Skip to main content
Reviews in Cardiovascular Medicine logoLink to Reviews in Cardiovascular Medicine
. 2025 Feb 20;26(2):26430. doi: 10.31083/RCM26430

Getting to the Heart of the Matter: Exploring the Intersection of Cardiovascular Disease, Sex and Race and How Exercise, and Gut Microbiota Influence these Relationships

John J Guers 1, Kevin S Heffernan 2, Sara C Campbell 3,4,*
Editor: Carl J Lavie
PMCID: PMC11868917  PMID: 40026503

Abstract

Cardiovascular disease (CVD) is the leading cause of death worldwide, with physical inactivity being a known contributor to the global rates of CVD incidence. CVD incidence, however, is not uniform with recognized sex differences as well and racial and ethnic differences. Furthermore, gut microbiota have been associated with CVD, sex, and race/ethnicity. Researchers have begun to examine the interplay of these complicated yet interrelated topics. This review will present evidence that CVD (risk and development), and gut microbiota are distinct between the sexes and racial/ethnic groups, which appear to be influenced by acculturation, discrimination, stress, and lifestyle factors like exercise. Furthermore, this review will address the beneficial impacts of exercise on the cardiovascular system and will provide recommendations for future research in the field.

Keywords: cardiovascular disease, ethnicity, microbiome, heart, intestine, physical activity

1. Introduction and a Brief Overview of Topics

Cardiovascular disease (CVD) remains the leading cause of mortality and morbidity worldwide [1]. CVDs include atherosclerosis, myocardial infarction, stroke, heart failure, and hypertension, among others. Risk factors for CVD can be categorized as either modifiable (habitual alcohol and tobacco use, high blood lipids, high blood pressure, excess adiposity/body fat, poor glucose control/diabetes, physical inactivity, and high-fat “Western” diet) or nonmodifiable (age, biological sex, genetics). Physical inactivity is a known contributor to the global rates of CVD [2]. The United States “Physical Activity Guidelines for Americans” recommend that adults engage in 150–300 minutes of moderate or 75 minutes of vigorous physical activity each week [2].

This narrative review will build on our previous work by presenting novel data that shows a clear relationship between CVD, exercise, sex, race and gut microbiota. Specifically, we highlight how biological sex and race impact gut microbiota and how exercise can be used to improve gut health while minimizing disparities. These factors are all linked in a complicated system that ultimately can strongly influence cardiovascular health. This review will provide a brief outline of each topic, take a deep dive into the impacts of exercise on CVD with considerations for sex, race and gut microbiota, truly getting to the heart of the matter.

2. Topical Overviews

2.1 Exercise Promotes Longevity and Health

It is well known that exercise preserves health. Studies conducted as early as the 1910’s highlight the protective effects of manual labor on degenerative diseases [3]. Similar reports reinforced the notion that physical activity can help prevent disease [4]. More recently, studies have shown that aerobic capacity correlates with an increased lifespan and increased “healthspan” [5]. Exercise is known to decrease all-cause mortality, and we know that cardiorespiratory fitness correlates with longevity [6]. Over the past several decades researchers have become interested in which potential mechanisms are responsible for these protective effects. For the purposes of this paper, we will focus on the mechanisms involved with exercise-induced protection of the cardiovascular system.

2.2 Gut Microbiota and Exercise

The gut microbiota consists of trillions of microbial cells such as bacteria, fungi, viruses, and archaea [7]. Regarding gut bacteria, there are over 1100 genera, and approximately 90% fall under the phylum Bacteroidota and Bacillota (formerly known as Bacteroidetes and Firmicutes [8], respectively) while, the minority of gut bacteria are Pseudomonadota, Actinomycetota, Fusobacteriota, and Verrucomicrobiota (formerly known as Proteobacteria, Actinobacteria, Fusobacteria, Verrucomicrobia [8], respectively) phyla [9]. Commonly observed in a healthy gut microbiota is a decreased Bacillota to Bacteroidota ratio, stable community, and greater species diversity [10].

The gut microbiota is now recognized as being critical for the maintenance of optimal human health. When the gut microbiota is in symbiosis with the host, microbes can promote health. However, when in dysbiosis (unbalanced gut microbes) with the host, the bacteria can contribute to chronic disease. In a healthy host, the gut microbiota favorably affects digestion, nutrient absorption, and production of folate, vitamins, and short chain fatty acids (SCFAs).

Our lab [10], and others [11, 12, 13] have examined the link between the gut microbiota and exercise in animal models. The gut microbiota of sedentary individuals differs from active individuals [14, 15, 16, 17]. Results from humans and animal studies clearly show that exercise is central to healthful aging, improves the diversity of microbes within the Bacillota phylum [10, 13, 14], and increases the abundance of beneficial bacteria such as Roseburia intestinalis, Faecalibacterium prausnitzii, and Akkermansia muciniphila [15, 18].

