1
|
Wayne N, Singamneni VS, Venkatesh R, Cherlin T, Verma SS, Guerraty MA. Genetic Insights Into Coronary Microvascular Disease. Microcirculation 2025; 32:e12896. [PMID: 39755372 DOI: 10.1111/micc.12896] [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: 09/18/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Coronary microvascular disease (CMVD) affects the coronary pre-arterioles, arterioles, and capillaries and can lead to blood supply-demand mismatch and cardiac ischemia. CMVD can present clinically as ischemia or myocardial infarction with no obstructive coronary arteries (INOCA or MINOCA, respectively). Currently, therapeutic options for CMVD are limited, and there are no targeted therapies. Genetic studies have emerged as an important tool to gain rapid insights into the molecular mechanisms of human diseases. For example, coronary artery disease (CAD) genome-wide association studies (GWAS) have enrolled hundreds of thousands of patients and have identified > 320 loci, elucidating CAD pathogenic pathways and helping to identify therapeutic targets. Here, we review the current landscape of genetic studies of CMVD, consisting mostly of genotype-first approaches. We then present the hypothesis that CAD GWAS have enrolled heterogenous populations and may be better characterized as ischemic heart disease (IHD) GWAS. We discuss how several of the genetic loci currently associated with CAD may be involved in the pathogenesis of CMVD. Genetic studies could help accelerate progress in understanding CMVD pathophysiology and identifying putative therapeutic targets. Larger phenotype-first genomic studies into CMVD with adequate sex and ancestry representation are needed. Given the extensive CAD genetic and functional validation data, future research should leverage these loci as springboards for CMVD genomic research.
Collapse
Affiliation(s)
- Nicole Wayne
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Venkata S Singamneni
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rasika Venkatesh
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tess Cherlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Marie A Guerraty
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Kalaria R, Englund E. Neuropathological features of cerebrovascular diseases. Pathology 2024:S0031-3025(24)00291-5. [PMID: 39718486 DOI: 10.1016/j.pathol.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 12/25/2024]
Abstract
Optimal blood flow through a patent cerebral circulation is critical for supply of oxygen and nutrients for brain function. The integrity of vascular elements within arterial vessels of any calibre can be compromised by various disease processes. Pathological changes in the walls of veins and the venous system may also alter the dynamics of cerebral perfusion. The consequences of both systemic vascular and cerebrovascular diseases range from acute focal changes to irreversible chronic restructuring of the brain parenchyma. Cerebral infarcts of different sizes may instigate a cascade of programmed cell death mechanisms including autophagy and mitophagy and processes that range from necroptosis to ferroptosis. Recent advances also emphasise the role of the vascular inflammasome in the pathology of cerebral infarction. Here, we summarise current knowledge on frequencies, epidemiological features and the neuropathology of common cerebrovascular disorders among which cerebral small vessel diseases have become of particular interest. We also highlight the current spectrum of monogenic and polygenic genetic disorders affecting the intracranial vasculature. With the advent of DNA screening technologies, it is now realised that several cerebrovascular disorders exhibit strong genetic traits. Whilst several gene defects and their aberrant products are identified, the precise role or mechanisms of how they influence angiogenesis, vasculogenesis, vessel integrity or the extracellular matrix remain largely unclear. Despite such genetic advances, histopathological examination remains the gold standard for diagnosis and characterisation of most cerebrovascular disorders.
Collapse
Affiliation(s)
- Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom.
| | - Elisabet Englund
- Institutionen för kliniska vetenskaper, Lunds Universitet, Klinisk Patologi & Medicinsk Service, Region Skåne, Lund, Sweden
| |
Collapse
|
3
|
Aherrahrou R, Reinberger T, Hashmi S, Erdmann J. GWAS breakthroughs: mapping the journey from one locus to 393 significant coronary artery disease associations. Cardiovasc Res 2024; 120:1508-1530. [PMID: 39073758 DOI: 10.1093/cvr/cvae161] [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: 01/27/2024] [Revised: 03/20/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Coronary artery disease (CAD) poses a substantial threat to global health, leading to significant morbidity and mortality worldwide. It has a significant genetic component that has been studied through genome-wide association studies (GWAS) over the past 17 years. These studies have made progress with larger sample sizes, diverse ancestral backgrounds, and the discovery of multiple genomic regions related to CAD risk. In this review, we provide a comprehensive overview of CAD GWAS, including information about the genetic makeup of the disease and the importance of ethnic diversity in these studies. We also discuss challenges of identifying causal genes and variants within GWAS loci with a focus on non-coding regions. Additionally, we highlight tissues and cell types relevant to CAD, and discuss clinical implications of GWAS findings including polygenic risk scores, sex-specific differences in CAD genetics, ethnical aspects of personalized interventions, and GWAS guided drug development.
Collapse
Affiliation(s)
- Rédouane Aherrahrou
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Tobias Reinberger
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Satwat Hashmi
- Department of Biological and Biomedical Sciences, Aga Khan University, Stadium Road, 74800 Karachi, Pakistan
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| |
Collapse
|
4
|
Wang C, Geng L, Hou L. Analysis of carotid ultrasound in a high-stroke-risk population. Medicine (Baltimore) 2024; 103:e40383. [PMID: 39496038 PMCID: PMC11537612 DOI: 10.1097/md.0000000000040383] [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: 05/09/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
This study aims to explore the risk factors for carotid plaque (CP) and carotid common artery intima-media thickening (CCAIMT) and clarify the relationship between the risk factors with the number of CPs and the side of CCAIMT in a high-stroke-risk population in Qujing, Yunnan, China. Carotid ultrasonography was performed in 430 participants with high stroke risk, who were divided into different groups according to their ultrasound results. The risk factors and blood biochemical indices were recorded for assessment. The prevalence rates of CP and CCAIMT were 88.1% and 70.5%, respectively. Multivariate logistic regression analysis identified age and lack of physical exercise as risk factors of CP. Compared to participants without CP, participants who performed little physical exercise were prone to have one CP, while participants with risk factors for smoking, older age, and physical inactivity were more likely to have several CPs. Risk factors for CCAIMT were older age, male, and the levels of low density lipoprotein cholesterol. Risk factors for left CCAIMT included a history of hyperlipidemia and low density lipoprotein cholesterol, while male sex was the sole risk factor for right CCAIMT. Finally, male sex and advanced age were identified as risk factors for dual CCAIMT. The research reveals the risk factors for CP and CCAIMT, also clarifies the relationship between the risk factors, CP numbers, and the side of CCAIMT.
Collapse
Affiliation(s)
- ChunFang Wang
- Department of Neurology, Second People’s Hospital of Qujing, Qujing, China
| | - Lirong Geng
- Department of Neurology, Second People’s Hospital of Qujing, Qujing, China
| | - Lijun Hou
- Department of Neurology, Second People’s Hospital of Qujing, Qujing, China
| |
Collapse
|
5
|
Kozbenko T, Adam N, Grybas VS, Smith BJ, Alomar D, Hocking R, Abdelaziz J, Pace A, Boerma M, Azimzadeh O, Blattnig S, Hamada N, Yauk C, Wilkins R, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to abnormal vascular remodeling. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:4-30. [PMID: 39440813 DOI: 10.1002/em.22636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular diseases (CVDs) are complex, encompassing many types of heart pathophysiologies and associated etiologies. Radiotherapy studies have shown that fractionated radiation exposure at high doses (3-17 Gy) to the heart increases the incidence of CVD. However, the effects of low doses of radiation on the cardiovascular system or the effects from space travel, where radiation and microgravity are important contributors to damage, are not clearly understood. Herein, the adverse outcome pathway (AOP) framework was applied to develop an AOP to abnormal vascular remodeling from the deposition of energy. Following the creation of a preliminary pathway with the guidance of field experts and authoritative reviews, a scoping review was conducted that informed final key event (KE) selection and evaluation of the Bradford Hill criteria for the KE relationships (KERs). The AOP begins with a molecular initiating event of deposition of energy; ionization events increase oxidative stress, which when persistent concurrently causes the release of pro-inflammatory mediators, suppresses anti-inflammatory mechanisms and alters stress response signaling pathways. These KEs alter nitric oxide levels leading to endothelial dysfunction, and subsequent abnormal vascular remodeling (the adverse outcome). The work identifies evidence needed to strengthen understanding of the causal associations for the KERs, emphasizing where there are knowledge gaps and uncertainties in both qualitative and quantitative understanding. The AOP is anticipated to direct future research to better understand the effects of space on the human body and potentially develop countermeasures to better protect future space travelers.
Collapse
Affiliation(s)
- Tatiana Kozbenko
- Health Canada, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | - Amanda Pace
- Carleton University, Ottawa, Ontario, Canada
| | - Marjan Boerma
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, Neuherberg, Germany
| | | | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Carole Yauk
- University of Ottawa, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
6
|
Franza L, Caldarelli M, Villani ER, Cianci R. Sex Differences in Cardiovascular Diseases: Exploring the Role of Microbiota and Immunity. Biomedicines 2024; 12:1645. [PMID: 39200110 PMCID: PMC11352091 DOI: 10.3390/biomedicines12081645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of mortality and morbidity in Western countries, thus representing a global health concern. CVDs show different patterns in terms of the prevalence and presentation in men and women. The role of sex hormones has been extensively implicated in these sex-specific differences, due to the presence of the menstrual cycle and menopause in women. Moreover, the gut microbiota (GM) has been implicated in cardiovascular health, considering the growing evidence that it is involved in determining the development of specific diseases. In particular, gut-derived metabolites have been linked to CVDs and kidney disorders, which can in turn promote the progression of CVDs. Considering the differences in the composition of GM between men and women, it is possible that gut microbiota act as a mediator in regard to the sex disparities in CVDs. This narrative review aims to comprehensively review the interplay between sex, GM, and CVDs, discussing potential mechanisms and therapeutic options.
Collapse
Affiliation(s)
- Laura Franza
- Emergency, Anesthesiological and Reanimation Sciences Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS of Rome, 00168 Rome, Italy;
- Emergency Department, Azienda Ospedaliero-Universitaria di Modena, Largo del Pozzo, 71, 41125 Modena, Italy
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Emanuele Rocco Villani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- UOC Geriatra-Disturbi Cognitivi e Demenze, Dipartimento di Cure Primarie, AUSL Modena, 41012 Modena, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| |
Collapse
|
7
|
Lee C. Age-specific heterogeneity of genetic susceptibility to cardiovascular disease might have opposite outcomes depending on the presence of prediabetes. World J Diabetes 2024; 15:1381-1383. [PMID: 38983829 PMCID: PMC11229971 DOI: 10.4239/wjd.v15.i6.1381] [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: 02/27/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024] Open
Abstract
Examining age-specific heterogeneity of susceptibility to cardiovascular disease is also essential in individuals without prediabetes to determine its relative size and direction compared to those with prediabetes. Of particular interest, age-specific heterogeneity in genetic susceptibility may exhibit opposite directions depending on the presence or absence of prediabetes.
Collapse
Affiliation(s)
- Chaeyoung Lee
- Department of Bioinformatics and Life Science, Soongsil University, Seoul 06978, South Korea
| |
Collapse
|
8
|
McClain AK, Monteleone PP, Zoldan J. Sex in cardiovascular disease: Why this biological variable should be considered in in vitro models. SCIENCE ADVANCES 2024; 10:eadn3510. [PMID: 38728407 PMCID: PMC11086622 DOI: 10.1126/sciadv.adn3510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024]
Abstract
Cardiovascular disease (CVD), the world's leading cause of death, exhibits notable epidemiological, clinical, and pathophysiological differences between sexes. Many such differences can be linked back to cardiovascular sexual dimorphism, yet sex-specific in vitro models are still not the norm. A lack of sex reporting and apparent male bias raises the question of whether in vitro CVD models faithfully recapitulate the biology of intended treatment recipients. To ensure equitable treatment for the overlooked female patient population, sex as a biological variable (SABV) inclusion must become commonplace in CVD preclinical research. Here, we discuss the role of sex in CVD and underlying cardiovascular (patho)physiology. We review shortcomings in current SABV practices, describe the relevance of sex, and highlight emerging strategies for SABV inclusion in three major in vitro model types: primary cell, stem cell, and three-dimensional models. Last, we identify key barriers to inclusive design and suggest techniques for overcoming them.
