooth muscle cells. Cell Commun Signal 2024; 22:41. [PMID: 38229167 PMCID: PMC10790416 DOI: 10.1186/s12964-024-01480-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/05/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Tar is the main toxic of cigarettes, and its effect on atherosclerosis progression and the underlying mechanisms remain largely unknown. Vascular smooth muscle cells (VSMCs) play a key role in atherogenesis and plaque vulnerability. The present study sought to investigate the mechanism of atherosclerosis progression through tar-induced VSMC necroptosis, a recently described form of necrosis. METHODS The effect of tar on atherosclerosis progression and VSMC necroptosis was examined in ApoE-/- mice and cultured VSMCs. The role of necroptosis in tar-induced plaque development was evaluated in RIPK3-deletion mice (ApoE-/-RIPK3-/-). The key proteins of necroptosis in carotid plaques of smokers and non-smokers were also examined. Quantitative proteomics of mice aortas was conducted to further investigate the underlying mechanism. Pharmacological approaches were then applied to modulate the expression of targets to verify the regulatory process of tar-induced necroptosis. RESULTS Tar administration led to increased atherosclerotic plaque area and reduced collagen and VSMCs in ApoE-/- mice. The expression of RIPK1、RIPK3、and MLKL in VSMCs of plaques were all increased in tar-exposed mice and smokers. RIPK3 deletion protected against VSMC loss and plaque progression stimulated by tar. In mechanistic studies, quantitative proteomics analysis of ApoE-/- mice aortas suggested that tar triggered endoplasmic reticulum (ER) stress. PERK-eIF2α-CHOP axis was activated in tar-treated VSMCs and atherosclerotic plaque. Inhibition of ER stress using 4PBA significantly reduced plaque progression and VSMC necroptosis. Further study revealed that ER stress resulted in calcium (Ca2+) release into mitochondria and cytoplasm. Elevated Ca2+ levels lead to mitochondrial dysfunction and excessive reactive oxygen species (ROS) production, which consequently promote RIPK3-dependent necroptosis. In addition, Ca2+/calmodulin-dependent protein kinase II (CaMKII) activated by cytosolic Ca2+ overload binds to RIPK3, accounting for necroptosis. CONCLUSION The findings revealed that cigarette tar promoted atherosclerosis progression by inducing RIPK3-dependent VSMC necroptosis and identified novel avenues of ER stress and Ca2+ overload.
Collapse
Affiliation(s)
- Xiaoxuan Bai
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Ying Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xing Luo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xiaoyi Bao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xiuzhu Weng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Yuwu Chen
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Shan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Ying Lv
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xinyu Dai
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Ming Zeng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Dan Yang
- Department of Forensic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Sining Hu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Ji Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Yong Ji
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Key Laboratory of Cardiovascular Medicine Research and NHC Key Laboratory of Cell Transplantation, Harbin, 150001, China
| | - Haibo Jia
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China.
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China.
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
31
|
Choradia A, Bai K, Soni S, Nguyen N, Adhikari S, Rahul DK, Gupta R. Beyond hot flashes: Exploring the role of estrogen therapy in postmenopausal women for myocardial infarction prevention and recovery. BIOMOLECULES & BIOMEDICINE 2024; 24:4-13. [PMID: 37650466 PMCID: PMC10787608 DOI: 10.17305/bb.2023.9535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
Myocardial infarction (MI) commonly known as "heart attack" results from the blockage of blood flow to the heart. Postmenopausal women face an elevated risk of MI due to declining estrogen levels, a hormone pivotal in maintaining cardiovascular health. It promotes vasodilation, reduce inflammation, and improves lipid profiles. While estrogen therapy shows promise in mitigating MI risk for postmenopausal woman, its efficacy in prevention and recovery remains a subject of debate. This review provides a critical assessment of existing evidence on estrogen therapy's cardioprotective effects for postmenopausal women. It delves into estrogen's role in vascular function enhancement, inflammation reduction, and lipid metabolism modulation. Additionally, it addresses the various forms of estrogen therapy, administration methods, dosage considerations, safety implications, and associated risks. The review highlights the existing controversies and knowledge gaps related to estrogen therapy for MI prevention. It underscores the urgency for in-depth research to decipher the nexus between estrogen therapy and MI risk, especially concerning primary prevention and specific postmenopausal subgroups. Future studies should investigate optimal formulations, doses, and administration routes of estrogen therapy as well as assess treatment timing and duration. Comparative studies and long-term follow-up are necessary to inform clinical decision-making and improve patient care. Addressing these research gaps will empower clinicians to make more judicious choices about estrogen therapy for MI prevention and recovery in postmenopausal women, aiming for enhanced patient outcomes.