In addition, the gut microbiota appears to adapt to the unique demands of exercise [19, 20, 21]. Changes in the gut microbiota that occur with exercise generate metabolites that further provide the host with performance advantages [19, 20, 21, 22, 23, 24]. Athletes typically have improved carbohydrate metabolism, higher tolerance to oxidative stress, greater insulin sensitivity, enhanced muscle tissue repair, and greater energy harvesting [14, 25, 26, 27].

Moreover, results from antibiotic and germ-free mouse models demonstrate a bidirectional relationship between gut microbiota and exercise. Results show that gut microbiota must be intact for exercise performance and various aspects of maintenance of exercise training but perhaps not for adapting to exercise training [12, 19, 20, 21, 22, 23, 24, 28].

In summary, habitually exercise-trained individuals have a beneficial gut microbiota. Additionally, sedentary individuals who undertake exercise training can improve the abundance of beneficial gut microbes. Importantly, exercise-induced microbial changes in human studies are observed across the lifespan and are seen in both men and women. It is important to underscore that the favorable gut modifications that come with habitual exercise training are lost with cessation of exercise (“use it or lose it”). In conclusion, an intact gut microbiota must be present to fully adapt to exercise-induced training adaptations, including muscle hypertrophy.

2.3 Sex and Exercise Differences

There are established sex differences in heart size, stroke volume, and hemoglobin content contributing to exercise performance [29, 30, 31]. Among humans, sex differences in heart size do not manifest until puberty. By adulthood, hearts are approximately 30% larger in males compared to females, primarily due to greater myocyte hypertrophy among males [32]. These observed sex-based differences in heart size are the primary factors contributing to larger stroke volume among males compared to females [33, 34, 35]. However, there does not appear to be a difference in maximum heart rate by sex [33]. Hemoglobin concentration in blood is higher for males compared to females, contributing to sex differences in oxygen carrying capacity [36]. Although males have larger muscle fibers and more capillaries per fiber, capillary density does not differ between sexes [37]. Furthermore, while skeletal muscles of men are usually stronger and more powerful than women, men are often more fatigable than women for sustained or intermittent isometric contractions performed at a similar relative intensity [38]. Importantly, these fundamental differences between biologic males and females emerge at the onset of puberty, suggesting that sex hormones may be responsible for conferring sex-based differences. This is relevant because exercise motivation, particularly in females, has been shown to be regulated by estrogen. Krause et al. [39] demonstrated that in estrogen deficiency there was reduced melanocortin-4 signaling which lowered the drive to exercise, illuminating the power of estrogen during the reproductive cycle in motivating behavior and maintaining an active lifestyle in women. Intriguingly, estrogen deficiency (menopause) is also when CVD risk increases [40], meaning not only are women at high risk of CVD, but they may be less likely to want to engage in exercise which would help in the prevention of CVD and other metabolic risk factors.

2.4 Sex Differences in Gut Microbiota Considering Lifespan

Studies comparing compositional differences in the microbiota between males and females often find differences between each sex, but not always [41]. This may indicate that the sex differences are context-dependent. For example, in several studies, compositional differences were described as females having higher levels of Clostridium from the Bacillota (formerly Firmicutes) phylum and males having higher levels of Prevotella from the Bacteroidota (formerly Bacteroidetes) phylum and Lactobacillus from the Bacillota phylum [42, 43, 44]. Other observations include males having less microbial diversity compared to females [42]. These compositional differences are not always consistent between the sexes, particularly when a study alters an additional factor like diet [42].

2.4.1 Birth and Childhood

A variety of factors impact microbiota in the early years of life including mode of birth, breastfeeding or formula feeding, antibiotic treatment, genetics, sex, and more [41]. Consequently, these microbes likely affect human development in a sex-dependent manner. Even from birth, some studies show different microbial communities between males and females [42]. For example, females delivered by asthmatic mothers are prone to Bacteroidaceae microbes compared to males that tend to harbor Lactobacilli [45]. Another example of early sex differences observing 300 infants is the temperament of males appears to be more positive when Bifidobacterium of the Actinomycetota (formerly Actinobacteria) phyla and Clostridiaceae of the Bacillota phyla are present [46]. Female members that have gut communities with Veillonella tend to be more risk averse [46]. Using reverse-transcriptase qPCR a study showed that boys had higher abundance of several Bifidobacterium spp. over three years [47]. A study examined how normal weight pre-puberty girls have increased Bacteroidota compared to obese girls [48]. Interestingly, these differences were not seen in boys of the same age [48]. Obesity in girls of this group had more developed adrenal glands and an underexpression of gonadal estradiol, the predominant estrogen [49]. Boys in this group had increased dehydroepiandrosterone (DHEA) [49]. Given that other studies have linked estrogen levels with certain groups of microbes, it would suggest that these girls could have gut microbes that play a role in estrogen-driven diseases.