Collapse
Affiliation(s)
- Anna K. McClain
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78751, USA
| | - Peter P. Monteleone
- Ascension Texas Cardiovascular, Austin, TX 78705, USA
- Dell School of Medicine, The University of Texas at Austin, Austin, TX 78712, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78751, USA
| |
Collapse
|
9
|
Russo I, Dun W, Mehta S, Ahmed S, Tzimas C, Fukuma N, Tsai EJ. Extracellular Matrix Instability and Chronic Inflammation Underlie Maladaptive Right Ventricular Pressure Overload Remodeling and Failure in Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.588013. [PMID: 38617374 PMCID: PMC11014567 DOI: 10.1101/2024.04.03.588013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Background Right ventricular dysfunction (RVD) portends increased death risk for heart failure (HF) and pulmonary arterial hypertension (PAH) patients, regardless of left ventricular function or etiology. In both, RVD arises from the chronic RV pressure overload, and represents advanced cardiopulmonary disease. RV remodeling responses and survival rates of HF and PAH patients, however, differ by sex. Men develop more severe RVD and die at younger ages than do women. Mechanistic details of this sexual dimorphism in RV remodeling are incompletely understood. We sought to elucidate the cardiac pathophysiology underlying the sex-specific RV remodeling phenotypes, RV failure (RVF) versus compensated RVD. Methods We subjected male (M-) and female (F-) adult mice to moderate pulmonary artery banding (PAB) for 9wks. Mice underwent serial echocardiography, cardiac MRI, RV pressure-volume loop recordings, histologic and molecular analyses. Results M-PAB developed severe RVD with RVF, increased RV collagen deposition and degradation, extracellular matrix (ECM) instability, and activation and recruitment of macrophages. Despite the same severity and chronicity of RV pressure overload, F-PAB had more stable ECM, lacked chronic inflammation, and developed mild RVD without RVF. Conclusions ECM destabilization and chronic activation of recruited macrophages are associated with maladaptive RV remodeling and RVF in male PAB mice. Adaptive RV remodeling of female PAB mice lacked these histopathologic changes. Our findings suggest that these two pathophysiologic processes likely contribute to the sexual dimorphism of RV pressure overload remodeling. Further mechanistic studies are needed to assess their pathogenic roles and potential as targets for RVD therapy and RVF prevention. CLINICAL PERSPECTIVE What is new?: In a mouse model of pure PH, males but not females showed an association between ECM instability, chronic inflammation with activation of recruited macrophages, and severe RV dysfunction and failure.What are the clinical implications?: In male HF and PH patients, enhancing ECM stability and countering the recruitment and activation of macrophages may help preserve RV function such that RVF can be prevented or delayed. Further preclinical mechanistic studies are needed to assess the therapeutic potential of such approaches. RESEARCH PERSPECTIVE What new question does this study raise? What question should be addressed next?: What mechanisms regulate RV ECM stability and macrophage recruitment and activation in response to chronic RV pressure overload? Are these regulatory mechanisms dependent upon or independent of sex hormone signaling?
Collapse
|
10
|
Vo V, Lopez G, Malay S, Roman YM. Cardiovascular Risk Factors Among Asian Americans: Perspectives on the Role of Acculturation in Cardiovascular Diseases Health Disparities. J Immigr Minor Health 2024; 26:409-420. [PMID: 37222869 DOI: 10.1007/s10903-023-01489-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
The growing prevalence of cardiovascular diseases in the United States (US) has disproportionately affected minority populations more than their white counterparts. A population that is often overlooked is the Asian American population, particularly Southeastern Asian immigrants. Despite having relatively favorable socioeconomic indicators compared to the general US population, Asian Americans, specifically Southeast Asian individuals, face a significant burden of traditional cardiovascular risk factors and are considered a high cardiovascular disease risk group. In addition, most studies have aggregated Asian populations into one major racial group rather than analyzing the different ethnicities among the Asian categorization. While some studies suggest that the acculturation process has some degree of impact on cardiovascular health, there has not been a widely-used tool to measure or ascertain the totality of acculturation. Instead, multiple proxies have been used to measure acculturation, and prior studies have argued for more culturally-tailored acculturation proxies. This paper aims to assess the implications of different acculturation measures on cardiovascular health among Asian Americans, particularly Southeastern Asian immigrants. The following proxies were expanded on in this paper: English spoken at home, length of stay in the US, religiosity and spirituality, and admixed family structures. Previous studies showed that as the length of stay in the US increases, the burden of cardiovascular risk factors increases. However, the impact of English spoken at home, religiosity, and admixed family structure are still inconclusive given the extent of current studies. While most studies suggest that an increase in acculturation increases the risk of cardiovascular disease, it is critical to note that acculturation is a multifaceted process. Therefore, more studies are necessary to appropriately examine the implications of various acculturation processes on cardiovascular risk factors in Asians, specifically Southeastern Asian individuals in the US.
Collapse
Affiliation(s)
- Victoria Vo
- Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA, 23298, USA
| | - Glydel Lopez
- Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Shravani Malay
- Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA, 23298, USA
| | - Youssef M Roman
- Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA, 23298, USA.
| |
Collapse
|
11
|
You W, Henneberg M. Modern medical services, a double-edged sword manages symptoms, but accumulates genetic background of cardiovascular diseases: A cross populational analysis of 217 countries. Health Sci Rep 2024; 7:e1828. [PMID: 38260183 PMCID: PMC10802089 DOI: 10.1002/hsr2.1828] [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: 08/29/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Background and Aims Through reduced natural selection, measured with Biological State Index (I bs), modern medicine enables most people to survive well beyond the reproductive lifespan leading to deleterious gene accumulation in population. This study explored the role of reduced natural selection in increasing cardiovascular disease (CVD) incidence worldwide. Methods Country-specific estimates of CVD incidence and the index of reduced natural selection were captured for analysis of their correlation. Aging, affluence, obesity prevalence, and urbanization were considered as the potential confounders in the analyses. Results Worldwide, I bs was significantly correlated with CVD incidence in the bivariate correlation analyses. This relationship remains when the contributing effects from aging, affluence, obesity prevalence, and urbanization are removed in partial correlation model. Multiple linear regression (enter) shows that I bs is a significant predictor of CVD incidence. Stepwise multiple linear regression selects I bs as the variable having the second greatest influence on CVD incidence after ageing. I bs showed a significantly greater correlation with CVD incidence in low- and middle-income countries (LMICs) than in high-income countries. Conclusion Worldwide, through reducing natural selection, the side effects of healthcare services may have been partially contributing to the increase of CVD incidence worldwide with special regard to LMICs.
Collapse
Affiliation(s)
- Wenpeng You
- Heart and LungRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
- Adelaide Medical SchoolThe University of AdelaideAdelaideSouth AustraliaAustralia
- CardiologyBox Hill HospitalMelbourneAustralia
| | - Maciej Henneberg
- Institute of Evolutionary MedicineUniversity of ZurichZurichSwitzerland
| |
Collapse
|
12
|
Ajayi OA, Okonkwo P, Ojo TO, Ajayi OK, Ogunsola O, Osayi E, Onwuatuelo I, Samuels JO. Time to incident hypertension and independent predictors among people living with HIV in Nigeria. Ther Adv Infect Dis 2024; 11:20499361241289800. [PMID: 39430413 PMCID: PMC11489917 DOI: 10.1177/20499361241289800] [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: 02/17/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024] Open
Abstract
Background Understanding the time to hypertension occurrence after antiretroviral treatment (ART) initiation in people living with HIV (PLHIV) and its determinants is important for designing interventions for control. Objective This study sought to estimate the median time of ART use to hypertension onset and its predictors in Nigerian PLHIV. Design A retrospective longitudinal study. Methods This retrospective review of 2503 normotensive adult PLHIV (⩾18 years) from 2004 to 2020 in two HIV clinics in Nigeria. Incident hypertension was based on clinical diagnosis or two consecutive blood pressure readings ⩾140/90 mmHg, taken during the 8 months of data collection. Survival event was defined as incident hypertension during follow-up or interview day for observed patients unless they were right censored. The Kaplan-Meier survival curve was used to estimate the survival probabilities of hypertension. The Cox proportional hazard model was fitted to identify predictors of hypertension at p < 0.05. Results A total of 2503 PLHIV was followed up. The majority were females (74.6%) and on Dolutegravir-based therapy (93.0%). About 22 (0.9%) were diabetic. Median age at ART initiation was 35 (interquartile range: 29-41) years. The median period of follow-up was 12.0 ± 3.9 years. The cumulative incidence of hypertension was 32.5% (381/2540), with an incidence rate of 40.1/1000 person-years. The median time to incident hypertension was 17.0 years (95% CI: 12.5-21.5 years). Shorter hypertension-free survival times were seen in males, those aged 60+, with diabetes, unsuppressed viral load, history of tuberculosis, other opportunistic infections, or co-trimoxazole use. Significant risk factors included male sex (adjusted odds ratio (AOR) = 1.3, 95% CI = 1.1-1.6), middle age (AOR = 2.3, 95% CI = 1.7-3.2), old age (AOR = 5.6, 95% CI = 3.9-8.4), and unsuppressed viral load (AOR = 1.9, 95% CI = 1.3-2.7). Conclusion Hypertension is commoner among PLHIV with unsuppressed viral load, males, and persons older than 40 years. Effective ART with viral suppression remains essential. Incorporating regular hypertension screening and treatment into HIV care is necessary for optimum health outcomes.
Collapse
Affiliation(s)
- Oluseye Ayodele Ajayi
- APIN Public Health Initiatives, Plot 1551, Zone E, Apo Resettlement, Apo, Abuja, Nigeria
| | - Prosper Okonkwo
- Program Department, APIN Public Health Initiatives, Headquarter Office, Abuja, Nigeria
| | - Temitope Olumuyiwa Ojo
- Department of Community Health, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | | | - Olabanjo Ogunsola
- Prevention and Community Services, APIN Public Health Initiatives, Ibadan Office, Nigeria
| | - Emmanuel Osayi
- Clinical Services, APIN Public Health Initiatives, Plateau Office, Nigeria
| | - Ifeyinwa Onwuatuelo
- Program Department, APIN Public Health Initiatives, Headquarter Office, Abuja, Nigeria
| | - Jay Osi Samuels
- Program Department, APIN Public Health Initiatives, Headquarter Office, Abuja, Nigeria
| |
Collapse
|
13
|
Zeidan RS, McElroy T, Rathor L, Martenson MS, Lin Y, Mankowski RT. Sex differences in frailty among older adults. Exp Gerontol 2023; 184:112333. [PMID: 37993077 DOI: 10.1016/j.exger.2023.112333] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
By definition, aging is a natural, gradual and continuous process. On the other hand, frailty reflects the increase in vulnerability to stressors and shortens the time without disease (health span) while longevity refers to the length of life (lifespan). The average life expectancy has significantly increased during the last few decades. A longer lifespan has been accompanied by an increase in frailty and decreased independence in older adults, with major differences existing between men and women. For example, women tend to live longer than men but also experience higher rates of frailty and disability. Sex differences prevent optimization of lifestyle interventions and therapies to effectively prevent frailty. Sex differences in frailty and aging are rooted in a complex interplay between uncontrollable (genetic, epigenetic, physiological), and controllable factors (psychosocial and lifestyle factors). Thus, understanding the underlying causes of sex differences in frailty and aging is essential for developing personalized interventions to promote healthy aging and improve quality of life in older men and women. In this review, we have discussed the key contributors and knowledge gaps related to sex differences in aging and frailty.
Collapse
Affiliation(s)
- Rola S Zeidan
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Taylor McElroy
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Laxmi Rathor
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Matthew S Martenson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Yi Lin
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| | - Robert T Mankowski
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
14
|
Najafi MT, Abedini R, Ghandi N, Seraji S, Sadeghi Y. Is the severity of alopecia areata associated with arterial stiffness? JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2023; 28:80. [PMID: 38292334 PMCID: PMC10826848 DOI: 10.4103/jrms.jrms_375_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/20/2023] [Accepted: 08/16/2023] [Indexed: 02/01/2024]
Abstract
Background This study aimed to evaluate the severity of alopecia areata (AA) associations with metabolic syndrome, body composition evaluated by bioimpedance techniques, and arterial stiffness based on pulse-wave velocity analysis. Materials and Methods This cross-sectional study was conducted on patients referred to AA Clinic at Razi Hospital in 2021 and 2022. Patients with AA with the Severity of Alopecia Tool (SALT) score above 20% and receiving no systemic therapy were included. Patient demographic and clinical information, symptoms of metabolic syndrome, and bioimpedance factors were collected, and the relationship between disease severity, metabolic syndrome, and bioimpedance indicators was evaluated. Results In this study, 59 patients were examined, with 26 (44.07%) being female and 33 (55.93%) being male. The mean age of the patients was 37.42 years (standard deviation [SD] =11.28). The severity of the disease was assessed using the SALT score, with the mean severity in terms of the percentage being 69.83% (SD = 28.57%). In the regression model, SALT score was independently related to the severity of vascular stiffness after adjusting for the effect of other variables (beta = 0.033, 95% CI = 0.009-0.057, P = 0.046). Moreover, SALT score was significantly related to metabolic syndrome after adjusting for the effect of other variables (OR = 1.035, 95% CI = 1.012-1.059, P = 0.002). Conclusion This study found that AA severity is associated with a higher chance of having metabolic syndrome and arterial stiffness which may lead to cardiovascular diseases in patients with AA, and screening patients regarding cardiometabolic diseases is mandated.