Collapse
Affiliation(s)
| | | | - Suha Soni
- University of Texas Health Science Center School of Public Health, Texas, USA
| | | | | | | | | |
Collapse
|
32
|
Alake SE, Ice J, Robinson K, Price P, Hatter B, Wozniak K, Lin D, Chowanadisai W, Smith BJ, Lucas EA. Reduced estrogen signaling contributes to bone loss and cardiac dysfunction in interleukin-10 knockout mice. Physiol Rep 2024; 12:e15914. [PMID: 38217044 PMCID: PMC10787104 DOI: 10.14814/phy2.15914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
Characterization of the interleukin (IL)-10 knockout (KO) mouse with chronic gut inflammation, cardiovascular dysfunction, and bone loss suggests a critical role for this cytokine in interorgan communication within the gut, bone, and cardiovascular axis. We sought to understand the role of IL-10 in the cross-talk between these systems. Six-week-old IL-10 KO mice and their wild type (WT) counterparts were maintained on a standard rodent diet for 3 or 6 months. Gene expression of proinflammatory markers and Fgf23, serum 17β-estradiol (E2), and cardiac protein expression were assessed. Ileal Il17a and Tnf mRNA increased while Il6 mRNA increased in the bone and heart by at least 2-fold in IL-10 KO mice. Bone Dmp1 and Phex mRNA were repressed at 6 months in IL-10 KO mice, resulting in increased Fgf23 mRNA (~4-fold) that contributed to increased fibrosis. In the IL-10 KO mice, gut bacterial β-glucuronidase activity and ovarian Cyp19a1 mRNA were lower (p < 0.05), consistent with reduced serum E2 and reduced cardiac pNOS3 (Ser1119 ) in these mice. Treatment of ileal lymphocytes with E2 reduced gut inflammation in WT but not IL-10 KO mice. In conclusion, our data suggest that diminished estrogen and defective bone mineralization increased FGF23 which contributed to cardiac fibrosis in the IL-10 KO mouse.
Collapse
Affiliation(s)
- Sanmi E. Alake
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - John Ice
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Kara Robinson
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Payton Price
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Bethany Hatter
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Karen Wozniak
- Department of Microbiology and Molecular GeneticsOklahoma State UniversityStillwaterOklahomaUSA
| | - Dingbo Lin
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Winyoo Chowanadisai
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Brenda J. Smith
- Department of Obstetrics and GynecologyIndiana School of MedicineIndianapolisIndianaUSA
- Indiana Center for Musculoskeletal HealthIndiana School of MedicineIndianapolisIndianaUSA
| | - Edralin A. Lucas
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| |
Collapse
|
33
|
Cote S, Perron TL, Baillargeon JP, Bocti C, Lepage JF, Whittingstall K. Association of Cumulative Lifetime Exposure to Female Hormones With Cerebral Small Vessel Disease in Postmenopausal Women in the UK Biobank. Neurology 2023; 101:e1970-e1978. [PMID: 37758482 PMCID: PMC10662980 DOI: 10.1212/wnl.0000000000207845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/03/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Rates of cerebrovascular disease increase after menopause, which is often attributed to the absence of hormones. It remains unknown whether the cumulative exposure to hormones across a female person's premenopausal life extends the window of cerebrovascular protection to the postmenopausal period. To investigate this, we examined the relationship between lifetime hormone exposure (LHE) and cerebral small vessel disease in more than 9,000 postmenopausal women in the UK-Biobank. METHODS The cohort consisted of women (aged 40-69 years) who attended one of 22 research centers across the United Kingdom between 2006 and 2010. Women were excluded if they were premenopausal when scanned, had missing reproductive history data, self-reported neurologic disorders, brain cancer, cerebral vascular incidents, head or neurologic injury, and nervous system infection. Endogenous LHE (LHEEndo) was estimated by summing the number of years pregnant (LHEParity) with the duration of the reproductive period (LHECycle = age menopause - age menarche). Exogenous LHE (LHEExo) was estimated by summing the number of years on oral contraceptives and hormone replacement therapy. Cerebral small vessel disease was determined by estimating white matter hyperintensity volume (WMHV) from T2-fluid-attenuated inversion recovery brain MRI (acquired between 2014 and 2021), normalized to intracranial volume and log-transformed. Multiple linear regressions were used to assess the relationship between LHEEndo on WMHV adjusted for age, cardiovascular risk factors, sociodemographics, and LHEExo. RESULTS A total of 9,163 postmenopausal women (age 64.21 ± 6.81 years) were retained for analysis. Average LHEEndo was 39.77 ± 3.59 years. Women with higher LHEEndo showed smaller WMHV (adj-R 2 = 0.307, LHEEndo β = -0.007 [-0.012 to -0.002], p < 0.01). LHEParity and LHECycle were independent contributors to WMHV (adj-R 2 = 0.308, p << 0.001; LHEParity β = -0.022 [-0.042 to -0.002], p < 0.05; LHECycle β = -0.006 [-0.011 to -0.001], p < 0.05). LHEExo was not significantly related to WMHV (LHEExo β = 0.001 [-0.001 to 0.002], p > 0.05). DISCUSSION Women with more prolonged exposure to endogenous hormones show relatively smaller burden of cerebral small vessel disease independent of the history of oral contraceptive use or hormone replacement therapy. Our results highlight the critical role endogenous hormones play in female brain health and provide real-world evidence of the protective effects premenopausal endogenous hormone exposure plays on postmenopausal cerebrovascular health.