2.4.2 Puberty

During puberty, the difference in levels of sex hormones between males and females increases, and the effects they have on the microbiome appear to be more prominent as well [50]. For example, in a human twin study of teenagers, there was greater dissimilarity of the gut microbiota between opposite-sex twins than same-sex twins during puberty [51]. In another study using mice, the alpha-diversity of females changed significantly compared to males after puberty and the sex-related compositional differences disappeared after these male mice were castrated [52]. Interestingly, in a study by Yuan et al. (2020) [53] there was no difference in alpha-and beta-diversity of girls and boys before puberty, but there was an association of certain microbes to testosterone including Adlercreutzia, Ruminococcus, Dorea, Clostridium, and Parabacteroides. Similarly, male mice undergoing a gonadectomy were administered testosterone and subsequently, did not exhibit the microbiota changes [52]. Another group of mice that had a gonadectomy that did not receive testosterone supplementation did exhibit microbial changes [52]. This highlights testosterone as a key factor in microbial change. Similar studies performing ovariectomies on mice showed changes in microbiota including a reduction of Pseudomonadota (formerly Proteobacteria), higher Akkermansia, and a decreased ratio of Bacillota to Bacteroidota [54].

2.4.3 Adulthood

During adulthood, estrogen and testosterone are described as potent modifiers of the human body and the microbiota [55]. And due to the different concentrations of sex hormones in males and females, the microbiota and its effects are modulated in a sex-dependent manner [55]. The adult microbiota is also characterized as being more stable compared to other stages of life [42]. In a human study of 516 Japanese males and females, Prevotellaceae was more abundant in males and Ruminococcaceae was more abundant in females [44]. The microbiota from 91 pregnant women were transplanted via fecal microbiota transfer (FMT) into germ-free (GF) mice in the 1st and 3rd trimester [56]. Mice receiving FMT from third trimester (T3) showed pregnancy-like effects like increased adiposity and insulin sensitivity, but FMT from first trimester (T1) did not show these effects [56]. Additionally, there was no correlation between the microbiota compared to estrogen levels throughout the menstrual cycle of 17 females [57]. Importantly, adulthood is when many diseases can progress, and this can have sex-dependent effects on the microbiota as well. In a study by Mahnic et al. (2018) [58], they also found higher levels of Bacteroides and Prevotella in males compared to females. To understand these relationships fully, the mechanisms that influence them should be investigated.

2.4.4 Old Age

As people age, the microbial changes between males and females become less prominent [42]. It is important to note that this is also when male and female hormone levels become more similar [41]. These events are likely not a coincidence. In a study by Santos-Marcos et al. (2018) [59], the microbiota of human males and post-menopausal females were compared to measure any differences between each sex. The Bacillota/Bacteroidota ratio was different between males and females as well as the amount of saccharolytic activity [59]. More specifically, pre-menopausal women versus post-menopausal women and pre-menopausal females versus males were most different [59]. Given that estrogen levels are greatly reduced in post-menopausal women, the data suggests that the changes in the microbiota are influenced by the changes in sex hormones [59]. Interestingly, Deltaproteobacteria in the cecum increased in abundance as mice aged [60]. This raises the question of how age may impact the microbiota differently depending on where along the gastrointestinal tract the sample is taken.

2.5 Race, Sex, and Exercise Differences

According to the 2022 Centers for Disease Control, National Center of Health Statistics Data Brief on physical activity in the United States (US) the percentage of adults who met the guidelines for both aerobic and muscle-strengthening activities varied by race and Hispanic origin [61]. In general, in 2020, 24.2% of adults aged 18 and over met the 2018 Physical Activity Guidelines for Americans for both aerobic and muscle-strengthening activities [61]. When accounting for race/ethnicity Hispanic men (23.5%) were less likely to meet both physical activity guidelines than non-Hispanic White (30.5%), non-Hispanic Asian (30.2%), and non-Hispanic Black (29.7%) men [61]. Non-Hispanic White women (24.3%) were more likely to meet both guidelines than Hispanic (18.0%), non-Hispanic Asian (16.7%), and non-Hispanic Black (16.5%) women [61]. Across all race and Hispanic-origin groups, men were more likely than women to meet the guidelines for both aerobic and muscle-strengthening activities [61]. The percentage of men who met both physical activity guidelines increased as family income increased, from 16.2% of men with a family income of less than 100% of the federal poverty level (FPL), to 20.0% of men with income at 100%–199% of FPL, and 32.4% of those with income at 200% of FPL or more [61]. The percentage of women who met both physical activity guidelines increased as family income increased, from 9.9% of women with family income less than 100% of FPL, to 13.6% of women with income at 100%–199% of FPL, and 25.9% of those with income at 200% of FPL or more [61]. Across all income groups, men were more likely than women to meet the guidelines for both types of activity [61].