Collapse
Affiliation(s)
| | - Robabeh Abedini
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Ghandi
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadab Seraji
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Sadeghi
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Sawan MA, Steinberg RS, Sayegh MN, Devlin C, Behbahani-Nejad O, Wenger NK. Chest Pain in Women: Gender- and Sex-based Differences in the Presentation and Diagnosis of Heart Disease. US CARDIOLOGY REVIEW 2023; 17:e19. [PMID: 39559518 PMCID: PMC11571392 DOI: 10.15420/usc.2022.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 10/01/2023] [Indexed: 11/20/2024] Open
Abstract
Ischemic heart disease is a major cause of morbidity and mortality in the US, affecting both men and women significantly. The presentation of chest pain is largely similar in female and male patients, but additional non-chest pain symptoms can confound timely diagnosis in women. Management, diagnostic evaluation, and clinical outcomes for patients admitted with chest pain differ significantly between men and women, and understanding of these discrepancies is limited. The objective of this review is to familiarize readers with gender- and sex-specific differences in the presentation, diagnosis, and management of chest pain.
Collapse
Affiliation(s)
- Mariem A Sawan
- Department of Medicine, Division of Cardiology, Emory University School of MedicineAtlanta, GA
| | - Rebecca S Steinberg
- Department of Internal Medicine, Emory University School of MedicineAtlanta, GA
| | - Michael N Sayegh
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBoston, MA
| | - Christian Devlin
- Department of Internal Medicine, Emory University School of MedicineAtlanta, GA
| | - Omid Behbahani-Nejad
- Department of Medicine, Division of Cardiology, Emory University School of MedicineAtlanta, GA
| | - Nanette K Wenger
- Department of Medicine, Division of Cardiology, Emory University School of MedicineAtlanta, GA
| |
Collapse
|
16
|
Sakkers TR, Mokry M, Civelek M, Erdmann J, Pasterkamp G, Diez Benavente E, den Ruijter HM. Sex differences in the genetic and molecular mechanisms of coronary artery disease. Atherosclerosis 2023; 384:117279. [PMID: 37805337 DOI: 10.1016/j.atherosclerosis.2023.117279] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/09/2023] [Accepted: 09/01/2023] [Indexed: 10/09/2023]
Abstract
Sex differences in coronary artery disease (CAD) presentation, risk factors and prognosis have been widely studied. Similarly, studies on atherosclerosis have shown prominent sex differences in plaque biology. Our understanding of the underlying genetic and molecular mechanisms that drive these differences remains fragmented and largely understudied. Through reviewing genetic and epigenetic studies, we identified more than 40 sex-differential candidate genes (13 within known CAD loci) that may explain, at least in part, sex differences in vascular remodeling, lipid metabolism and endothelial dysfunction. Studies with transcriptomic and single-cell RNA sequencing data from atherosclerotic plaques highlight potential sex differences in smooth muscle cell and endothelial cell biology. Especially, phenotypic switching of smooth muscle cells seems to play a crucial role in female atherosclerosis. This matches the known sex differences in atherosclerotic phenotypes, with men being more prone to lipid-rich plaques, while women are more likely to develop fibrous plaques with endothelial dysfunction. To unravel the complex mechanisms that drive sex differences in CAD, increased statistical power and adjustments to study designs and analysis strategies are required. This entails increasing inclusion rates of women, performing well-defined sex-stratified analyses and the integration of multi-omics data.
Collapse
Affiliation(s)
- Tim R Sakkers
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508, GA, Utrecht, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508, GA, Utrecht, the Netherlands; Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508, GA, Utrecht, the Netherlands
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, 1335 Lee St, Charlottesville, VA, 22908, USA; Department of Biomedical Engineering, University of Virginia, 351 McCormick Road, Charlottesville, VA, 22904, USA
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508, GA, Utrecht, the Netherlands
| | - Ernest Diez Benavente
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508, GA, Utrecht, the Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3508, GA, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Marjani A, Poursharifi N, Hashemi MM, Sajedi A, Tatari M. The association of paraoxonase I gene polymorphisms Q192R (rs662) and L55M (rs854560) and its activity with metabolic syndrome components in fars ethnic group. Horm Mol Biol Clin Investig 2023; 44:295-303. [PMID: 36793191 DOI: 10.1515/hmbci-2022-0064] [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: 06/26/2022] [Accepted: 01/22/2023] [Indexed: 02/17/2023]
Abstract
OBJECTIVES Metabolic syndrome (MetS) may cause premature development of some diseases. PON1 genes may be involved in the pathogenesis of MetS. The aim of study was to evaluate the association between Q192R and L55M gene polymorphisms and its enzyme activity with the MetS components in subjects with and without MetS. METHODS Polymerase Chain Reaction and Restriction Fragment Length Polymorphism analysis were performed to determine polymorphisms of the paraoxonase1 gene in subjects with and without metabolic syndrome. Biochemical parameters were measured by spectrophotometer. RESULTS The MM, LM, and LL genotype frequencies of the PON1 L55M polymorphism were 10.5, 43.4, and 46.1%, and 22.4, 46.6, and 31% and; the QQ, QR, and RR genotype frequencies of the PON1 Q192R polymorphism were 55.4, 38.6 and 6%; and 56.5, 34.8 and 8.7% in subjects with and without MetS, respectively. The L and M allele frequencies were 68 and 53%; and 32 and 47% for PON1 L55M in subjects with and without MetS, respectively. The Q and R allele frequencies for PON1 Q192R were 74 and 26% in both groups. There were significant differences in HDL-cholesterol level and PON1 activity in the genotypes QQ, QR, and RR of the PON1 Q192R polymorphism in subjects with MetS. CONCLUSIONS The PON1 Q192R genotypes had only effected on PON1 activity and HDL-cholesterol level in subjects with MetS. Different genotypes of the PON1 Q192R seem to be important candidates to make the subjects susceptible to MetS in the Fars ethnic group.
Collapse
Affiliation(s)
- Abdoljalal Marjani
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Golestan, Gorgan, Iran
| | - Nahid Poursharifi
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Golestan, Gorgan, Iran
| | - Mohammad Mostakhdem Hashemi
- Student Research Committee, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Golestan, Gorgan, Iran
| | - Atefe Sajedi
- Student Research Committee, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Golestan, Gorgan, Iran
| | - Mahin Tatari
- Bioistatistics Counseling and Reproductive Health Research Center, Golestan University of Medical Sciences, Golestan, Gorgan, Iran
| |
Collapse
|
18
|
Oliveira GMMD, Almeida MCCD, Rassi DDC, Bragança ÉOV, Moura LZ, Arrais M, Campos MDSB, Lemke VG, Avila WS, Lucena AJGD, Almeida ALCD, Brandão AA, Ferreira ADDA, Biolo A, Macedo AVS, Falcão BDAA, Polanczyk CA, Lantieri CJB, Marques-Santos C, Freire CMV, Pellegrini D, Alexandre ERG, Braga FGM, Oliveira FMFD, Cintra FD, Costa IBSDS, Silva JSN, Carreira LTF, Magalhães LBNC, Matos LDNJD, Assad MHV, Barbosa MM, Silva MGD, Rivera MAM, Izar MCDO, Costa MENC, Paiva MSMDO, Castro MLD, Uellendahl M, Oliveira Junior MTD, Souza OFD, Costa RAD, Coutinho RQ, Silva SCTFD, Martins SM, Brandão SCS, Buglia S, Barbosa TMJDU, Nascimento TAD, Vieira T, Campagnucci VP, Chagas ACP. Position Statement on Ischemic Heart Disease - Women-Centered Health Care - 2023. Arq Bras Cardiol 2023; 120:e20230303. [PMID: 37556656 PMCID: PMC10382148 DOI: 10.36660/abc.20230303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023] Open
Affiliation(s)
| | | | | | | | | | | | | | | | - Walkiria Samuel Avila
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | | | | | | | | | - Andreia Biolo
- Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS - Brasil
| | | | | | | | | | - Celi Marques-Santos
- Universidade Tiradentes (UNIT), Aracaju, SE - Brasil
- Hospital São Lucas Rede D'Or São Luis, Aracaju, SE - Brasil
| | | | - Denise Pellegrini
- Hospital São Lucas da Pontifícia Universidade Católica do Rio Grande do Sul (PUC-RS), Porto Alegre, RS - Brasil
| | | | - Fabiana Goulart Marcondes Braga
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | | | | | | | | | - Lara Terra F Carreira
- Cardiologia Nuclear de Curitiba, Curitiba, PR - Brasil
- Hospital Pilar, Curitiba, PR - Brasil
| | | | | | | | | | | | | | | | | | | | | | - Marly Uellendahl
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP - Brasil
- DASA - Diagnósticos da América S/A, São Paulo, SP - Brasil
| | - Mucio Tavares de Oliveira Junior
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | | | | | - Ricardo Quental Coutinho
- Faculdade de Ciências Médicas da Universidade de Pernambuco (UPE), Recife, PE - Brasil
- Hospital Universitário Osvaldo Cruz da Universidade de Pernambuco (UPE), Recife, PE - Brasil
| | | | - Sílvia Marinho Martins
- Pronto Socorro Cardiológico de Pernambuco da Universidade de Pernambuco (PROCAPE/UPE), Recife, PE - Brasil
| | | | - Susimeire Buglia
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
- Instituto Dante Pazzanese de Cardiologia, São Paulo, SP - Brasil
| | | | | | - Thais Vieira
- Universidade Tiradentes (UNIT), Aracaju, SE - Brasil
- Rede D'Or, Aracaju, SE - Brasil
- Hospital Universitário da Universidade Federal de Sergipe (UFS), Aracaju, SE - Brasil
| | | | - Antonio Carlos Palandri Chagas
- Instituto do Coração (Incor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
- Centro Universitário Faculdade de Medicina ABC, Santo André, SP - Brasil
| |
Collapse
|
19
|
de Bakker M, Petersen TB, Akkerhuis KM, Harakalova M, Umans VA, Germans T, Caliskan K, Katsikis PD, van der Spek PJ, Suthahar N, de Boer RA, Rizopoulos D, Asselbergs FW, Boersma E, Kardys I. Sex-based differences in cardiovascular proteomic profiles and their associations with adverse outcomes in patients with chronic heart failure. Biol Sex Differ 2023; 14:29. [PMID: 37198662 DOI: 10.1186/s13293-023-00516-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Studies focusing on sex differences in circulating proteins in patients with heart failure with reduced ejection fraction (HFrEF) are scarce. Insight into sex-specific cardiovascular protein profiles and their associations with the risk of adverse outcomes may contribute to a better understanding of the pathophysiological processes involved in HFrEF. Moreover, it could provide a basis for the use of circulating protein measurements for prognostication in women and men, wherein the most relevant protein measurements are applied in each of the sexes. METHODS In 382 patients with HFrEF, we performed tri-monthly blood sampling (median follow-up: 25 [13-31] months). We selected all baseline samples and two samples closest to the primary endpoint (PEP: composite of cardiovascular death, heart transplantation, left ventricular assist device implantation, and HF hospitalization) or censoring. We then applied an aptamer-based multiplex proteomic assay identifying 1105 proteins previously associated with cardiovascular disease. We used linear regression models and gene-enrichment analysis to study sex-based differences in baseline levels. We used time-dependent Cox models to study differences in the prognostic value of serially measured proteins. All models were adjusted for the MAGGIC HF mortality risk score and p-values for multiple testing. RESULTS In 104 women and 278 men (mean age 62 and 64 years, respectively) cumulative PEP incidence at 30 months was 25% and 35%, respectively. At baseline, 55 (5%) out of the 1105 proteins were significantly different between women and men. The female protein profile was most strongly associated with extracellular matrix organization, while the male profile was dominated by regulation of cell death. The association of endothelin-1 (Pinteraction < 0.001) and somatostatin (Pinteraction = 0.040) with the PEP was modified by sex, independent of clinical characteristics. Endothelin-1 was more strongly associated with the PEP in men (HR 2.62 [95%CI, 1.98, 3.46], p < 0.001) compared to women (1.14 [1.01, 1.29], p = 0.036). Somatostatin was positively associated with the PEP in men (1.23 [1.10, 1.38], p < 0.001), but inversely associated in women (0.33 [0.12, 0.93], p = 0.036). CONCLUSION Baseline cardiovascular protein levels differ between women and men. However, the predictive value of repeatedly measured circulating proteins does not seem to differ except for endothelin-1 and somatostatin.
Collapse
Affiliation(s)
- Marie de Bakker
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Teun B Petersen
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Magdalena Harakalova
- Department of Cardiology, Division Heart and Lungs, Circulatory Health Research Center, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Victor A Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Kadir Caliskan
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter J van der Spek
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Navin Suthahar
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Eric Boersma
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Room Na-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Yun H, Lim JE, Lee EY. Genetic Risk Score for Prediction of Coronary Heart Disease in the Korean Genome and Epidemiology Study. Rev Cardiovasc Med 2023; 24:102. [PMID: 39076255 PMCID: PMC11273040 DOI: 10.31083/j.rcm2404102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/25/2023] [Accepted: 02/08/2023] [Indexed: 07/31/2024] Open
Abstract
Background Using a genetic risk score (GRS) to predict coronary heart disease (CHD) may detect disease earlier. The current study aims to assess whether GRS is associated with CHD incidence and whether it is clinically useful for improving prediction using traditional risk factors (TRFs) as well as family history. Methods Data from a total of 48,941 participants in the Korean Genome and Epidemiology Study were analyzed in the current study. The weighted GRS was constructed using 55 single-nucleotide polymorphisms based on published genome-wide association studies. The association of GRS with incident CHD was analyzed using Cox proportional hazard model. Discrimination and reclassification were assessed to demonstrate the clinical utility of GRS. The analyses were performed separately by sex. Results After adjusting for family history and TRFs, GRS was significantly associated with CHD incidence in men; compared to the low GRS group, men in the high GRS group had a 2.07-fold increased risk of CHD (95% confidence interval [CI]: 1.51-2.85). In men, the combination of TRFs, family history, and GRS had better performance than TRFs alone (C statistics for TRF-only model, 0.66, 95% CI, 0.64-0.69; C statistics for combination model, 0.68, 95% CI, 0.65-0.71; category-free reclassification index, 15%). In women, however, there was no significant association between GRS and CHD and no improvement between models. Conclusions GRS was associated with CHD incidence and contributed to a small improvement of CHD prediction in men. The potential clinical use of GRS may not outweigh the value of family history.