Collapse
Affiliation(s)
- Samantha Cote
- From the Department of Nuclear Medicine and Radiobiology (S.C.), Division of Neurology (T.-L.P., C.B.) and Endocrinology Division (J.-P.B.), Department of Medicine, Department of Pediatrics (J.-F.L.), and Diagnostic Radiology (K.W.), Department of Medicine, Université de Sherbrooke, Quebec, Canada
| | - Thomas-Louis Perron
- From the Department of Nuclear Medicine and Radiobiology (S.C.), Division of Neurology (T.-L.P., C.B.) and Endocrinology Division (J.-P.B.), Department of Medicine, Department of Pediatrics (J.-F.L.), and Diagnostic Radiology (K.W.), Department of Medicine, Université de Sherbrooke, Quebec, Canada
| | - Jean-Patrice Baillargeon
- From the Department of Nuclear Medicine and Radiobiology (S.C.), Division of Neurology (T.-L.P., C.B.) and Endocrinology Division (J.-P.B.), Department of Medicine, Department of Pediatrics (J.-F.L.), and Diagnostic Radiology (K.W.), Department of Medicine, Université de Sherbrooke, Quebec, Canada
| | - Christian Bocti
- From the Department of Nuclear Medicine and Radiobiology (S.C.), Division of Neurology (T.-L.P., C.B.) and Endocrinology Division (J.-P.B.), Department of Medicine, Department of Pediatrics (J.-F.L.), and Diagnostic Radiology (K.W.), Department of Medicine, Université de Sherbrooke, Quebec, Canada
| | - Jean-Francois Lepage
- From the Department of Nuclear Medicine and Radiobiology (S.C.), Division of Neurology (T.-L.P., C.B.) and Endocrinology Division (J.-P.B.), Department of Medicine, Department of Pediatrics (J.-F.L.), and Diagnostic Radiology (K.W.), Department of Medicine, Université de Sherbrooke, Quebec, Canada
| | - Kevin Whittingstall
- From the Department of Nuclear Medicine and Radiobiology (S.C.), Division of Neurology (T.-L.P., C.B.) and Endocrinology Division (J.-P.B.), Department of Medicine, Department of Pediatrics (J.-F.L.), and Diagnostic Radiology (K.W.), Department of Medicine, Université de Sherbrooke, Quebec, Canada.
| |
Collapse
|
34
|
Thachil J, Lassila R. What can historical literature on von Willebrand disease teach us? Res Pract Thromb Haemost 2023; 7:102244. [PMID: 38193051 PMCID: PMC10772893 DOI: 10.1016/j.rpth.2023.102244] [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: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 01/10/2024] Open
Abstract
Dr Erik von Willebrand first described a family with bleeding symptoms in a Finnish publication in 1926. A closer look at this landmark publication sheds light on some pathophysiological aspects of von Willebrand disease that may be applicable even in the current era. We attempt to relay in this article how the teachings from this original description may provide a benchmark for further research in this condition.
Collapse
Affiliation(s)
- Jecko Thachil
- Department of Haematology, Manchester University Hospitals, Manchester, United Kingdom
- University of Manchester, Manchester, United Kingdom
| | - Riitta Lassila
- Research Program Unit in Systems Oncology, Oncosys, Medical Faculty, University of Helsinki, Helsinki, Finland
- Coagulation Disorders Unit, Department of Hematology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
35
|
Wits M, Becher C, de Man F, Sanchez-Duffhues G, Goumans MJ. Sex-biased TGFβ signalling in pulmonary arterial hypertension. Cardiovasc Res 2023; 119:2262-2277. [PMID: 37595264 PMCID: PMC10597641 DOI: 10.1093/cvr/cvad129] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare cardiovascular disorder leading to pulmonary hypertension and, often fatal, right heart failure. Sex differences in PAH are evident, which primarily presents with a female predominance and increased male severity. Disturbed signalling of the transforming growth factor-β (TGFβ) family and gene mutations in the bone morphogenetic protein receptor 2 (BMPR2) are risk factors for PAH development, but how sex-specific cues affect the TGFβ family signalling in PAH remains poorly understood. In this review, we aim to explore the sex bias in PAH by examining sex differences in the TGFβ signalling family through mechanistical and translational evidence. Sex hormones including oestrogens, progestogens, and androgens, can determine the expression of receptors (including BMPR2), ligands, and soluble antagonists within the TGFβ family in a tissue-specific manner. Furthermore, sex-related genetic processes, i.e. Y-chromosome expression and X-chromosome inactivation, can influence the TGFβ signalling family at multiple levels. Given the clinical and mechanistical similarities, we expect that the conclusions arising from this review may apply also to hereditary haemorrhagic telangiectasia (HHT), a rare vascular disorder affecting the TGFβ signalling family pathway. In summary, we anticipate that investigating the TGFβ signalling family in a sex-specific manner will contribute to further understand the underlying processes leading to PAH and likely HHT.
Collapse
Affiliation(s)
- Marius Wits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Clarissa Becher
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Frances de Man
- Department of Pulmonary Medicine, Amsterdam University Medical Center (UMC) (Vrije Universiteit), 1081 HV Amsterdam, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
36
|
Huckaby LV, Leshnower BG. Sex and Gender Differences in Aortic Disease. US CARDIOLOGY REVIEW 2023; 17:e14. [PMID: 39559522 PMCID: PMC11571386 DOI: 10.15420/usc.2022.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 07/06/2023] [Indexed: 11/20/2024] Open
Abstract
Dilatation of the thoracic or abdominal aorta can progress to dissection or rupture with significant associated morbidity and mortality. Aortic disease remains a treatable contributor to mortality in the US and its burden is likely underestimated. Recent clinical studies have uncovered sex and gender distinctions in the epidemiology, pathophysiology, and outcomes of aortic disease. Despite this, there has been little progress in the application of these findings to clinical practice. Improved understanding of the sex-specific mechanisms of aortic disease may inform personalized indications for elective repair and thus reduce the morbidity of aortic catastrophe. The objective of this review is to summarize known clinical and biological sex differences in both thoracic and abdominal aortic disease and highlight promising areas for future investigation.