2.6 Racial Disparities and Gut Microbiota

Currently, human gut microbiota studies have had a narrow focus or simply describe broad population-level changes to gut communities in response to environmental variation. As such, only a few studies have been designed to address gut microbiota variation in relation to structural inequities, and even fewer have attempted to link host health to socially attributed variations in the gut microbiota [62, 63, 64, 65, 66]. Nevertheless, the small but existing literature does provide accumulating evidence that the social and environmental factors that contribute to health inequities may also predict gut microbiota characteristics. For example, measures of socioeconomic status (SES) across globally diverse populations, have been associated with a distinct gut microbiota in both adults [66, 67, 68] and children [69, 70, 71, 72, 73]. Similarly, the gut microbiota consistently varies with race (e.g., Asian, Black, Hispanic, White) and/or ethnicity/ancestry (Arapaho, Cheyenne, Dutch, Ghanaian, Moroccan) in adults [62, 63, 65, 74] and children [70, 71, 75, 76].

There is strong evidence linking structural inequities to gut microbiota variation in the context of SES. For example, neighborhood SES has been shown to explain 12–25% of the variation in adult gut microbiota composition, after adjustment for demographic and lifestyle factors, and was positively correlated with gut microbiota diversity [67]. Similar results noting an association between neighborhood SES and gut microbiota diversity were also obtained utilizing a discordant-twin analysis, which minimizes the possibility of confounding by shared genetic or family influences [68]. Finally, it has been shown that the relative abundance of taxa, accounting for 38.8% of the gut microbiota, varies in relation to indices of wealth appraised as personal yearly income and spending [66].

Despite the important contributions of these findings, most gut microbiota studies in minoritized populations do not operationally define structural inequities. Furthermore, race and ethnicity/ancestry are often incorrectly conflated. Whether the gut microbiota is impacted more by the personal lived experiences of perceived racism and discrimination (internalization) versus overt structural/systemic oppressive policies remains largely unknown. It is likely a combination of both. Similarly, the scale (i.e., household, neighborhood, and beyond) at which structural inequities might affect the gut microbiota is unclear. Nonetheless, the existing literature demonstrates that the same social inequities that predict disease disparities also predict variation in the gut microbiota. These relationships underscore the likely role of the gut microbiota in mediating socially driven health disparities.

3. Why is Exercise so Critical?

Exercise has many health benefits. These benefits apply to people of all ages, races and ethnicities, and sexes. Exercise helps individuals maintain a healthy weight, reduces the risk of depression and a decline in cognitive function and lowers a person’s risk for many diseases, such as CVD and other chronic health diseases [3, 4, 5, 6]. When done regularly, moderate- and vigorous-intensity physical activity strengthens the cardiac myocardium and improves the heart’s ability to distribute blood to the body, thereby reducing CVD risk. Exercise can reduce this risk through a variety of mechanisms including lowering blood pressure, and triglycerides, raising HDL (high-density lipoproteins), decreasing arterial stiffness, reducing the risk of being overweight or obese and maintaining a healthy weight, maintenaining in-range blood glucose and insulin levels, and reducing inflammation [3, 4, 5, 6]. This section of the review will highlight the impacts of exercise on the cardiovascular system and the mechanisms by which this occurs, providing a foundation for which we will later discuss the integrated roles of sex, race/ethnicity, CVD, and gut microbiota.

3.1 Impacts of Exercise on the Cardiovascular System

Broadly, exercise decreases CVD [77] and increased aerobic fitness has been shown to reduce mortality rates of individuals following myocardial infarction [78]. These improvements have been shown in various animal models [79, 80, 81] and human studies [82, 83, 84]. Specifically, it is believed that chronic shear stresses on the endothelial lining of the blood vessels and the endocardium, which are derived from exercise-induced increases in blood flow, increase nitric oxide (NO) bioavailability [85] (Fig. 1). NO is a vasoprotective molecule that prevents vascular dysfunction, platelet aggregation, leukocyte adhesion and vascular stiffening [86, 87]. Reductions in NO have been indicated in the development of hypertension and CVD [88, 89].

Fig. 1.

Fig. 1.

A representation of nitric oxide signaling. Shear stress increases intracellular calcium (Ca2+) which enhances endothelial nitric oxide synthase (eNOS) enzymatic action. eNOS catalyzes the synthesis of L-arginine to nitric oxide. Tetrahydrobiopterin (BH4) is a critical cofactor.

Furthermore, exercise augments anti-oxidant defense and decreases reactive oxygen species (ROS) production [90, 91, 92]. Production of ROS is known to increase the potential for cellular damage [93, 94] and can augment the severity of myocardial ischemia [95]. Previous work has shown that exercise-trained rodents have increased cardiac output compared with sedentary littermates following in-vivo myocardial ischemia [96]. Exercise has been long known to increase cardiac output via myocardial hypertrophy and proliferation [97]. More recently exercise has been shown to increase peroxisome proliferator-activated receptor-gamma coactivator-1α [98, 99], which has the potential to increase longevity and promote health [100]. Lastly, exercise has several indirect effects that improve cardiovascular health including weight reduction [101] and improved gut health [10]. In the following sections, we will review each of these mechanisms in detail.