Collapse
Affiliation(s)
- Hyunok Yun
- Department of Nursing, Catholic Kkottongnae University, 28211 Cheongju, Republic of Korea
| | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, 02447 Seoul, Republic of Korea
| | - Eun Young Lee
- Department of Nursing, Catholic Kkottongnae University, 28211 Cheongju, Republic of Korea
| |
Collapse
|
21
|
Lakbar I, Einav S, Lalevée N, Martin-Loeches I, Pastene B, Leone M. Interactions between Gender and Sepsis—Implications for the Future. Microorganisms 2023; 11:microorganisms11030746. [PMID: 36985319 PMCID: PMC10058943 DOI: 10.3390/microorganisms11030746] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/15/2023] Open
Abstract
Sex and gender dimorphisms are found in a large variety of diseases, including sepsis and septic shock which are more prevalent in men than in women. Animal models show that the host response to pathogens differs in females and males. This difference is partially explained by sex polarization of the intracellular pathways responding to pathogen–cell receptor interactions. Sex hormones seem to be responsible for this polarization, although other factors, such as chromosomal effects, have yet to be investigated. In brief, females are less susceptible to sepsis and seem to recover more effectively than males. Clinical observations produce more nuanced findings, but men consistently have a higher incidence of sepsis, and some reports also claim higher mortality rates. However, variables other than hormonal differences complicate the interaction between sex and sepsis, including comorbidities as well as social and cultural differences between men and women. Conflicting data have also been reported regarding sepsis-attributable mortality rates among pregnant women, compared with non-pregnant females. We believe that unraveling sex differences in the host response to sepsis and its treatment could be the first step in personalized, phenotype-based management of patients with sepsis and septic shock.
Collapse
Affiliation(s)
- Ines Lakbar
- Department of Anesthesiology and Intensive Care Unit, Assistance Publique Hôpitaux Universitaires de Marseille, Aix-Marseille University, Hospital Nord, 13015 Marseille, France
- CEReSS, Health Service Research and Quality of Life Centre, School of Medicine-La Timone Medical, Aix-Marseille University, 13015 Marseille, France
| | - Sharon Einav
- Intensive Care Unit, Shaare Zedek Medical Center, Jerusalem 23456, Israel
- Faculty of Medicine, Hebrew University, Jerusalem 23456, Israel
| | - Nathalie Lalevée
- INSERM, INRAE, Centre for Nutrition and Cardiovascular Disease (C2VN), Aix-Marseille University, 13005 Marseille, France
| | - Ignacio Martin-Loeches
- Intensive Care Unit, Trinity Centre for Health Science HRB-Wellcome Trust, St James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Bruno Pastene
- Department of Anesthesiology and Intensive Care Unit, Assistance Publique Hôpitaux Universitaires de Marseille, Aix-Marseille University, Hospital Nord, 13015 Marseille, France
- INSERM, INRAE, Centre for Nutrition and Cardiovascular Disease (C2VN), Aix-Marseille University, 13005 Marseille, France
| | - Marc Leone
- Department of Anesthesiology and Intensive Care Unit, Assistance Publique Hôpitaux Universitaires de Marseille, Aix-Marseille University, Hospital Nord, 13015 Marseille, France
- INSERM, INRAE, Centre for Nutrition and Cardiovascular Disease (C2VN), Aix-Marseille University, 13005 Marseille, France
- Correspondence:
| |
Collapse
|
22
|
Della Peruta C, Lozanoska-Ochser B, Renzini A, Moresi V, Sanchez Riera C, Bouché M, Coletti D. Sex Differences in Inflammation and Muscle Wasting in Aging and Disease. Int J Mol Sci 2023; 24:ijms24054651. [PMID: 36902081 PMCID: PMC10003083 DOI: 10.3390/ijms24054651] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Only in recent years, thanks to a precision medicine-based approach, have treatments tailored to the sex of each patient emerged in clinical trials. In this regard, both striated muscle tissues present significant differences between the two sexes, which may have important consequences for diagnosis and therapy in aging and chronic illness. In fact, preservation of muscle mass in disease conditions correlates with survival; however, sex should be considered when protocols for the maintenance of muscle mass are designed. One obvious difference is that men have more muscle than women. Moreover, the two sexes differ in inflammation parameters, particularly in response to infection and disease. Therefore, unsurprisingly, men and women respond differently to therapies. In this review, we present an up-to-date overview on what is known about sex differences in skeletal muscle physiology and disfunction, such as disuse atrophy, age-related sarcopenia, and cachexia. In addition, we summarize sex differences in inflammation which may underly the aforementioned conditions because pro-inflammatory cytokines deeply affect muscle homeostasis. The comparison of these three conditions and their sex-related bases is interesting because different forms of muscle atrophy share common mechanisms; for instance, those responsible for protein dismantling are similar although differing in terms of kinetics, severity, and regulatory mechanisms. In pre-clinical research, exploring sexual dimorphism in disease conditions could highlight new efficacious treatments or recommend implementation of an existing one. Any protective factors discovered in one sex could be exploited to achieve lower morbidity, reduce the severity of the disease, or avoid mortality in the opposite sex. Thus, the understanding of sex-dependent responses to different forms of muscle atrophy and inflammation is of pivotal importance to design innovative, tailored, and efficient interventions.
Collapse
Affiliation(s)
- Chiara Della Peruta
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
| | - Biliana Lozanoska-Ochser
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
- Department of Medicine and Surgery, LUM University, 70010 Bari, Italy
| | - Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
| | - Viviana Moresi
- Institute of Nanotechnology (Nanotec), National Research Council (CNR), c/o Sapienza University of Rome, 00185 Roma, Italy
| | - Carles Sanchez Riera
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
| | - Marina Bouché
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
- Correspondence:
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00161 Roma, Italy
- Biological Adaptation and Ageing (B2A), Institut de Biologie Paris-Seine, Sorbonne Université, CNRS UMR 8256, Inserm U1164, 75005 Paris, France
| |
Collapse
|
23
|
Arslani K, Tontsch J, Todorov A, Gysi B, Kaufmann M, Kaufmann F, Hollinger A, Wildi K, Merdji H, Helms J, Siegemund M, Gebhard C, Gebhard CE. Temporal trends in mortality and provision of intensive care in younger women and men with acute myocardial infarction or stroke. Crit Care 2023; 27:14. [PMID: 36635740 PMCID: PMC9835383 DOI: 10.1186/s13054-022-04299-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Timely management of acute myocardial infarction (AMI) and acute stroke has undergone impressive progress during the last decade. However, it is currently unknown whether both sexes have profited equally from improved strategies. We sought to analyze sex-specific temporal trends in intensive care unit (ICU) admission and mortality in younger patients presenting with AMI or stroke in Switzerland. METHODS Retrospective analysis of temporal trends in 16,954 younger patients aged 18 to ≤ 52 years with AMI or acute stroke admitted to Swiss ICUs between 01/2008 and 12/2019. RESULTS Over a period of 12 years, ICU admissions for AMI decreased more in women than in men (- 6.4% in women versus - 4.5% in men, p < 0.001), while ICU mortality for AMI significantly increased in women (OR 1.2 [1.10-1.30], p = 0.032), but remained unchanged in men (OR 0.99 [0.94-1.03], p = 0.71). In stroke patients, ICU admission rates increased between 3.6 and 4.1% per year in both sexes, while ICU mortality tended to decrease only in women (OR 0.91 [0.85-0.95, p = 0.057], but remained essentially unaltered in men (OR 0.99 [0.94-1.03], p = 0.75). Interventions aimed at restoring tissue perfusion were more often performed in men with AMI, while no sex difference was noted in neurovascular interventions. CONCLUSION Sex and gender disparities in disease management and outcomes persist in the era of modern interventional neurology and cardiology with opposite trends observed in younger stroke and AMI patients admitted to intensive care. Although our study has several limitations, our data suggest that management and selection criteria for ICU admission, particularly in younger women with AMI, should be carefully reassessed.
Collapse
Affiliation(s)
- Ketina Arslani
- grid.410567.1Department of Cardiology, University Hospital Basel, Basel, Switzerland ,grid.4973.90000 0004 0646 7373Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Janna Tontsch
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland ,grid.410567.1Department of Anesthesiology, University Hospital Basel, Basel, Switzerland
| | - Atanas Todorov
- grid.412004.30000 0004 0478 9977Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland ,grid.7400.30000 0004 1937 0650Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Bianca Gysi
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Mark Kaufmann
- grid.410567.1Department of Anesthesiology, University Hospital Basel, Basel, Switzerland ,grid.6612.30000 0004 1937 0642University of Basel, Basel, Switzerland
| | - Fabian Kaufmann
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Alexa Hollinger
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland ,grid.6612.30000 0004 1937 0642University of Basel, Basel, Switzerland
| | - Karin Wildi
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland ,grid.1003.20000 0000 9320 7537Critical Care Research Group, The University of Queensland, Brisbane, Australia ,Cardiovascular Research Group, Basel, Switzerland
| | - Hamid Merdji
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland ,grid.11843.3f0000 0001 2157 9291Université de Strasbourg (UNISTRA), Faculté de Médecine; Hôpitaux universitaires de Strasbourg, Service de Médecine Intensive-Réanimation, Nouvel Hôpital Civil, Strasbourg, France ,grid.503388.5INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Julie Helms
- grid.11843.3f0000 0001 2157 9291Université de Strasbourg (UNISTRA), Faculté de Médecine; Hôpitaux universitaires de Strasbourg, Service de Médecine Intensive-Réanimation, Nouvel Hôpital Civil, Strasbourg, France ,grid.503388.5INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Martin Siegemund
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland ,grid.6612.30000 0004 1937 0642University of Basel, Basel, Switzerland
| | - Catherine Gebhard
- grid.412004.30000 0004 0478 9977Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland ,grid.7400.30000 0004 1937 0650Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland ,grid.411656.10000 0004 0479 0855Department of Cardiology, University Hospital Bern, Bern, Switzerland
| | - Caroline E. Gebhard
- grid.410567.1Intensive Care Unit, Department of Acute Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland ,grid.6612.30000 0004 1937 0642University of Basel, Basel, Switzerland
| | | |
Collapse
|
24
|
De Paoli M, Wood DW, Bohn MK, Pandey AK, Borowitz DK, Fang S, Patel Z, Venegas-Pino DE, Shi Y, Werstuck GH. Investigating the protective effects of estrogen on β-cell health and the progression of hyperglycemia-induced atherosclerosis. Am J Physiol Endocrinol Metab 2022; 323:E254-E266. [PMID: 35830687 DOI: 10.1152/ajpendo.00353.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sex differences in the prevalence and development of diabetes and associated cardiometabolic complications are well established. The objective of this study was to analyze the effects of estrogen on the maintenance of β-cell health/function and atherosclerosis progression, using a mouse model of hyperglycemia-induced atherosclerosis, the ApoE-/-:Ins2+/Akita mouse. ApoE-/-:Ins2+/Akita mice exhibit sexual dimorphism in the control of blood glucose levels. Male ApoE-/-:Ins2+/Akita mice are chronically hyperglycemic due to a significant reduction in pancreatic β-cell mass. Female mice are only transiently hyperglycemic, maintain β-cell mass, and blood glucose levels normalize at 35 ± 1 days of age. To determine the effects of estrogen on pancreatic β-cell health and function, ovariectomies and estrogen supplementation experiments were performed, and pancreatic health and atherosclerosis were assessed at various time points. Ovariectomized ApoE-/-:Ins2+/Akita mice developed chronic hyperglycemia with significantly reduced β-cell mass. To determine whether the observed effects on ovariectomized ApoE-/-:Ins2+/Akita mice were due to a lack of estrogens, slow-releasing estradiol pellets were inserted subcutaneously. Ovariectomized ApoE-/-:Ins2+/Akita mice treated with exogenous estradiol showed normalized blood glucose levels and maintained β-cell mass. Exogenous estradiol significantly reduced atherosclerosis in both ovariectomized female and male ApoE-/-:Ins2+/Akita mice relative to controls. Together, these findings suggest that estradiol confers significant protection to pancreatic β-cell health and can directly and indirectly slow the progression of atherosclerosis.NEW & NOTEWORTHY This study examines the effect(s) of estrogen on β cell and cardiometabolic health/function in a novel mouse model of hyperglycemia-induced atherosclerosis (ApoE-/-:Ins2+/Akita). Using a combination of estrogen deprivation (ovariectomy) and supplementation strategies, we quantify effects on glucose homeostasis and atherogenesis. Our results clearly show a protective role for estrogen on pancreatic β-cell health and function and glucose homeostasis. Furthermore, estrogen supplementation dramatically reduces atherosclerosis progression in both male and female mice.