Collapse
|
37
|
Saddic L, Escopete S, Zilberberg L, Kalsow S, Gupta D, Eghbali M, Parker S. 17 β-Estradiol Impedes Aortic Root Dilation and Rupture in Male Marfan Mice. Int J Mol Sci 2023; 24:13571. [PMID: 37686377 PMCID: PMC10487461 DOI: 10.3390/ijms241713571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Marfan syndrome causes a hereditary form of thoracic aortic aneurysms with worse outcomes in male compared to female patients. In this study, we examine the effects of 17 β-estradiol on aortic dilation and rupture in a Marfan mouse model. Marfan male mice were administered 17 β-estradiol, and the growth in the aortic root, along with the risk of aortic rupture, was measured. Transcriptomic profiling was used to identify enriched pathways from 17 β-estradiol treatments. Aortic smooth muscle cells were then treated with cytokines to validate functional mechanisms. We show that 17 β-estradiol decreased the size and rate of aortic root dilation and improved survival from rupture. The Marfan transcriptome was enriched in inflammatory genes, and the addition of 17 β-estradiol modulated a set of genes that function through TNFα mediated NF-κB signaling. In addition, 17 β-estradiol suppressed the induction of these TNFα induced genes in aortic smooth muscle cells in vitro in an NF-κB dependent manner, and 17 β-estradiol decreased the formation of adventitial inflammatory foci in aortic roots in vivo. In conclusion, 17 β-estradiol protects against the dilation and rupture of aortic roots in Marfan male mice through the inhibition of TNFα-NF-κB signaling.
Collapse
Affiliation(s)
- Louis Saddic
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA (M.E.)
| | - Sean Escopete
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA (L.Z.); (S.K.); (D.G.)
| | - Lior Zilberberg
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA (L.Z.); (S.K.); (D.G.)
| | - Shannon Kalsow
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA (L.Z.); (S.K.); (D.G.)
| | - Divya Gupta
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA (L.Z.); (S.K.); (D.G.)
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA (M.E.)
| | - Sarah Parker
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA (L.Z.); (S.K.); (D.G.)
| |
Collapse
|
38
|
Lira-Silva E, del Valle Mondragón L, Pérez-Torres I, Posadas-Sánchez R, Roldán Gómez FJ, Posadas-Romero C, Vargas-Barrón J, Pavón N. Possible implication of estrogenic compounds on heart disease in menopausal women. Biomed Pharmacother 2023; 162:114649. [PMID: 37023620 DOI: 10.1016/j.biopha.2023.114649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Epidemiological studies imply there is a higher risk of cardiovascular disease in menopausal women. Some explanations suggest a lack of estrogens as the cause, but estrogens do not disappear completely and are just transformed into different products called estrogenic degradation metabolites (EDMs). When estrogens are metabolized, reactive oxygen species (ROS) increase, causing DNA damage and increasing oxidative stress. These conditions are associated to neurodegenerative diseases and different types of cancer. However, their effect on the cardiovascular system remains unknown. This paper compares estrogenic metabolite levels in serum from post-menopausal women with cardiovascular risk (CAC>1) and with establish cardiovascular disease (CVD), against levels in healthy women (Ctrl). Sample sera were obtained from the Genetics of Atherosclerotic Disease (GEA) Mexican Study. Serum levels of eleven estrogenic metabolites were quantified by High performance liquid chromatography (HPLC) and oxidative stress markers such as ROS, lipoperoxidation levels (TBARS), total antioxidant capacity (TAC), super oxide dismutase activity (SOD) and cytokine levels were evaluated. 8-hydroxy-2-deoxyguanosine (8-OHdG) was also determined as a marker of nuclear damage.There were significant differences between serum levels of some EDMs in CAC> 1 and CVD vs. serum levels in Ctrl women. Results also revealed an increase in oxidative stress and a diminished capacity to manage oxidative stress. These findings provide an overview, and suggest that some estrogenic metabolites may be associated with an increased risk of CVD in menopausal women. However, additional studies are needed to evaluate the impact of these EDMs directly on cardiovascular function.
Collapse
|
39
|
Saddic L, Escopete S, Zilberberg L, Kalsow S, Gupta D, Egbhali M, Parker S. 17 β-estradiol impedes aortic root dilation and rupture in male Marfan mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.540071. [PMID: 37215011 PMCID: PMC10197695 DOI: 10.1101/2023.05.09.540071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Marfan syndrome causes a hereditary form of thoracic aortic aneurysms with dilation of the aortic root. Human and animal models suggest a worse phenotype for males compared to females with respect to aneurysm size and risk of dissection. In this study we examine the effects of 17 β-estradiol on aortic dilation and rupture in a Marfan mouse model. Marfan male mice were administered 17 β-estradiol and the growth in aortic root size along with the risk of aortic rupture or dissection with the addition of angiotensin II was measured. Transcriptomic profiling was used to identify enriched pathways from 17 β-estradiol treatment. Aortic smooth muscle cells were then treated with cytokines in order to validate the mechanism of 17 β-estradiol protection. We show that 17 β-estradiol decreased the size and rate of aortic root dilation and improved survival from rupture and dissection after treatment with angiotensin II. The Marfan transcriptome was enriched in inflammatory genes and the addition of 17 β-estradiol modulated a set of genes that function through TNFα mediated NF-κB signaling. These included many proteins known to play a role in the phenotypic shift of aortic smooth muscle cells from a contractile to a more inflammatory-like state such as Vcam-1, Mcp-1, Lgals3, Il-6, Il-1b, and C3. In addition, 17 β-estradiol suppressed the induction of these TNFα induced genes in aortic smooth muscle cells in vitro and this effect appears to be NF-κB dependent. In conclusion, 17 β-estradiol protects against the dilation and rupture of aortic roots in Marfan male mice through the inhibition of TNFα -NF-κB signaling and thus prevents the phenotypic switch of aortic smooth muscle cells from a contractile to an inflammatory state.