3.2 Exercise Mediated NO Production

Endothelium-derived NO is essential for cardiovascular health [86, 87] and its production is augmented with acute [102] and chronic exercise [103]. Endothelial-derived NO is synthesized from L-arginine by endothelial nitric oxide synthase (eNOS) and released by endothelial cells [104, 105]. Shear stresses placed on the endothelial cells of blood vessels cause the release of NO, which triggers vasodilation [104, 105]. The repeated shear stresses which are associated with repeated bouts of exercise are thought to increase NO bioavailability by chronically stimulating its release [85].

Improvements in rodent vascular NO bioavailability are often indicated in-vivo by examining endothelial-dependent dilation (EDD) in the blood vessel of interest [90]. Because NO is a key regulator of vasodilation, reductions in EDD can be indicative of diminished NO bioavailability. Rodent exercise perturbations ranging from 2–13 weeks have been shown to improve EDD [90, 103, 106] and thus NO bioavailability. This was confirmed in an acute study consisting of two to four weeks of treadmill training in healthy rats. Dose-dependent EDD was improved in the skeletal muscle arterioles of the exercise-trained rats [107], while endothelial independent dilation was not changed. In a 13-week exercise intervention, EDD and NO production in the femoral artery were increased in Wistar-Kyoto rats following treadmill training [108]. Both eNOS expression and phosphorylated eNOS (Ser1177) expression were increased in trained rats when compared to their sedentary littermates.

Exercise also has a vascular protective effect in several models of rodent vascular dysfunction. In a study by Guers et al. [109], 6 weeks of voluntary wheel running protected against salt-induced (4% NaCl chow) losses in EDD in rat femoral arteries. Western blot analysis demonstrated that this may have been mediated through a decrease in protein concentration of the reactive oxygen species: nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) and Gp91-phox, two subunits of NADPH oxidase. Protein concentrations of both NOX4 and Gp91-phox were initially increased following 6-weeks of a high salt diet in rodents. Exercise also led to an upregulation of the antioxidant superoxide dismutase-2 (SOD2). Collectively, there was a reduction in overall oxidative stress and thus an increase in vascular eNOS bioavailability. eNOS tends to become uncoupled with high levels of oxidative stress [110] and thus becomes unable to synthesize NO [111].

Exercise not only augments NO production in blood vessels but also in the heart [112]. In a study by Kuczmarski et al. [113], 4 weeks of voluntary wheel running helped maintain left ventricular (LV) cardiac function following an ischemia-perfusion injury in rats in a model of chronic kidney disease. Kuczmarski found that wheel running protected against losses in LV NO levels and improved overall cardiac redox status [113]. Specifically, this appeared to be mediated through an upregulation of the antioxidant SOD2 [113]. Furthermore, similar to blood vessels, eNOS is upregulated in the heart with chronic aerobic exercise [112]. Dogs who were treadmill trained for 10 days experienced increases in dose dependent EDD in both coronary arteries and the microvasculature of the heart [114]. The authors also found an increase in the constitutive nitric oxide (ECNOS) gene. Together these data further support the notion of an increase in NO bioavailability in the heart as a result of exercise.

Exercise also has the potential to increase NO bioavailability in humans [115, 116]. Performing moderate aerobic exercise for 1 hour a day for a month increased NO generation and reduced resting blood pressure. This effect was thought to be mediated through an increase in antioxidant enzymes in blood monocytes [115]. In another study by Tanaka et al. [117], the authors discovered that individuals who have high levels of aerobic fitness do not experience the typical age-related decreases in vascular function as measured by EDD. Furthermore, 12 weeks of brisk walking restored losses in EDD in previously sedentary middle-aged and old individuals [117]. Lastly, four weeks of home-based exercise restored losses in forearm EDD in individuals with hypercholesterolemia independent of dietary modifications [118].

3.3 Exercise and Heart Failure

Collectively, patients with heart failure tend to have a significant reduction in aerobic capacity [119]. This appears to be at least partially mediated through a reduction in NO [120]. Heart failure patients also consistently have a reduction in EDD [121] which can be partially restored with supplementation of L-Arginine, a precursor of NO [122]. A hallmark of heart failure tends to be the reduction in blood flow back towards the heart which diminishes pre-load. Exercise training has been shown to improve outcomes in patients with heart failure by increasing NO bioavailability and in turn blood flow and preload. Further to this, 12 weeks of aerobic exercise training increases forearm EDD in hypertensive individuals [123].