Collapse
Affiliation(s)
- Monica De Paoli
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dempsey W Wood
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Mary K Bohn
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Arjun K Pandey
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Dana K Borowitz
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Susanna Fang
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Zinal Patel
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Daniel E Venegas-Pino
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yuanyuan Shi
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Nonterah EA, Crowther NJ, Klipstein‐Grobusch K, Oduro AR, Kavousi M, Agongo G, Anderson TJ, Asiki G, Boua PR, Choma SSR, Couper DJ, Engström G, de Graaf J, Kauhanen J, Lonn EM, Mathiesen EB, Micklesfield LK, Okazaki S, Polak JF, Rundek T, Salonen JT, Tollman SM, Tuomainen T, Grobbee DE, Ramsay M, Bots ML. Racial and Ethnic Differences in the Association Between Classical Cardiovascular Risk Factors and Common Carotid Intima-Media Thickness: An Individual Participant Data Meta-Analysis. J Am Heart Assoc 2022; 11:e023704. [PMID: 35876421 PMCID: PMC9375511 DOI: 10.1161/jaha.121.023704] [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: 10/14/2021] [Accepted: 03/21/2022] [Indexed: 01/02/2023]
Abstract
Background The major risk factors for atherosclerotic cardiovascular disease differ by race or ethnicity but have largely been defined using populations of European ancestry. Despite the rising prevalence of cardiovascular disease in Africa there are few related data from African populations. Therefore, we compared the association of established cardiovascular risk factors with carotid-intima media thickness (CIMT), a subclinical marker of atherosclerosis, between African, African American, Asian, European, and Hispanic populations. Methods and Results Cross-sectional analyses of 34 025 men and women drawn from 15 cohorts in Africa, Asia, Europe, and North America were undertaken. Classical cardiovascular risk factors were assessed and CIMT measured using B-mode ultrasound. Ethnic differences in the association of established cardiovascular risk factors with CIMT were determined using a 2-stage individual participant data meta-analysis with beta coefficients expressed as a percentage using the White population as the reference group. CIMT adjusted for risk factors was the greatest among African American populations followed by Asian, European, and Hispanic populations with African populations having the lowest mean CIMT. In all racial or ethnic groups, men had higher CIMT levels compared with women. Age, sex, body mass index, and systolic blood pressure had a significant positive association with CIMT in all races and ethnicities at varying magnitudes. When compared with European populations, the association of age, sex, and systolic blood pressure with CIMT was weaker in all races and ethnicities. Smoking (beta coefficient, 0.39; 95% CI, 0.09-0.70), body mass index (beta coefficient, 0.05; 95% CI, 0.01-0.08) and glucose (beta coefficient, 0.13; 95% CI, 0.06-0.19) had the strongest positive association with CIMT in the Asian population when compared with all other racial and ethnic groups. High-density lipoprotein-cholesterol had significant protective effects in African American (beta coefficient, -0.31; 95% CI, -0.42 to -0.21) and African (beta coefficient, -0.26; 95% CI, -0.31 to -0.19) populations only. Conclusions The strength of association between established cardiovascular risk factors and CIMT differed across the racial or ethnic groups and may be due to lifestyle risk factors and genetics. These differences have implications for race- ethnicity-specific primary prevention strategies and also give insights into the differential contribution of risk factors to the pathogenesis of cardiovascular disease. The greatest burden of subclinical atherosclerosis in African American individuals warrants further investigations.
Collapse
Affiliation(s)
- Engelbert A. Nonterah
- Navrongo Health Research CentreGhana Health ServiceNavrongoGhana
- Julius Global HealthJulius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Nigel J. Crowther
- Department of Chemical PathologyFaculty of Health SciencesNational Health Laboratory ServiceUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Kerstin Klipstein‐Grobusch
- Julius Global HealthJulius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
- Division of Epidemiology and BiostatisticsSchool of Public HealthFaculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Abraham R. Oduro
- Navrongo Health Research CentreGhana Health ServiceNavrongoGhana
| | - Maryam Kavousi
- Department of EpidemiologyErasmus University Medical CentreRotterdamthe Netherlands
| | - Godfred Agongo
- Navrongo Health Research CentreGhana Health ServiceNavrongoGhana
- Department of Biochemistry and Forensic ScienceCK Tedam University of Technology and Applied Sciences (UTAS)NavrongoGhana
| | - Todd J. Anderson
- Department of Cardiac Sciences and Libin Cardiovascular Institute of AlbertaUniversity of CalgaryAlbertaCanada
| | - Gershim Asiki
- African Population and Health Research Centre (APHRC)NairobiKenya
| | - Palwendé R. Boua
- Clinical Research Unit of NanoroInstitut de Recherché en Sciences de la SantéNanoroBurkina Faso
| | - Solomon S. R. Choma
- Department of Pathology and Medical SciencesDIMAMO Health and Demographic Surveillance SystemUniversity of LimpopoSouth Africa
| | - David J. Couper
- Collaborative Studies Coordinating CenterDepartment of BiostatisticsUniversity of North Carolina at Chapel HillNC
| | - Gunnar Engström
- Department of Clinical Sciences in MalmöLund UniversitySkåne University HospitalMalmöSweden
| | - Jacqueline de Graaf
- Division of Vascular MedicineDepartment of General Internal MedicineNijmegen University Medical CentreNijmegenthe Netherlands
| | - Jussi Kauhanen
- The Institute of Public Health and clinical NutritionSchool of MedicineFaculty of Health SciencesUniversity of Eastern Finland (UEF)HelsinkiFinland
| | - Eva M. Lonn
- Division of Cardiology and Population Health Research InstituteDepartment of MedicineMcMaster UniversityHamiltonOntarioCanada
| | - Ellisiv B. Mathiesen
- Brain and Circulation Research GroupInstitute of Clinical MedicineUniversity of TromsøNorway
| | - Lisa K. Micklesfield
- South African Medical Research Council/Developmental Pathways for Health Research Unit (DPHRU)Department of PaediatricsSchool of Clinical MedicineFaculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Shuhei Okazaki
- Department of Neurology, Stroke CenterOsaka University Graduate School of MedicineOsakaJapan
| | | | | | | | - Stephen M. Tollman
- South African Medical Research Council/Wits Rural Public Health and Health Transitions Research Unit (Agincourt)School of Public HealthFaculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Tomi‐Pekka Tuomainen
- The Institute of Public Health and clinical NutritionSchool of MedicineFaculty of Health SciencesUniversity of Eastern Finland (UEF)HelsinkiFinland
| | - Diederick E. Grobbee
- Julius Global HealthJulius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Michéle Ramsay
- Sydney Brenner Institute of Molecular BioscienceFaculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Michiel L. Bots
- Julius Global HealthJulius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | | |
Collapse
|
26
|
Abstract
Sex is a key risk factor for many types of cardiovascular disease. It is imperative to understand the mechanisms underlying sex differences to devise optimal preventive and therapeutic approaches for all individuals. Both biological sex (determined by sex chromosomes and gonadal hormones) and gender (social and cultural behaviors associated with femininity or masculinity) influence differences between men and women in disease susceptibility and pathology. Here, we focus on the application of experimental mouse models that elucidate the influence of 2 components of biological sex-sex chromosome complement (XX or XY) and gonad type (ovaries or testes). These models have revealed that in addition to well-known effects of gonadal hormones, sex chromosome complement influences cardiovascular risk factors, such as plasma cholesterol levels and adiposity, as well as the development of atherosclerosis and pulmonary hypertension. One mechanism by which sex chromosome dosage influences cardiometabolic traits is through sex-biased expression of X chromosome genes that escape X inactivation. These include chromatin-modifying enzymes that regulate gene expression throughout the genome. The identification of factors that determine sex-biased gene expression and cardiometabolic traits will expand our mechanistic understanding of cardiovascular disease processes and provide insight into sex differences that remain throughout the lifespan as gonadal hormone levels alter with age.
Collapse
Affiliation(s)
- Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA
- Department of Medicine, David Geffen School of Medicine at UCLA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Carrie B. Wiese
- Department of Human Genetics, David Geffen School of Medicine at UCLA
| |
Collapse
|
27
|
Wren G, Davies W. Sex-linked genetic mechanisms and atrial fibrillation risk. Eur J Med Genet 2022; 65:104459. [PMID: 35189376 DOI: 10.1016/j.ejmg.2022.104459] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/11/2022] [Accepted: 02/16/2022] [Indexed: 01/14/2023]
Abstract
Atrial fibrillation (AF) is a cardiac condition characterised by an irregular heartbeat, atrial pathology and an elevated downstream risk of thrombosis and heart failure, as well as neurological sequelae including stroke and dementia. The prevalence and presentation of, risk factors for, and therapeutic responses to, AF differ by sex, and this sex bias may be partially explained in terms of genetics. Here, we consider four sex-linked genetic mechanisms that may influence sex-biased phenotypes related to AF and provide examples of each: X-linked gene dosage, X-linked genomic imprinting, sex-biased autosomal gene expression, and male-limited Y-linked gene expression. We highlight novel candidate risk genes and pathways that warrant further investigation in clinical and preclinical studies. Understanding the biological basis of sex differences in AF should allow better prediction of disease risk, identification of novel risk/protective factors, and the development of more effective sex-tailored interventions.
Collapse
Affiliation(s)
| | - William Davies
- School of Psychology, Cardiff University, UK; School of Medicine, Cardiff University, UK.
| |
Collapse
|
28
|
Lau ES, Binek A, Parker SJ, Shah SH, Zanni MV, Van Eyk JE, Ho JE. Sexual Dimorphism in Cardiovascular Biomarkers: Clinical and Research Implications. Circ Res 2022; 130:578-592. [PMID: 35175850 PMCID: PMC8883873 DOI: 10.1161/circresaha.121.319916] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Sex-based differences in cardiovascular disease presentation, diagnosis, and response to therapies are well established, but mechanistic understanding and translation to clinical applications are limited. Blood-based biomarkers have become an important tool for interrogating biologic pathways. Understanding sexual dimorphism in the relationship between biomarkers and cardiovascular disease will enhance our insights into cardiovascular disease pathogenesis in women, with potential to translate to improved individualized care for men and women with or at risk for cardiovascular disease. In this review, we examine how biologic sex associates with differential levels of blood-based biomarkers and influences the effect of biomarkers on disease outcomes. We further summarize key differences in blood-based cardiovascular biomarkers along central biologic pathways, including myocardial stretch/injury, inflammation, adipose tissue metabolism, and fibrosis pathways in men versus women. Finally, we present recommendations for leveraging our current knowledge of sex differences in blood-based biomarkers for future research and clinical innovation.
Collapse
Affiliation(s)
- Emily S. Lau
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Aleksandra Binek
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sarah J. Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Svati H. Shah
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Markella V. Zanni
- Metabolism Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jennifer E. Ho
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Ischemic preconditioning protects the heart against ischemia-reperfusion injury in chronic kidney disease in both males and females. Biol Sex Differ 2021; 12:49. [PMID: 34488888 PMCID: PMC8420010 DOI: 10.1186/s13293-021-00392-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Uremic cardiomyopathy is a common cardiovascular complication of chronic kidney disease (CKD) characterized by left ventricular hypertrophy (LVH) and fibrosis enhancing the susceptibility of the heart to acute myocardial infarction. In the early stages of CKD, approximately 60% of patients are women. We aimed to investigate the influence of sex on the severity of uremic cardiomyopathy and the infarct size-limiting effect of ischemic preconditioning (IPRE) in experimental CKD. METHODS CKD was induced by 5/6 nephrectomy in 9-week-old male and female Wistar rats. Two months later, serum and urine laboratory parameters were measured to verify the development of CKD. Transthoracic echocardiography was performed to assess cardiac function and morphology. Cardiomyocyte hypertrophy and fibrosis were measured by histology. Left ventricular expression of A- and B-type natriuretic peptides (ANP and BNP) were measured by qRT-PCR and circulating BNP level was measured by ELISA. In a subgroup of animals, hearts were perfused according to Langendorff and were subjected to 35 min global ischemia and 120 min reperfusion with or without IPRE (3 × 5 min I/R cycles applied before index ischemia). Then infarct size or phosphorylated and total forms of proteins related to the cardioprotective RISK (AKT, ERK1,2) and SAFE (STAT3) pathways were measured by Western blot. RESULTS The severity of CKD was similar in males and females. However, CKD males developed more severe LVH compared to females as assessed by echocardiography. Histology revealed cardiac fibrosis only in males in CKD. LV ANP expression was significantly increased due to CKD in both sexes, however, LV BNP and circulating BNP levels failed to significantly increase in CKD. In both sexes, IPRE significantly decreased the infarct size in both the sham-operated and CKD groups. IPRE significantly increased the phospho-STAT3/STAT3 ratio in sham-operated but not in CKD animals in both sexes. There were no significant differences in phospho-AKT/AKT and phospho-ERK1,2/ERK1,2 ratios between the groups. CONCLUSION The infarct size-limiting effect of IPRE was preserved in both sexes in CKD despite the more severe uremic cardiomyopathy in male CKD rats. Further research is needed to identify crucial molecular mechanisms in the cardioprotective effect of IPRE in CKD.