Collapse
Affiliation(s)
- Louis Saddic
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Sean Escopete
- Cedars-Sinai Medical Center, Smidt Heart Institute, Department of Cardiology, Los Angeles, California
| | - Lior Zilberberg
- Cedars-Sinai Medical Center, Smidt Heart Institute, Department of Cardiology, Los Angeles, California
| | - Shannon Kalsow
- Cedars-Sinai Medical Center, Smidt Heart Institute, Department of Cardiology, Los Angeles, California
| | - Divya Gupta
- Cedars-Sinai Medical Center, Smidt Heart Institute, Department of Cardiology, Los Angeles, California
| | - Mansoureh Egbhali
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Sarah Parker
- Cedars-Sinai Medical Center, Smidt Heart Institute, Department of Cardiology, Los Angeles, California
| |
Collapse
|
40
|
Kommaddi RP, Verma A, Muniz-Terrera G, Tiwari V, Chithanathan K, Diwakar L, Gowaikar R, Karunakaran S, Malo PK, Graff-Radford NR, Day GS, Laske C, Vöglein J, Nübling G, Ikeuchi T, Kasuga K, Ravindranath V. Sex difference in evolution of cognitive decline: studies on mouse model and the Dominantly Inherited Alzheimer Network cohort. Transl Psychiatry 2023; 13:123. [PMID: 37045867 PMCID: PMC10097702 DOI: 10.1038/s41398-023-02411-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Women carry a higher burden of Alzheimer's disease (AD) compared to men, which is not accounted entirely by differences in lifespan. To identify the mechanisms underlying this effect, we investigated sex-specific differences in the progression of familial AD in humans and in APPswe/PS1ΔE9 mice. Activity dependent protein translation and associative learning and memory deficits were examined in APPswe/PS1ΔE9 mice and wild-type mice. As a human comparator group, progression of cognitive dysfunction was assessed in mutation carriers and non-carriers from DIAN (Dominantly Inherited Alzheimer Network) cohort. Female APPswe/PS1ΔE9 mice did not show recall deficits after contextual fear conditioning until 8 months of age. Further, activity dependent protein translation and Akt1-mTOR signaling at the synapse were impaired in male but not in female mice until 8 months of age. Ovariectomized APPswe/PS1ΔE9 mice displayed recall deficits at 4 months of age and these were sustained until 8 months of age. Moreover, activity dependent protein translation was also impaired in 4 months old ovariectomized APPswe/PS1ΔE9 mice compared with sham female APPswe/PS1ΔE9 mice. Progression of memory impairment differed between men and women in the DIAN cohort as analyzed using linear mixed effects model, wherein men showed steeper cognitive decline irrespective of the age of entry in the study, while women showed significantly greater performance and slower decline in immediate recall (LOGIMEM) and delayed recall (MEMUNITS) than men. However, when the performance of men and women in several cognitive tasks (such as Wechsler's logical memory) are compared with the estimated year from expected symptom onset (EYO) we found no significant differences between men and women. We conclude that in familial AD patients and mouse models, females are protected, and the onset of disease is delayed as long as estrogen levels are intact.
Collapse
Affiliation(s)
- Reddy Peera Kommaddi
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India.
| | - Aditi Verma
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Graciela Muniz-Terrera
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland, UK
- The Department of Social Medicine, Ohio University, Athens, OH, 45701, USA
| | - Vivek Tiwari
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | | | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Ruturaj Gowaikar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Smitha Karunakaran
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| | - Palash Kumar Malo
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Neill R Graff-Radford
- Department of Neurology, Mayo Clinic Florida, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Mayo Clinic College of Medicine and Science, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Christoph Laske
- German Center for Neurodegenerative Diseases, Munich, Germany
- Section for Dementia Research, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Georg Nübling
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8585, Japan
| | - Vijayalakshmi Ravindranath
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
41
|
Ormeloxifene, a selective estrogen receptor modulator, protects against pulmonary hypertension. Eur J Pharmacol 2023; 943:175558. [PMID: 36731722 DOI: 10.1016/j.ejphar.2023.175558] [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: 10/19/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023]
Abstract
PURPOSE Protective effect of 17β-estradiol is well-known in pulmonary hypertension. However, estrogen-based therapy may potentially increase the risk of breast cancer, necessitating a search for novel drugs. This study, therefore, investigated the ameliorative effects of a selective estrogen receptor modulator, ormeloxifene, in pulmonary hypertension. METHODS Cardiomyocytes (H9C2) and human pulmonary arterial smooth muscle cells (HPASMCs) were exposed to hypoxia (1% O2) for 42 and 96 h, respectively, with or without ormeloxifene pre-treatment (1 μM). Also, female (ovary-intact or ovariectomized) and male Sprague-Dawley rats received monocrotaline (60 mg/kg, once, subcutaneously), with or without ormeloxifene treatment (2.5 mg/kg, orally) for four weeks. RESULTS Hypoxia dysregulated 17β-hydroxysteroid dehydrogenase (17βHSD) 1 & 2 expressions, reducing 17β-estradiol production and estrogen receptors α and β in HPASMC but increasing estrone, proliferation, inflammation, oxidative stress, and mitochondrial dysfunction. Similarly, monocrotaline decreased plasma 17β-estradiol and uterine weight in ovary-intact rats. Further, monocrotaline altered 17βHSD1 & 2 expressions and reduced estrogen receptors α and β, increasing right ventricular pressure, proliferation, inflammation, oxidative stress, endothelial dysfunction, mitochondrial dysfunction, and vascular remodeling in female and male rats, with worsened conditions in ovariectomized rats. Ormeloxifene was less uterotrophic; however, it attenuated both hypoxia and monocrotaline effects by improving pulmonary 17β-estradiol synthesis. Furthermore, ormeloxifene decreased cardiac hypertrophy and right ventricular remodeling induced by hypoxia and monocrotaline. CONCLUSION This study demonstrates that ormeloxifene promoted pulmonary 17β-estradiol synthesis, alleviated inflammation, improved the NOX4/HO1/Nrf/PPARγ/PGC-1α axis, and attenuated pulmonary hypertension. It is evidently safe at tested concentrations and may be effectively repurposed for pulmonary hypertension treatment.