In both the heart and blood vessels, as indicated in the aforementioned studies, oxidative stress appears to be one of the principal mediators in reducing NO levels consequently disrupting cardiovascular homeostasis. Oxidative stress is defined as an imbalance of free radical production and the production of free radical scavenging antioxidants [124]. Oxidative stress has been indicated in a number of pathologies including CVD [95, 123, 125]. As an example of this: NADPH oxidases were found to be significantly upregulated in aortic atherosclerotic lesions taken from human autopsies [126]. Furthermore, SOD2 knockout mice experienced increased mitochondrial oxidative stress which led to the onset of hypertension [127] and elevations in oxidative stress levels were associated with the severity of heart failure in both the left and right ventricles of mice following myocardial ischemia [128]. Lastly, a clinical studyhas found correlations between markers of oxidative stress and instances of heart failure [129]. Interestingly, in many cases exogenous antioxidants have been shown to improve outcomes in certain instances of CVD [130, 131].

3.4 Upregulation of Endogenous Antioxidant Defense

As mentioned earlier exercise has the capacity to increase antioxidant defenses and decrease oxidative stress levels which protects against a reduction in NO bioavailability and maintains normal cardiovascular function. SOD is an antioxidant that can be upregulated through exercise [109, 113]. SOD is critical in the maintenance of cardiovascular homeostasis as it prevents the breakdown of NO by the reactive oxygen species superoxide (O2.-) [132]. O2.- has a high affinity for NO and rapidly converts it to peroxynitrite (ONOO-) which can damage lipoproteins. SOD reacts and dismutates O2.- to H2O2 before this reaction can occur. An increase in O2.- disrupts vascular function [133] and elevations in ONOO- levels are associated with CVD [134] (Fig. 2).

Fig. 2.

Fig. 2.

A representation of free radicals being scavenged by endogenous antioxidants. Superoxide (O2-) reacts with nitric oxide (NO) to form peroxynitrate (ONOO-). Superoxide dismutase (SOD) catalyzes the reaction of O2- to hydrogen peroxide (H2O2), which participates in the formation of hydroxyl radicals (HO). Both catalase and glutathione peroxidase (GPx) reduce H2O2 to water (H2O) and oxygen (O2).

Therefore, a deficiency in SOD will lead to a decrease in NO bioavailability and diminishes vascular function. As an example, copper zinc SOD (CuZnSOD) deficient mice had a 2-fold increase in O2.- relative to their control littermates. Ultimately, this led to a decrease in dose-dependent EDD in the carotid artery [135]. Reduced SOD has also been associated with a number of pathologies including atherosclerosis, hypertension, and hypercholesterolemia [136]. Importantly, as mentioned previously aerobic exercise can increase SOD levels. In a study by Durrant et al. (2009) [103], old mice with access to a running wheel had greater levels of aortic SOD and lower levels of NADPH oxidases relative to their untrained littermates. This coincided with better dose-dependent EDD and higher levels of aortic eNOS and phosphorylated eNOS (Ser1177) expression [103].

H2O2, is the result of the dismutation of O2.- by SOD and elevated levels of H2O2 can also lead to oxidative stress [93] and vascular dysfunction [137]. The antioxidants, Glutathione peroxidase (GPx) and catalase are both capable of reducing H2O2 to oxygen and water. In humans, low levels of GPx are associated with an increased risk of CVD [138]. Furthermore, in mice, GPx deficiency led to a reduction in NO and a decrease in vascular function [139]. Similar to GPx, low levels of catalase are also associated with CVD [140]. Like SOD, several studies have shown that exercise increases levels of both GPx and catalase [141].

3.5 Protection from Arterial Stiffness

Arterial stiffness is a consistent independent predictor of all-cause mortality in individuals with hypertension [142]. Arterial stiffening is often associated with atherosclerosis, aging, smoking, obesity, and hyperlipidemia amongst other factors [143]. Over time, the structural properties of the vasculature can change. Collagen deposition in the tunica media and the degradation of elastin decreases the ability of arteries to dampen pulse waves and increases blood pressure [144]. Furthermore, chronic elevations in blood pressure increase LV overload which leads to the eventual development of LVventricular hypertrophy. Specifically, the loss of the ability to “dampen” a pulse wave in the aorta leaves organs with low vascular resistance vulnerable to injury [144]. One particular example is the kidneys where the exacerbation of damage is associated with the stiffening of both resistance arteries as well larger elastic arteries [145].

It has been established that exercise has the ability to slow down and help prevent vascular stiffening as well as decrease collagen levels in rodents [90, 91, 146] and humans [147, 148]. Further, arterial stiffness tends to be correlated with maximal aerobic capacity [130]. Fleenor et al. 2010 [146], found that 10–14 weeks of voluntary exercise was associated with decreased age-related vascular stiffness. Specifically, collagen I and III fibers were reduced. Another study examining a model of heart failure in mice discovered that 6 weeks of treadmill exercise was able to prevent the onset of aortic stiffening relative to sedentary mice [149]. Wheel running also protected young and old mice from arterial stiffness after consuming a Western-style diet (40% fat and 19% sucrose) for 10–14 weeks. Sedentary mice placed on a Western-style diet also had diminished EDD and NO bioavailability, exercise protected from losses in both. Lastly, rats placed on a high salt diet for 6 weeks experienced increased vascular stiffness and aortic collagen I protein expression [90]. All of these variables were attenuated when rats were given access to a running wheel during the same 6 weeks. Exercise-trained mice also had higher levels of aortic SOD2 protein expression when compared to sedentary rats who were placed on the same diet.