Collapse
|
30
|
Helms J, De Jong A, Einav S. Yentl syndrome and the ICU. Intensive Care Med 2021; 47:594-597. [PMID: 33950371 DOI: 10.1007/s00134-021-06420-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Julie Helms
- Faculté de Médecine, Université de Strasbourg (UNISTRA), Hôpitaux Universitaires de Strasbourg, Service de Médecine Intensive-Réanimation, Nouvel Hôpital Civil, Strasbourg, France.
- Laboratoire d'ImmunoRhumatologie Moléculaire, Faculté de Médecine, Institut National de la Santé et de la Recherche Médicale (INSERM)UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NGFédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.
| | - Audrey De Jong
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, University of Montpellier, PhyMedExp, INSERM U1046, CNRS UMR, 9214, CEDEX 5, Montpellier, France
| | - Sharon Einav
- University Faculty of Medicine, Intensive Care Unit of the Shaare Zedek Medical Medical Centre and 2Hebrew, Jerusalem, Israel
| |
Collapse
|
31
|
Association of cytochrome P450 1B1 gene polymorphisms and environmental biomarkers with hypertension in Slovak midlife women. ACTA ACUST UNITED AC 2021; 27:1287-1294. [PMID: 33110045 DOI: 10.1097/gme.0000000000001605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE This study investigated the association of the Leu432Val and Asn453Ser CYP1B1 polymorphisms and selected environmental biomarkers with hypertension (HT) in Slovak midlife women. METHODS We studied 575 women. Divided according to their blood pressure status: 255 with HT and 320 without HT. All data was obtained by using standard anthropometric, genetic methods and analyzed by regression models to adjust for HT risk factors such as age, obesity, smoking, and level of education. RESULTS Our findings revealed that CYP1B1 Leu432Val polymorphism was associated with HT, whereas no association was found between Asn453Ser polymorphism and HT. Women with at least one Val allele had significantly higher odds of HT compared to women with the Leu/Leu genotype in the total sample (Exp(B) = 1.82, CI 1.16-2.84, P = 0.009). After dividing women by menopausal status and the presence of HT environmental risk factor, the association between CYP1B1 polymorphism and HT was observed in pre/perimenopausal women (Exp(B), 2.36; 95% CI 1.13-4.92; P = 0.02), smokers (Exp(B), 3.40; 95% CI 1.48-7.82; P = 0.004), abdominal obesity (Exp(B), 2.41; 95% CI 1.23-4.75; P = 0.01) and in women with only basic education (Exp(B), 4.20, 95% CI 1.12-15.71; P = 0.03). However, general linear models did not reveal a statistically significant interactions between CYP1B1, menopausal status, and HT risk factors and their common association with HT (P > 0.05). CONCLUSIONS In this pilot study, we have provided novel data that supports the significant association of CYP1B1 Leu432Val gene polymorphism with HT in Slovak midlife women.
Collapse
|
32
|
Azizi Z, Gisinger T, Bender U, Deischinger C, Raparelli V, Norris CM, Kublickiene K, Herrero MT, Emam KE, Kautzky-Willer A, Pilote L. Sex, Gender, and Cardiovascular Health in Canadian and Austrian Populations. Can J Cardiol 2021; 37:1240-1247. [PMID: 33785367 DOI: 10.1016/j.cjca.2021.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/09/2021] [Accepted: 03/07/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Evidence differentiating the effect of biological sex from psychosociocultural factors (gender) in different societies and its relation to cardiovascular diseases is scarce. We explored the association between sex, gender, and cardiovascular health (CVH) among Canadian (CAN) and Austrian (AT) populations. METHODS The Canadian Community Health Survey (CCHS) (n = 63,522; 55% female) and Austrian Health Interview Survey (AT-HIS) (n = 15,771; 56% female) were analyzed in a cross-sectional survey design. The CANHEART/ATHEART index, a measure of ideal CVH composed of 6 cardiometabolic risk factors (smoking, physical activity, fruit and vegetable consumption, overweight/obesity, diabetes, and hypertension; range 0-6; higher scores reflecting better CVH) was calculated for both databases. A composite measure of psychosociocultural gender was computed for each country (range 0-1, higher score identifying characteristics traditionally ascribed to women). RESULTS Median CANHEART 4 (interquartile range 3-5) and CAN gender scores 0.55 (0.49-0.60) were similar to median ATHEART 4 (3-5) and AT gender scores 0.55 (0.46-0.64). Although higher gender scores (CCHS: β = -1.33, 95% confidence interval [CI] -1.44 to -1.22; AT-HIS: β = -1.08, 95% CI -1.26 to -0.89)) were associated with worse CVH, female sex (CCHS: β = 0.35, 95% CI (0.33-0.37); AT-HIS: β = 0.60, 95% CI (0.55-0.64)) was associated with better CVH in both populations. In addition, higher gender scores were associated with increased prevalence of heart disease compared with female sex. The magnitude of this risk was higher in Austrians. CONCLUSIONS These results demonstrate that individuals with characteristics typically ascribed to women reported poorer cardiovascular health and higher risk of heart disease, independently from biological sex and baseline CV risk factors, in both countries. Female sex exhibited better CV health and a lower prevalence of heart disease than male in both populations. However, gender factors and magnitude of gender impact varied by country.
Collapse
Affiliation(s)
- Zahra Azizi
- Centre for Outcomes Research and Evaluation, McGill University Health Centre Research Institute, Montréal, Québec, Canada
| | - Teresa Gisinger
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Vienna, Austria
| | - Uri Bender
- Centre for Outcomes Research and Evaluation, McGill University Health Centre Research Institute, Montréal, Québec, Canada
| | - Carola Deischinger
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Vienna, Austria
| | - Valeria Raparelli
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy; Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Colleen M Norris
- Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada; Heart and Stroke Strategic Clinical Networks, Alberta Health Services, Edmonton, Alberta, Canada
| | - Karolina Kublickiene
- Section for Renal Medicine, Department of Clinical Intervention, Science, and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | | | - Khaled El Emam
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Replica Analytics, Ottawa, Ontario, Canada
| | - Alexandra Kautzky-Willer
- Gender Medicine Unit, Division of Endocrinology and Metabolism, Department of Internal Medicine III, Vienna, Austria
| | - Louise Pilote
- Centre for Outcomes Research and Evaluation, McGill University Health Centre Research Institute, Montréal, Québec, Canada; Divisions of Clinical Epidemiology and General Internal Medicine, McGill University Health Centre Research Institute, Montréal, Québec, Canada.
| | | |
Collapse
|
33
|
Abstract
Black women in the United States have experienced substantial improvements in health during the last century, yet health disparities persist. These health disparities are in large part a reflection of the inequalities experienced by Black women on a host of social and economic measures. In this paper, we examine the structural contributors to social and economic conditions that create the landscape for persistent health inequities among Black women. Demographic measures related to the health status and health (in)equity of Black women are reviewed. Current rates of specific physical and mental health outcomes are examined in more depth, including maternal mortality and chronic conditions associated with maternal morbidity. We conclude by highlighting the necessity of social and economic equity among Black women for health equity to be achieved.
Collapse
Affiliation(s)
- Juanita J. Chinn
- Population Dynamics Branch, Division of Extramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Iman K. Martin
- Blood Epidemiology and Clinical Therapeutics Branch, Division of Blood Diseases and Resources, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Nicole Redmond
- Clinical Applications and Prevention Branch, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
34
|
Ye Y, Chen X, Han J, Jiang W, Natarajan P, Zhao H. Interactions Between Enhanced Polygenic Risk Scores and Lifestyle for Cardiovascular Disease, Diabetes, and Lipid Levels. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003128. [PMID: 33433237 DOI: 10.1161/circgen.120.003128] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Both lifestyle and genetic factors confer risk for cardiovascular diseases, type 2 diabetes, and dyslipidemia. However, the interactions between these 2 groups of risk factors were not comprehensively understood due to previous poor estimation of genetic risk. Here we set out to develop enhanced polygenic risk scores (PRS) and systematically investigate multiplicative and additive interactions between PRS and lifestyle for coronary artery disease, atrial fibrillation, type 2 diabetes, total cholesterol, triglyceride, and LDL-cholesterol. METHODS Our study included 276 096 unrelated White British participants from the UK Biobank. We investigated several PRS methods (P+T, LDpred, PRS continuous shrinkage, and AnnoPred) and showed that AnnoPred achieved consistently improved prediction accuracy for all 6 diseases/traits. With enhanced PRS and combined lifestyle status categorized by smoking, body mass index, physical activity, and diet, we investigated both multiplicative and additive interactions between PRS and lifestyle using regression models. RESULTS We observed that healthy lifestyle reduced disease incidence by similar multiplicative magnitude across different PRS groups. The absolute risk reduction from lifestyle adherence was, however, significantly greater in individuals with higher PRS. Specifically, for type 2 diabetes, the absolute risk reduction from lifestyle adherence was 12.4% (95% CI, 10.0%-14.9%) in the top 1% PRS versus 2.8% (95% CI, 2.3%-3.3%) in the bottom PRS decile, leading to a ratio of >4.4. We also observed a significant interaction effect between PRS and lifestyle on triglyceride level. CONCLUSIONS By leveraging functional annotations, AnnoPred outperforms state-of-the-art methods on quantifying genetic risk through PRS. Our analyses based on enhanced PRS suggest that individuals with high genetic risk may derive similar relative but greater absolute benefit from lifestyle adherence.
Collapse
Affiliation(s)
- Yixuan Ye
- Program of Computational Biology and Bioinformatics (Y.Y., H.Z.), Yale University
| | - Xi Chen
- Department of Statistics and Data Science (X.C., J.H.), Yale University.,Department of Molecular Biophysics and Biochemistry (X.C., J.H.), Yale University
| | - James Han
- Department of Statistics and Data Science (X.C., J.H.), Yale University.,Department of Molecular Biophysics and Biochemistry (X.C., J.H.), Yale University
| | - Wei Jiang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT (W.J., H.Z.)
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.).,Program in Medical and Population Genetics, Broad Institute of Harvard & Massachusetts Institute of Technology, Cambridge, MA (P.N.)
| | - Hongyu Zhao
- Program of Computational Biology and Bioinformatics (Y.Y., H.Z.), Yale University.,Department of Biostatistics, Yale School of Public Health, New Haven, CT (W.J., H.Z.)
| |
Collapse
|
35
|
The Y Chromosome: A Complex Locus for Genetic Analyses of Complex Human Traits. Genes (Basel) 2020; 11:genes11111273. [PMID: 33137877 PMCID: PMC7693691 DOI: 10.3390/genes11111273] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/19/2020] [Accepted: 10/26/2020] [Indexed: 12/29/2022] Open
Abstract
The Human Y chromosome (ChrY) has been demonstrated to be a powerful tool for phylogenetics, population genetics, genetic genealogy and forensics. However, the importance of ChrY genetic variation in relation to human complex traits is less clear. In this review, we summarise existing evidence about the inherent complexities of ChrY variation and their use in association studies of human complex traits. We present and discuss the specific particularities of ChrY genetic variation, including Y chromosomal haplogroups, that need to be considered in the design and interpretation of genetic epidemiological studies involving ChrY.
Collapse
|
36
|
Anderson WD, Soh JY, Innis SE, Dimanche A, Ma L, Langefeld CD, Comeau ME, Das SK, Schadt EE, Björkegren JLM, Civelek M. Sex differences in human adipose tissue gene expression and genetic regulation involve adipogenesis. Genome Res 2020; 30:1379-1392. [PMID: 32967914 PMCID: PMC7605264 DOI: 10.1101/gr.264614.120] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Sex differences in adipose tissue distribution and function are associated with sex differences in cardiometabolic disease. While many studies have revealed sex differences in adipocyte cell signaling and physiology, there is a relative dearth of information regarding sex differences in transcript abundance and regulation. We investigated sex differences in subcutaneous adipose tissue transcriptional regulation using omic-scale data from ∼3000 geographically and ethnically diverse human samples. We identified 162 genes with robust sex differences in expression. Differentially expressed genes were implicated in oxidative phosphorylation and adipogenesis. We further determined that sex differences in gene expression levels could be related to sex differences in the genetics of gene expression regulation. Our analyses revealed sex-specific genetic associations, and this finding was replicated in a study of 98 inbred mouse strains. The genes under genetic regulation in human and mouse were enriched for oxidative phosphorylation and adipogenesis. Enrichment analysis showed that the associated genetic loci resided within binding motifs for adipogenic transcription factors (e.g., PPARG and EGR1). We demonstrated that sex differences in gene expression could be influenced by sex differences in genetic regulation for six genes (e.g., FADS1 and MAP1B). These genes exhibited dynamic expression patterns during adipogenesis and robust expression in mature human adipocytes. Our results support a role for adipogenesis-related genes in subcutaneous adipose tissue sex differences in the genetic and environmental regulation of gene expression.