Collapse
|
42
|
Sex differences in the association between systemic oxidative stress status and optic nerve head blood flow in normal-tension glaucoma. PLoS One 2023; 18:e0282047. [PMID: 36827337 PMCID: PMC9955941 DOI: 10.1371/journal.pone.0282047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
PURPOSE To investigate the association of systemic oxidative stress markers and optic nerve head (ONH) blood flow in normal-tension glaucoma (NTG) patients, as well as sex differences in this association. METHODS This was a cross-sectional study of 235 eyes with NTG of 134 patients (56 male, 78 female; mean age, 60.9±14.1 years). Laser speckle flowgraphy (LSFG) was used to measure ONH blood flow (mean blur rate in the tissue area of the ONH; MBR-T) and LSFG pulse-waveform parameters, including flow acceleration index in the tissue area of the ONH (FAI-T). Oxidative stress markers, diacron-reactive oxygen metabolites (d-ROMs), and biological antioxidant potential (BAP) were measured with a free radical elective evaluator. Spearman's rank correlation test and a multivariate linear mixed-effect model were used to investigate factors associated with ONH blood flow. RESULTS MBR-T was significantly correlated with age (rs = -0.28, p < 0.001), mean arterial pressure (rs = -0.20, p = 0.002), intraocular pressure (rs = 0.24, p < 0.001), peripapillary retinal nerve fiber layer thickness (rs = 0.62, p < 0.001), and disc area (rs = -0.26, p < 0.001), but not with serum d-ROM level. Separate analyses of the subjects divided by sex showed that BAP was positively correlated to MBR-T (rs = 0.21, p = 0.036) and FAI-T (rs = 0.36, p < 0.001) only in male subjects. Similarly, BAP was significantly associated with MBR-T (β = 0.25, p = 0.026) and FAI-T (β = 0.37, p < 0.001) in male subjects in a multivariate linear mixed-effect model. CONCLUSION A lower serum antioxidant level, as indicated by BAP, was associated with reduced ONH blood flow only in male NTG patients. Our findings suggest that there are sex differences in the involvement of oxidative stress in the pathogenesis of reduced ocular blood flow in NTG.
Collapse
|
43
|
Ezzatvar Y, García-Hermoso A. Global estimates of diabetes-related amputations incidence in 2010-2020: A systematic review and meta-analysis. Diabetes Res Clin Pract 2023; 195:110194. [PMID: 36464091 DOI: 10.1016/j.diabres.2022.110194] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
AIMS/HYPOTHESIS This study sought to provide up-to-date pooled global estimates of diabetes-related amputation incidence from 2010 to 2020. METHODS Embase and Medline databases were searched for studies reporting the incidence rate (IR) of diabetes-related amputations from 2010 to 2020. IR estimates of diabetes-related amputations with associated 95% confidence interval (CI) per 100,000 individuals with diabetes were calculated. RESULTS 23 studies were included, reporting 505,390 diabetes-related lower extremity amputations. IR of minor amputations was 139.97 (95% CI 88.18-222.16) per 100,000 individuals with diabetes, among patients with type 1 diabetes was 148.59 (95% CI 65.00-339.68) and in type 2 diabetes was 75.53 (95% CI 29.94-190.54). IR of major amputations was 94.82 (95% CI 56.62-158.80) per 100,000 individuals with diabetes, among patients with type 1 diabetes was 100.76 (95% CI 53.71-189.01) and among type 2 diabetes was 40.58 (95% CI 11.03-149.28). There were 83.84 annual amputations (95% CI 41.67-168.65) per 100,000 women with diabetes and 178.04 (95% CI 81.16-390.55) per 100,000 men. CONCLUSIONS Globally, annual incidence of diabetes-related amputations from 2010 to 2020 has shown to disproportionately affect men and individuals with type 1 diabetes mellitus, although its incidence is not uniform across countries.
Collapse
Affiliation(s)
- Yasmin Ezzatvar
- Department of Nursing, Universitat de València, Valencia, Spain.
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| |
Collapse
|
44
|
Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Sex-specific and hormone-related differences in vascular remodelling in atherosclerosis. Eur J Clin Invest 2023; 53:e13885. [PMID: 36219492 DOI: 10.1111/eci.13885] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022]
Abstract
Atherosclerosis, a lipid-driven inflammatory disease, is the main underlying cause of cardiovascular diseases (CVDs) both in men and women. Sex-related dimorphisms regarding CVDs and atherosclerosis were observed since more than a decade ago. Inflammatory mediators such as cytokines, but also endothelial dysfunction, vascular smooth muscle cell migration and proliferation lead to vascular remodelling but are differentially affected by sex. Each year a greater number of men die of CVDs compared with women and are also affected by CVDs at an earlier age (40-70 years old) while women develop atherosclerosis-related complications mainly after menopause (60+ years). The exact biological reasons behind this discrepancy are still not well-understood. From the numerous animal studies on atherosclerosis, only a few include both sexes and even less investigate and highlight the sex-specific differences that may arise. Endogenous sex hormones such as testosterone and oestrogen modulate the atherosclerotic plaque composition and the frequency of such plaques. In men, testosterone seems to act like a double-edged sword as its decrease with ageing correlates with an increased risk of atherosclerotic CVDs, while testosterone is also reported to promote inflammatory immune cell recruitment into the atherosclerotic plaque. In premenopausal women, oestrogen exerts anti-atherosclerotic effects, which decline together with its level after menopause resulting in increased CVD risk in ageing women. However, the interplay of sex hormones, sex-specific immune responses and other sex-related factors is still incompletely understood. This review highlights reported sex differences in atherosclerotic vascular remodelling and the role of endogenous sex hormones in this process.