Arterial stiffening and oxidative stress tend to go hand in hand. Oxidative stress is a known initiator of vascular inflammation [150]. Studies have shown that antioxidant therapy is successful at decreasing oxidative stress and arterial stiffness. While this appears evident in animal models [131, 151] the results tend to be mixed in human trials [150, 152]. When TEMPOL (4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl), a superoxide dismutase mimetic was given to aging mice, not only was EDD improved there was lower levels of oxidative stress and large artery stiffness decreased [131]. Mitoquinone (MitoQ), an antioxidant which targets mitochondrial specific reactive oxygen species, not only reduced oxidative stress in aging mice but decreased aortic stiffness [153]. MitoQ was also shown to be effective in healthy older adults. Following chronic supplementation brachial flow-mediated dilation and aortic stiffness were lower [151].

Therefore, reduction and protection from arterial stiffness may be related to the ability of exercise to reduce oxidative stress. Spontaneous hypertensive rats had reduced vascular stiffness in the mesenteric and coronary arteries following 12 weeks of treadmill training. Authors found that these mice also had high NO bioavailability and less evidence of oxidative stress when compared to the spontaneous hypertensive rats who did not exercise [92]. Finally, voluntary wheel running reversed aortic stiffening in old mice. There was also a subsequent reduction in aortic O2 bioavailability [154].

4. Getting to the Heart of the Matter

4.1 The Gut-Heart Axis and CVD: An Update

We have previously reviewed the strong connection between the gut microbiome and cardiovascular disease, showing how dysbiosis and specific gut-derived metabolites can cause endothelial dysfunction, large artery stiffening, hypertension, and ultimately CVD [155]. Since our review on this topic, the literature has continued to evolve and continues to support a strong association between the gut microbiome and CVD. Here, we will summarize seminal new findings on the gut-heart axis since the publication of our previous review.

Studies since our last review have focused on understanding the role of gut microbial derived metabolites in CVD [156, 157, 158]. These studies have produced equivocal results with some metabolites like Indole-3-Propionic acid protecting against heart failure in patients with preserved ejection fraction [159], but others like butyrate showing no signs of altering, perhaps increasing CVD related diseases like hypertension [160] while gut microbial metabolite imidazole propionate (ImP) is increased in individuals with heart failure and is a predictor of overall survival [161].

With regards to studies associating specific gut microbiota to CVD, there have been some recent advances. Okami et al. [162], showed that as coronary artery calcification (CAC) scores rose in Japanese men, so did the Bacillota to Bacteroidota ratio, suggesting a relationship between higher gram-positive microbes and artery calcification. Given this is at such a high level of taxonomic resolution, the authors further reported that Lactobacillales were associated with a 1.3- to 1.4-fold higher risk of CVD and a higher CAC score. In addition, presence of Streptococcaceae and Streptococcus were linked to a higher risk of CVD while Enterobacteriaceae correlated with CAC scores. Sayols-Baixeras et al. [163], showed that Streptococcus anginosus and Streptococcus oralis had the strongest associations to CAC. Keeping in mind findings at the level of species and strain could be beneficial for the generation of -biotics, using bugs and drugs. Salvado et al. [164], showed that early vascular aging was associated with Bilophila, Faecalibacterium sp.UBA1819 and Phocea. Furthermore, when logistic regression analysis was completed, Bilophila remained significant. This is important because animal work has shown that Bilophila. wadsworthia caused systemic inflammation, suggesting the pathogenicity of this bacterium [165]. Guo et al. [166], showed that the genera Escherichia-Shigella, Lactobacillus, Enterococcus were more abundant in patients with resistant hypertension compared to normotensive adults.

While trimethylamine N-oxide (TMAO) continues to be a major gut microbial-derived metabolite of focus for CVD [167], an emerging metabolite phenylacetylgutamine (PAGln) has received a lot of attention recently [168]. In 2020, PAGln was discovered and is both associated with atherothrombotic heart disease in humans [169, 170, 171], and mechanistically linked to cardiovascular disease pathogenesis in animal models via modulation of adrenergic receptor signaling [172, 173]. Since then, Romano et al. [174] demonstrated that circulating PAGln levels were dose-dependently associated with heart failure presence and indices of severity (reduced ventricular ejection fraction, elevated N-terminal pro-B-type natriuretic peptide) independent of traditional risk factors and renal function, with associations between TMAO and incident heart failure being stronger among Black and Hispanic/Latino adults compared to White adults. Similar findings were shown by Tang et al. [175], which extended the work to show that PAGIn levels, independent of TMAO, may be used as a predictor of future CV events.