Collapse
Affiliation(s)
- Warren D Anderson
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Joon Yuhl Soh
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Sarah E Innis
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Alexis Dimanche
- Physics Department, Southwestern University, Georgetown, Texas 78626, USA
| | - Lijiang Ma
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Mary E Comeau
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Swapan K Das
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Eric E Schadt
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Johan L M Björkegren
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA
| |
Collapse
|
37
|
Sex-related impacts on clinical outcomes after percutaneous coronary intervention. Sci Rep 2020; 10:15262. [PMID: 32943716 PMCID: PMC7498594 DOI: 10.1038/s41598-020-72296-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/28/2020] [Indexed: 11/16/2022] Open
Abstract
The aim of this study is to investigate sex-related impacts on clinical outcomes after percutaneous coronary intervention (PCI). We analyzed 90,305 patients (29.0% of women) with the first episode of coronary artery disease who underwent PCI from the Korean National Health Insurance claims database between July 2013 and June 2017. Women were significantly older than men (71.5 ± 10.5 vs. 61.8 ± 11.7 years, p < 0.001). The study population had a median follow-up of 2.2 years (interquartile range, 1.2–3.3). In the propensity-score matched angina population (15,104 pairs), the in-hospital mortality of women was not different from men (odds ratio, 0.87; 95% confidence interval: 0.71–1.08, p = 0.202). However, the post-discharge mortality of women was significantly lower (hazard ratio, 0.74; 95% confidence interval: 0.69–0.80, p < 0.001) than that of men. In the propensity-score matched acute myocardial infarction (AMI) population (8,775 pairs), the in-hospital mortality of women was significantly higher than that of men (odds ratio, 1.19; 95% confidence interval: 1.05–1.34, p = 0.006). Meanwhile, there was no difference in mortality after discharge (hazard ratio, 0.98; 95% confidence interval: 0.91–1.06, p = 0.605). The post-discharge mortality of women was not higher than men under the contemporary PCI treatment. Altered sex-related impacts on clinical outcomes might be attributed to improved medical and procedural strategies.
Collapse
|
38
|
Miller LM, Jenny NS, Rawlings AM, Arnold AM, Fitzpatrick AL, Lopez OL, Odden MC. Sex Differences in the Association Between Pentraxin 3 and Cognitive Decline: The Cardiovascular Health Study. J Gerontol A Biol Sci Med Sci 2020; 75:1523-1529. [PMID: 31808814 PMCID: PMC7357589 DOI: 10.1093/gerona/glz217] [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: 03/07/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The importance of systemic inflammation, measured by C-reactive protein, in cognitive decline has been demonstrated; however, the role of vascular inflammation is less understood. Pentraxin 3 (PTX3) is a novel marker of vascular inflammation. METHODS We followed adults 65 and older, free of cardiovascular disease (CVD) for up to 9 years (n = 1,547) in the Cardiovascular Health Study. We evaluated the relationship between PTX3 and change in cognitive function, measured using the Modified Mini-Mental State Examination (3MSE), and incident cognitive impairment (3MSE < 80). Mediation by CVD events, and effect modification by sex and apolipoprotein E ɛ4 allele (APOE4) were also examined. RESULTS The average decline in 3MSE was 0.77 points per year. The association between PTX3 and change in 3MSE differed between women and men (p = .02). In the adjusted model, each standard deviation higher in PTX3 was associated with a 0.20 greater decline in 3MSE score per year in women over follow-up (95% CI: -0. 37, -0.03; p = .02), compared to no change in men (β = 0.07; 95% CI: -0.08, 0.22). CVD events had a minor effect on the associations. No effect modification by APOE4 was found, although we observed the association of PTX3 and cognitive impairment in women was attenuated and nonsignificant after adjustment for APOE4. There was a paradoxical protective association between PTX3 and reduced cognitive impairment in men, even after adjustment for APOE4. CONCLUSIONS We found that vascular inflammation was significantly associated with cognitive decline in older women, but not men.
Collapse
Affiliation(s)
- Lindsay M Miller
- Division of Nephrology-Hypertension, Department of Medicine, University of San Diego, La Jolla
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis
| | - Nancy S Jenny
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington
| | - Andreea M Rawlings
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis
- Kaiser Permanente Center for Health Research, Portland, Oregon
| | - Alice M Arnold
- Department of Biostatistics, University of Washington, Seattle
| | - Annette L Fitzpatrick
- Departments of Family Medicine, Epidemiology, and Global Health, University of Washington, Seattle
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pennsylvania
| | - Michelle C Odden
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis
- Department of Health Research and Policy, Stanford University, Stanford, California
| |
Collapse
|
39
|
Vallabhajosyula S, Ponamgi SP, Shrivastava S, Sundaragiri PR, Miller VM. Reporting of sex as a variable in cardiovascular studies using cultured cells: A systematic review. FASEB J 2020; 34:8778-8786. [PMID: 32946179 PMCID: PMC7383819 DOI: 10.1096/fj.202000122r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
Reporting the sex of biological material is critical for transparency and reproducibility in science. This study examined the reporting of the sex of cells used in cardiovascular studies. Articles from 16 cardiovascular journals that publish peer-reviewed studies in cardiovascular physiology and pharmacology in the year 2018 were systematically reviewed using terms "cultured" and "cells." Data were collected on the sex of cells, the species from which the cells were isolated, and the type of cells, and summarized as a systematic review. Sex was reported in 88 (38.6%) of the 228 studies meeting inclusion criteria. Reporting rates varied with Circulation, Cardiovascular Research and American Journal of Physiology: Heart and Circulatory Physiology having the highest rates of sex reporting (>50%). A majority of the studies used cells from male (54.5%) or both male and female animals (32.9%). Humans (31.8%), rats (20.4%), and mice (43.8%) were the most common sources for cells. Cardiac myocytes were the most commonly used cell type (37.0%). Overall reporting of sex of experimental material remains below 50% and is inconsistent among journals. Sex chromosomes in cells have the potential to affect protein expression and molecular signaling pathways and should be consistently reported.
Collapse
Affiliation(s)
- Saraschandra Vallabhajosyula
- Department of Cardiovascular MedicineMayo ClinicRochesterMNUSA
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineMayo ClinicRochesterMNUSA
- Center for Clinical and Translational ScienceMayo Clinic Graduate School of Biomedical SciencesMayo ClinicRochesterMNUSA
| | - Shiva P. Ponamgi
- Division of Hospital Internal MedicineDepartment of MedicineMayo ClinicRochesterMNUSA
| | | | | | - Virginia M. Miller
- Department of SurgeryMayo ClinicRochesterMNUSA
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| |
Collapse
|
40
|
Abstract
The development of transportation and technology has spread human movements more quickly and widely. As a result, our societies are becoming more complex, composed of people of more diverse races, cultures, religions, and languages. In this study, we focus on the origins of ethnicity while analyzing the background of social members. To track the origin of the ethnicities of which a society is composed, we established a surname-nationality prediction model by learning from a Recurrent Neural Network (RNN) with data recorded by business peoples’ surnames and nationalities to predict nationality with high accuracy through surnames. This study is meaningful because it approaches the social scientific problems of ethnic composition within society through massive data and machine learning: the informatics approach.
Collapse
|
41
|
Chalikiopoulou C, Bizjan BJ, Leventopoulos G, Smaili K, Blagus T, Menti A, Liopetas J, John A, Ali BR, Dolzan V, Hahalis GN, Patrinos GP, Katsila T. Multiomics Analysis Coupled with Text Mining Identify Novel Biomarker Candidates for Recurrent Cardiovascular Events. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:205-215. [PMID: 32176569 DOI: 10.1089/omi.2019.0216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recurrent cardiovascular events remain an enigma that accounts for >30% of deaths worldwide. While heredity and human genetics variation play a key role, host-environment interactions offer a sound conceptual framework to dissect the molecular basis of recurrent cardiovascular events from genes and proteins to metabolites, thus accounting for environmental contributions as well. We report here a multiomics systems science approach so as to map interindividual variability in susceptibility to recurrent cardiovascular events. First, we performed data and text mining through a mixed-methods content analysis to select genomic variants, 10 single nucleotide polymorphisms, and microRNAs (miR-10a, miR-21, and miR-20a), minimizing bias in candidate marker selection. Next, we validated our in silico data in a patient cohort suffering from recurrent cardiovascular events (a cross-sectional study design and sampling). Our findings report a key role in low-density lipoprotein clearance for rs11206510 (p < 0.01) and rs515135 (p < 0.05). miR-10a (p < 0.05) was significantly associated with heart failure, while increased expression levels for miR-21 and miR-20a associated with atherosclerosis. In addition, liquid chromatography-mass spectrometry-based (LC-MS-based) proteomics analyses identified that vascular diameter and cholesterol levels are among the key factors to be considered in recurrent cardiovascular events. From a methodology innovation standpoint, this study offers a strategy to enhance the signal-to-noise ratios in mapping novel biomarker candidates wherein each research and conceptual step were interrogated for their validity and in turn, enriched one another, ideally translating information growth to knowledge growth.
Collapse
Affiliation(s)
| | - Barbara Jenko Bizjan
- Unit of Special Laboratory Diagnostics, University Children's Hospital, UMC, Ljubljana, Slovenia
| | | | - Kalliopi Smaili
- Department of Cardiology, Patras University Hospital Rio, Patras, Greece
| | - Tanja Blagus
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ariadni Menti
- Department of Cardiology, Patras University Hospital Rio, Patras, Greece
| | - John Liopetas
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | - Anne John
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Vita Dolzan
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - George N Hahalis
- Department of Cardiology, Patras University Hospital Rio, Patras, Greece
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece.,Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Zayed Center of Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Theodora Katsila
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece.,Institute of Chemical Biology, National Hellenic Research Centre, Athens, Greece
| |
Collapse
|
42
|
Moyer AM, Matey ET, Miller VM. Individualized medicine: Sex, hormones, genetics, and adverse drug reactions. Pharmacol Res Perspect 2019; 7:e00541. [PMID: 31844524 PMCID: PMC6897337 DOI: 10.1002/prp2.541] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
Clinically relevant adverse drug reactions differ between men and women. The underlying physiological and pharmacological processes contributing to these differences are infrequently studied or reported. As gene expression, cellular regulatory pathways, and integrated physiological functions differ between females and males, aggregating data from combined groups of men and women obscures the ability to detect these differences. This paper summarizes how genetic sex, that is, the presence of sex chromosomes XY for male or XX for female, and the influence of sex hormones affect transporters, receptors, and enzymes involved in drug metabolism. Changing levels of sex steroids throughout life, including increases at puberty, changes with pregnancy, and decreases with age, may directly and indirectly affect drug absorption, distribution, metabolism, and elimination. The direct and indirect effects of sex steroids in the form of exogenous hormones such as those used in hormonal contraceptives, menopausal hormone treatments, transgender therapy, and over-the-counter performance enhancing drugs may interfere with metabolism of other pharmaceuticals, and these interactions may vary by dose, formulation, and mode of delivery (oral, injection, or transdermal) of the steroid hormones. Few drugs have sex-specific labeling or dosing recommendations. Furthermore, there is limited literature evaluating how the circulating levels of sex steroids impact drug efficacy or adverse reactions. Such research is needed in order to improve the understanding of the impact of sex hormones on pharmacological therapies, particularly as medicine moves toward individualizing treatments.
Collapse
Affiliation(s)
- Ann M. Moyer
- Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Eric T. Matey
- Medical Therapy Management and Center for Individualized MedicineMayo ClinicRochesterMNUSA
| | - Virginia M. Miller
- Departments of Surgery, and Physiology and Biomedical EngineeringWomen's Health Research CenterMayo ClinicRochesterMNUSA
| |
Collapse
|
43
|
Abstract
OBJECTIVE Statins are a class of drugs that competitively bind to the active site of HMG-CoA reductase enzyme, thereby inhibiting the initial steps in cholesterol synthesis. Originally approved for use in lowering serum cholesterol, a risk factor for developing atherosclerosis and coronary heart disease, statins have subsequently been noted to have myriad extrahepatic effects, including potential effects on cognition, diabetes, breast cancer, bone, and muscle. This narrative review assesses the current state of the science regarding the risks and benefits of statin therapy in women to identify areas where additional research is needed. METHODS Basic and clinical studies were identified by searching PubMed with particular attention to inclusion of female animals, women, randomized controlled trials, and sex-specific analyses. RESULTS Statin therapy is generally recommended to reduce the risk of cardiovascular disease. None of the current clinical guidelines, however, offer sex-specific recommendations for women due to lack of understanding of sex differences and underlying mechanisms of disease processes. In addition, conclusions regarding efficacy of treatments do not consider lipid solubility for the drug, dosing, duration of treatment, interactions with estrogen, or comorbidities. Pleiotropic effects of statins are often derived from secondary analysis of studies with cardiovascular events as primary outcomes. CONCLUSIONS Many of the trials that have established the efficacy and safety of statins were conducted predominantly or entirely in men, with results extrapolated to women. Additional research is needed to guide clinical recommendations specific to women. : Video Summary:http://links.lww.com/MENO/A462.