Collapse
Affiliation(s)
- Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
45
|
Low expression of ESR1 correlates with ascending aortic dilation and acute type A aortic dissection. Gene 2023; 851:147001. [DOI: 10.1016/j.gene.2022.147001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022]
|
46
|
Chen MC, Chang JJ, Chen MF, Wang TY, Huang CE, Lee KD, Chen CY. Risk of critical limb ischemia in long-term uterine cancer survivors: A population-based study. World J Clin Cases 2022; 10:13293-13303. [PMID: 36683627 PMCID: PMC9851000 DOI: 10.12998/wjcc.v10.i36.13293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/19/2022] [Accepted: 12/05/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The risk of critical limb ischemia (CLI) which causes ischemic pain or ischemic loss in the arteries of the lower extremities in long-term uterine cancer (UC) survivors remains unclear, especially in Asian patients, who are younger at the diagnosis of UC than their Western counterparts. AIM To conduct a nationwide population-based study to assess the risk of CLI in UC long-term survivors. METHODS UC survivors, defined as those who survived for longer than 5 years after the diagnosis, were identified and matched at a 1:4 ratio with normal controls. Stratified Cox models were used to assess the risk of CLI. RESULTS From 2000 to 2005, 1889 UC survivors who received surgery alone or surgery combined with radiotherapy (RT) were classified into younger (onset age < 50 years, n = 894) and older (onset age ≥ 50 years, n = 995) groups. While compared with normal controls, the younger patients with diabetes, hypertension, and receiving hormone replacement therapy (HRT) were more likely to develop CLI. In contrast, the risk of CLI was associated with adjuvant RT, obesity, hypertension, and HRT in the older group. Among the UC survivors, those who were diagnosed at an advanced age (> 65 years, aHR = 2.48, P = 0.011), had hypertension (aHR = 2.18, P = 0.008) or received HRT (aHR = 3.52, P = 0.020) were at a higher risk of CLI. CONCLUSION In this nationwide study, we found that the risk factors associated with CLI were similar in both cohorts except for adjuvant RT that was negligible in the younger group, but positive in the older group. Among the survivors, hypertension, advanced age, and HRT were more hazardous than RT. Secondary prevention should include CLI as a late complication in UC survivorship programs.
Collapse
Affiliation(s)
- Min-Chi Chen
- Department of Public Health, Chang Gung University, Taoyuan 333, Taiwan
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chiayi 612, Taiwan
| | - Jung-Jung Chang
- Division of Cardiovascular Disease, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi 612, Taiwan
| | - Miao-Fen Chen
- Department of Radiation Oncology, Change Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Ting-Yao Wang
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi 612, Taiwan
| | - Cih-En Huang
- Division of Hematology and Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi 612, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Kuan-Der Lee
- Department of Medicine, Taipei Medical University Hospital, Taipei 100, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Chao-Yu Chen
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chiayi 612, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Early Childhood Care and Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821, Taiwan
| |
Collapse
|
47
|
Early Changes in Androgen Levels in Individuals with Spinal Cord Injury: A Longitudinal SwiSCI Study. J Clin Med 2022; 11:jcm11216559. [DOI: 10.3390/jcm11216559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/18/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
We aimed to explore longitudinal changes in androgen levels in individuals with spinal cord injury (SCI) within initial inpatient rehabilitation stay and identify clinical/injury characteristics associated with hormone levels. Linear regression analysis was applied to explore the association between personal/injury characteristics and androgen hormones (total testosterone, free testosterone, sex hormone-binding globulin (SHBG), dehydroepiandrosterone (DHEA), and dehydroepiandrosterone sulfate (DHEA-S)) at admission to rehabilitation. Longitudinal changes in androgen levels were studied using linear mixed models. Analyses were stratified by sex and by injury type. We included 70 men and 16 women with SCI. We observed a non-linear association between age, time since injury, and androgens at baseline. At admission to initial rehabilitation, mature serum SHBG (full-length, protein form which lacks the N-terminal signaling peptide) was higher, while DHEA and DHEA-S were lower among opioid users vs. non-users. Serum levels of total testosterone and DHEA-S increased over rehabilitation period [β 3.96 (95%CI 1.37, 6.56), p = 0.003] and [β 1.77 (95%CI 0.73, 2.81), p = 0.01], respectively. We observed no significant changes in other androgens. Restricting our analysis to men with traumatic injury did not materially change our findings. During first inpatient rehabilitation over a median follow up of 5.6 months, we observed an increase in total testosterone and DHEA-S in men with SCI. Future studies need to explore whether these hormonal changes influence neurological and functional recovery as well as metabolic parameters during initial rehabilitation stay.
Collapse
|
48
|
Raguindin PF, Cardona I, Muka T, Lambrinoudaki I, Gebhard C, Franco OH, Marques‐Vidal P, Glisic M. Does reproductive stage impact cardiovascular disease risk factors? Results from a population-based cohort in Lausanne (CoLaus study). Clin Endocrinol (Oxf) 2022; 97:568-580. [PMID: 35377481 PMCID: PMC9790265 DOI: 10.1111/cen.14730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/30/2022]
Abstract
CONTEXT Menopause has been associated with adverse cardiovascular disease (CVD) risk profile, yet it is unclear whether the changes in CVD risk factors differ by reproductive stage independently of underlying ageing trajectories. DESIGN The CoLaus study is a prospective population-based cohort study in Lausanne, Switzerland. PATIENTS We used data from women at baseline and follow-up (mean: 5.6 ± 0.5 years) from 2003 to 2012 who did not use hormone therapy. We classified women into (i) premenopausal, (ii) menopausal transition, (iii) early (≤5 years) and (iv) late (>5 years) postmenopausal by comparing their menstruation status at baseline and follow-up. MEASUREMENTS We measured fasting lipids, glucose and cardiovascular inflammatory markers. We used repeated measures (linear mixed models) for longitudinal analysis, using premenopausal women as a reference category. We adjusted analyses for age, medications and lifestyle factors. RESULTS We used the data from 1710 women aged 35-75 years. Longitudinal analysis showed that the changes in CVD risk factors were not different in the other three menopausal categories compared to premenopausal women. When age was used as a predictor variable and adjusted for menopause status, most CVD risk factors increased, while interleukin-6 and interleukin-1β decreased with advancing age. CONCLUSION The current study suggests that women have a worsening cardiovascular risk profile as they age, and although menopausal women may have higher levels of cardiovascular risk factors compared to premenopausal women at any given time, the 5-year changes in cardiovascular risk factors may not depend on the reproductive stage.
Collapse
Affiliation(s)
- Peter Francis Raguindin
- Institute of Social and Preventive Medicine (ISPM)University of BernBernSwitzerland
- Swiss Paraplegic ResearchNottwilSwitzerland
- Graduate School for Health SciencesUniversity of BernBernSwitzerland
| | - Isabel Cardona
- Institute of Social and Preventive Medicine (ISPM)University of BernBernSwitzerland
| | - Taulant Muka
- Institute of Social and Preventive Medicine (ISPM)University of BernBernSwitzerland
| | - Irene Lambrinoudaki
- 2nd Department of Obstetrics and Gynecology, Medical School, Aretaieio HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Catherine Gebhard
- Department of Nuclear MedicineUniversity Hospital ZurichZurichSwitzerland
| | - Oscar H. Franco
- Institute of Social and Preventive Medicine (ISPM)University of BernBernSwitzerland
| | - Pedro Marques‐Vidal
- Department of Nuclear Medicine, Lausanne University Hospital (CHUV)University of LausanneLausanneSwitzerland
| | - Marija Glisic
- Institute of Social and Preventive Medicine (ISPM)University of BernBernSwitzerland
- Swiss Paraplegic ResearchNottwilSwitzerland
| |
Collapse
|
49
|
Mildly elevated diastolic blood pressure increases subsequent risk of breast cancer in postmenopausal women in the Health Examinees-Gem study. Sci Rep 2022; 12:15995. [PMID: 36163474 PMCID: PMC9512811 DOI: 10.1038/s41598-022-19705-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
Epidemiological evidence suggests that hypertension is associated with breast cancer risk. However, previous studies disregard blood pressure components in the healthy population. We aimed to examine the relationship between systolic and diastolic blood pressure and breast cancer risk in a Korean population-based prospective cohort. A total of 73,031 women from the Health Examinees Gem Study were followed from baseline (2004 to 2013) through 2018. Systolic and diastolic blood pressure were measured by trainee physicians at baseline recruitment and then categorized based on the international guidelines for clinical hypertension. Associations between systolic and diastolic blood pressure with overall breast cancer and stratified by premenopausal and postmenopausal status were evaluated using adjusted multivariable Cox proportional hazard regression. A total of 858 breast cancer cases were recorded for a median follow-up period of 9 years. Compared with the normal DBP category (< 85 mmHg), the normal-high category was positively associated with breast cancer risk in postmenopausal women (85–89 mmHg, HR 1.73 95% CI 1.28–2.33), but not in premenopausal women (85–89 mmHg, HR 0.87 95% CI 0.56–1.35). Similar results were found when all cases of self-reported hypertension were excluded. Results for SBP did not show a significant association with breast cancer risk. The association between DBP and breast cancer suggests DBP could be an important factor in cancer prevention, especially for women after menopause. Our study provides a first detailed approach to understanding the importance of diastolic blood pressure for breast cancer prevention and warrants further investigation.
Collapse
|
50
|
Sabetta A, Lombardi L, Stefanini L. Sex differences at the platelet-vascular interface. Intern Emerg Med 2022; 17:1267-1276. [PMID: 35576047 PMCID: PMC9352612 DOI: 10.1007/s11739-022-02994-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022]
Abstract
Platelets are multifunctional cells that ensure the integrity of the vascular wall and modulate the immune response at the blood/vascular interface. Their pathological activation results in both thrombosis and inflammation and implicates them in the pathogenesis of vascular disease. Vascular diseases are sexually dimorphic in terms of incidence, clinical presentation, outcome, and efficacy of anti-platelet therapy. We here provide an overview of what is known about the role of platelets in the initiation and progression of vascular diseases and summarize what is known about the sex differences in platelet reactivity and in the thromboinflammatory mechanisms that drive these diseases, with a particular focus on atherosclerosis, obstructive and non-obstructive coronary artery disease, and ischemic stroke. Understanding the sex differences at the platelet-vascular interface is clinically relevant as it will enable: (1) to design new therapeutic strategies that prevent the detrimental effects of the immune-modulatory function of platelets taking sex into account, and (2) to evaluate if sex-specific anti-platelet drug regimens should be used to reduce the risk not only of thrombosis but also of vascular disease progression.
Collapse
Affiliation(s)
- Annamaria Sabetta
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università, 37, 00185, Rome, Italy
| | - Ludovica Lombardi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università, 37, 00185, Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università, 37, 00185, Rome, Italy.
| |
Collapse
|