Collapse
Affiliation(s)
- Stephanie S. Faubion
- Center for Women’s Health, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Ekta Kapoor
- Center for Women’s Health, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN
| | - Ann M. Moyer
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN
| | - Howard N. Hodis
- Atherosclerosis Research Unit, Departments of Medicine and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Virginia M. Miller
- Departments of Surgery and Physiology & Biomedical Engineering, Women’s Health Research Center, Mayo Clinic, Rochester, MN
| |
Collapse
|
44
|
Yogasundaram H, Qi A, Nguyen Q, Oudit GY. Battle of the Sexes: Differential Prognosis by Sex in Dilated Cardiomyopathy. Can J Cardiol 2019; 36:7-10. [PMID: 31537392 DOI: 10.1016/j.cjca.2019.06.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/28/2019] [Accepted: 07/07/2019] [Indexed: 01/15/2023] Open
Affiliation(s)
- Haran Yogasundaram
- Division of Cardiology, Department of Medicine, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Arthur Qi
- Division of Cardiology, Department of Medicine, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Quynh Nguyen
- Division of Cardiology, Department of Medicine, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
45
|
Copy number variants in lipid metabolism genes are associated with gallstones disease in men. Eur J Hum Genet 2019; 28:264-273. [PMID: 31485028 PMCID: PMC6974590 DOI: 10.1038/s41431-019-0501-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/02/2019] [Indexed: 11/21/2022] Open
Abstract
Gallstones Disease (GSD) is one of the most common digestive diseases requiring hospitalization and surgical procedures in the world. GSD has a high prevalence in populations with European or Amerindian ancestry (10–20%) and the influence of genetic factors is broadly acknowledged. However, known genetic variants do not entirely explain the disease heritability suggesting that additional genetic variants remain to be identified. Here, we examined the association of copy number variants (CNVs) with GSD in a sample of 4778 individuals (1929 GSD cases and 2849 controls) including two European cohorts from Germany (n = 3702) and one admixed Latin American cohort from Chile (n = 1076). We detected 2936 large and rare CNVs events (size > 100 kb, frequency < 1%). Case-control burden analysis and generalized linear regression models revealed significant association of CNVs with GSD in men, with the strongest effect observed with CNVs overlapping lipid metabolism genes (p-value = 6.54 × 10–4; OR = 2.76; CI 95% = 1.53–4.89). Our results indicate a clear link between CNVs and GSD in men and provides additional evidence that the genetic components of risk for GSD are complex, can be sex specific and include CNVs affecting genes involved in lipid metabolism.
Collapse
|
46
|
Miller VM, Naftolin F, Asthana S, Black DM, Brinton EA, Budoff MJ, Cedars MI, Dowling NM, Gleason CE, Hodis HN, Jayachandran M, Kantarci K, Lobo RA, Manson JE, Pal L, Santoro NF, Taylor HS, Harman SM. The Kronos Early Estrogen Prevention Study (KEEPS): what have we learned? Menopause 2019; 26:1071-1084. [PMID: 31453973 PMCID: PMC6738629 DOI: 10.1097/gme.0000000000001326] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/20/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The Kronos Early Estrogen Prevention Study (KEEPS) was designed to address gaps in understanding the effects of timely menopausal hormone treatments (HT) on cardiovascular health and other effects of menopause after the premature termination of the Women's Health Initiative. METHOD The KEEPS was a randomized, double-blinded, placebo-controlled trial to test the hypothesis that initiation of HT (oral conjugated equine estrogens [o-CEE] or transdermal 17β-estradiol [t-E2]) in healthy, recently postmenopausal women (n = 727) would slow the progression of atherosclerosis as measured by changes in carotid artery intima-media thickness (CIMT). RESULTS After 4 years, neither HT affected the rate of increase in CIMT. There was a trend for reduced accumulation of coronary artery calcium with o-CEE. There were no severe adverse effects, including venous thrombosis. Several ancillary studies demonstrated a positive effect on mood with o-CEE, and reduced hot flashes, improved sleep, and maintenance of bone mineral density with both treatments. Sexual function improved with t-E2. There were no significant effects of either treatment on cognition, breast pain, or skin wrinkling. Variants of genes associated with estrogen metabolism influenced the age of menopause and variability in effects of the HT on CIMT. Platelet activation associated with the development of white matter hyperintensities in the brain. CONCLUSIONS KEEPS and its ancillary studies have supported the value and safety of the use of HT in recently postmenopausal women and provide a perspective for future research to optimize HT and health of postmenopausal women. The KEEPS continuation study continues to pursue these issues.
Collapse
Affiliation(s)
- Virginia M. Miller
- Departments of Surgery and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Fredrick Naftolin
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY
| | - Sanjay Asthana
- Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, University of Wisconsin School of Medicine and Public Health and the Geriatric Research, Madison, WI
| | - Dennis M. Black
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, CA
| | | | - Matthew J. Budoff
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California Los Angeles, Torrance, CA
| | - Marcelle I. Cedars
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA
| | - N. Maritza Dowling
- Departments of Acute and Chronic Care, Epidemiology and Biostatistics, George Washington University School of Nursing and Milken Institute School of Public Health, Washington, DC
| | - Carey E. Gleason
- Division of Geriatrics, Department of Medicine, University of Wisconsin School of Medicine and Public Health and the William S. Middleton Memorial VA, Geriatric Research, Education and Clinical Center, Madison, WI
| | - Howard N. Hodis
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, CA
| | - Muthuvel Jayachandran
- Department of Physiology and Biomedical Engineering, Division of Nephrology and Hypertension, Division of Hematology Research, Mayo Clinic, Rochester, MN
| | | | - Rogerio A. Lobo
- Department of Obstetrics and Gynecology, Columbia University, New York, NY
| | - JoAnn E. Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lubna Pal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Nanette F. Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | | |
Collapse
|
47
|
Tao K, Hu Z, Zhang Y, Jiang D, Cheng H. LncRNA CASC11 improves atherosclerosis by downregulating IL-9 and regulating vascular smooth muscle cell apoptosis and proliferation. Biosci Biotechnol Biochem 2019; 83:1284-1288. [PMID: 30915898 DOI: 10.1080/09168451.2019.1597621] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
ABSTRACT
LncRNA CASC11 is an oncogene in several types of cancer, while its role in atherosclerosis is unknown. In the present study we found that CASC11 was downregulated, while IL-9 was upregulated in plasma of atherosclerosis patients compared with healthy controls. Altered plasma levels of CASC11 and IL-9 distinguished atherosclerosis patients from healthy controls. CASC11 and IL-9 were significantly and inversely correlated in atherosclerosis patients but not in healthy controls. Exogenous IL-9 treatment failed to significantly affect expression levels of CASC11 in vascular smooth muscle cells (VSMC), while CASC11 overexpression resulted in the downregulation of IL-9. CASC11 overexpression also resulted in the downregulation of proliferation and promoted apoptosis of VSMC. Therefore, CASC11 may improve atherosclerosis by downregulating IL-9 and regulating VSMC apoptosis and proliferation.
Collapse
Affiliation(s)
- Kaiyu Tao
- Department of cardiovascular surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Zhanglong Hu
- Department of cardiovascular surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Yigong Zhang
- Department of cardiovascular surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Daming Jiang
- Department of cardiovascular surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Haifeng Cheng
- Department of cardiovascular surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| |
Collapse
|
48
|
XX sex chromosome complement promotes atherosclerosis in mice. Nat Commun 2019; 10:2631. [PMID: 31201301 PMCID: PMC6643208 DOI: 10.1038/s41467-019-10462-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/08/2019] [Indexed: 12/27/2022] Open
Abstract
Men and women differ in circulating lipids and coronary artery disease (CAD). While sex hormones such as estrogens decrease CAD risk, hormone replacement therapy increases risk. Biological sex is determined by sex hormones and chromosomes, but effects of sex chromosomes on circulating lipids and atherosclerosis are unknown. Here, we use mouse models to separate effects of sex chromosomes and hormones on atherosclerosis, circulating lipids and intestinal fat metabolism. We assess atherosclerosis in multiple models and experimental paradigms that distinguish effects of sex chromosomes, and male or female gonads. Pro-atherogenic lipids and atherosclerosis are greater in XX than XY mice, indicating a primary effect of sex chromosomes. Small intestine expression of enzymes involved in lipid absorption and chylomicron assembly are greater in XX male and female mice with higher intestinal lipids. Together, our results show that an XX sex chromosome complement promotes the bioavailability of dietary fat to accelerate atherosclerosis. Men and women differ in their risk of developing coronary artery disease, in part due to differences in their levels of sex hormones. Here, AlSiraj et al. show that the XX sex genotype regulates lipid metabolism and promotes atherosclerosis independently of sex hormones in mice.
Collapse
|
49
|
Associations between coffee consumption and all-cause and cause-specific mortality in a Japanese city: the Takayama study. Public Health Nutr 2019; 22:2561-2568. [PMID: 31107195 DOI: 10.1017/s1368980019000764] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Epidemiological studies suggest that coffee consumption is inversely associated with all-cause and cause-specific mortality. Evidence from studies targeting non-white, non-Western populations is still sparse, although coffee is popular and widely consumed in Asian countries. DESIGN Population-based, prospective cohort study. We used Cox proportional hazards models with adjustment for dietary and lifestyle factors to estimate associations between coffee consumption and all-cause and cause-specific mortality. Dietary intake including coffee consumption was assessed only at baseline using a validated FFQ. SETTING A Japanese city. PARTICIPANTS Individuals aged 35 years or older without cancer, CHD and stroke at baseline (n 29 079) and followed from 1992 to 2008. RESULTS From 410 352 person-years, 5339 deaths were identified (mean follow-up = 14·1 years). Coffee consumption was inversely associated with mortality from all causes and CVD among all participants, but not from cancer. Compared with the category of 'none', the multivariate hazard ratio (95 % CI) for all-cause mortality was 0·93 (0·86, 1·00) for <1 cup/d, 0·84 (0·76, 0·93) for 1 cup/d and 0·81 (0·71, 0·92) for 2-3 cups/d. The multivariate hazard ratio (95 % CI) for cardiovascular mortality were 0·87 (0·77, 0·99) for <1 cup/d, 0·76 (0·63, 0·92) for 1 cup/d and 0·67 (0·50, 0·89) for 2-3 cups/d. Inverse associations were also observed for mortality from other causes, specifically infectious and digestive diseases. CONCLUSION Drinking coffee, even 1 cup/d, was inversely associated with all-cause mortality and mortality from cardiovascular, infectious and digestive diseases.
Collapse
|
50
|
Nafakhi H, Al-Mosawi AA, Mudhafar MM, Al-Nafakh HA, Alsaady R. Digit ratio and hair color relationships with coronary atherosclerotic markers. Asian Cardiovasc Thorac Ann 2019; 27:362-368. [PMID: 31018656 DOI: 10.1177/0218492319847379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective We aimed to investigate the association between body phenotype markers and coronary atherosclerosis markers. Methods Eighty-nine patients (mean age 57 ± 9 years, 50.5% male) who were assessed for coronary artery disease by 64-slice multidetector computed tomography angiography were enrolled in the study. Results A significant positive association was observed between coronary artery calcification and the second to fourth digit ratio ( r = 0.2, p = 0.00); more notably in males compared to females ( p = 0.03 vs. p = 0.08). There was a significant association between coronary artery calcification and pure white hair ( p = 0.00). There was no significant association between pericardial fat volume and second to fourth digit ratio ( p = 0.8). Pericardial fat volume was significantly associated with pure white hair ( p = 0.02). A significant association was observed between coronary plaque and a higher second to fourth digit ratio ( p = 0.01) and this was more notable in males ( p = 0.04). A significant association was observed between pure white hair and coronary plaque ( p = 0.00). After adjustment for conventional cardiac risk factors, the association of coronary artery calcification and coronary plaque with the second to fourth digit ratio did not persist, whereas the association of pericardial fat volume and coronary artery calcification with pure white hair remained significant ( p = 0.01 and p = 0.00, respectively). Conclusion These results suggest a possible predictive value of hair color, rather than digit ratio, in assessing increased risk of coronary atherosclerosis and cardiac fat deposition.
Collapse
Affiliation(s)
- Hussein Nafakhi
- 1 Internal Medicine Department, Medicine College, University of Kufa, Najaf, Iraq
| | | | - Mundher M Mudhafar
- 3 Radiology Department, Jabir Ibn Hayyan Medical University, Najaf, Iraq
| | - Hasan A Al-Nafakh
- 2 Radiology Department, Medicine College, University of Kufa, Najaf, Iraq
| | - Rana Alsaady
- 2 Radiology Department, Medicine College, University of Kufa, Najaf, Iraq
| |
Collapse
|