Despite these recent advances, mechanistic studies are still either in their infancy or lacking in the field and even more importantly studies which compare sex and race/ethnicity need urgent attention. Knowledge of which gut microbes may be involved is a good start, but understanding their function and role in the development of CVD is still lacking. Finally, there has been a lot of attention on ways to manipulate the gut microbiota via fecal transplants, symbiotics, probiotics, high-fiber diets and prebiotics, while this is outside the scope of this review, it has recently been reviewed elsewhere and the authors call your attention to Theofilis et al. [176].

4.2 Racial Variation in CVD

Despite trends for reductions in mortality rates from CVD in the US between 1980 and 2010, deaths attributable to CVD are once again on the rise. One pattern that has remained constant during this time is that racial and ethnic minority groups in the US (and globally) experience a disproportionate burden of CVD compared to their White counterparts [177, 178, 179, 180]. Overall, CVD prevalence remains highest among non-Hispanic Black women (59%) and non-Hispanic Black men (58.9%) [179, 181]. Black women and Black men are more than twice as likely to die of CVD, relative to White women and White men [179, 181] and among young and middle-aged adult survivors of a myocardial infarction, Black patients have a 2-fold higher risk of adverse outcomes [182].

It has been suggested that hypertensive target organ damage is widespread in Black and African American adults [183]. Young Black patients have an increasing burden of CVD risk factors [177]. Individuals of Black and African American ancestry experience hypertensive target organ damage earlier in life compared with White Americans [184]. Black/African American adults may also be more susceptible to the damaging effects of high blood pressure [185, 186]. Numerous studies note large disparities in measures of vascular health, with Black/African American adults displaying lower NO-mediated EDD and higher large artery stiffness and pressure from wave reflections compared with White Americans [187, 188, 189]. We and others have shown that disparities in these vascular health measures can be seen in childhood and correlate with proxies of target organ damage such as carotid intima-media thickness, LV mass, myocardial work, and coronary perfusion [190, 191, 192, 193]. Such “early vascular aging” in Black/African American adults likely serves as the catalyst for detrimental LV remodeling, heart failure, and future CVD [194]. For the past several decades, racial differences in CVD were ascribed to biological (“genetic”) differences (e.g., biological differences in inflammation, oxidative stress, NO metabolism, renin-angiotensin-aldosterone system, and autonomic nervous system function), neglecting the crucial role of the environment on risk [195, 196, 197]. It is now commonly recognized that cardiovascular health disparities are driven largely by deep-rooted structural racism and not race per se [178, 198, 199, 200].

Individuals who self-identify as members of a racial or ethnic minority group experience greater obstacles to health due to social, economic, and/or environmental disadvantages [199]. Systemic oppressive structures, policies, and practices in the US (i.e., social injustice) have created inequity in access to resources, services, and opportunities in minoritized (and marginalized) groups, driving disparities in SES and cardiovascular health [201]. Minority-related psychosocial stressors experienced by marginalized groups such as prejudice, discrimination, pressure to conform to a group stereotype by members of the same marginalized group, and pressure to acculturate/acculturation, are emerging as powerful risk factors for CVD and cardiovascular mortality [202]. Indeed, perceived discrimination is associated with increased risk for hypertension, systemic inflammation and oxidative stress, subclinical atherosclerosis, and detrimental vascular remodeling (increased carotid intima-media thickness, coronary artery calcification, and large artery stiffness), target organ damage, myocardial infarction, heart failure, and stroke [203, 204, 205, 206, 207]. Other factors related to structural racism such as lower SES, educational attainment, place of birth, neighborhood safety and food insecurity from residential segregation, and built environment (i.e., access to blue and green space, also shaped by neighborhood-level racial residential segregation) are barriers to ideal cardiovascular health [208, 209, 210, 211, 212, 213]. Moreover, each of these social determinants of health (SDoH) along with others such as stress from the incarceration of family or friends, job insecurity, violence in the home setting, and healthcare access are also associated with hypertension, inflammation, and oxidative stress, subclinical atherosclerosis, detrimental vascular remodeling, target organ damage, and ultimately CVD [214, 215, 216, 217, 218, 219, 220, 221]. We have recently shown that environmental toxicants found in higher concentrations in areas of lower SES are “cardiovascular disruptors” in children, contributing to altered vascular reactivity (greater blood pressure and vascular resistance in response to psychological stress) and subclinical CVD measured as carotid intima-media thickness at a young age [222, 223, 224, 225]. Additionally, we have shown that relative to White children, Black children have significantly greater hair cortisol levels and flatter diurnal slopes, which were in turn associated with subclinical CVD (measured as carotid intima-media thickness and aortic stiffness) [222]. Black children experienced significantly more environmental stress than White children with income inequality partially explaining the higher subclinical CVD risk in Black children [222]. Taken together, psychosocial determinants are the likely drivers of early (premature) vascular aging in Black and African American people in the US, some of which may be transmitted intergenerationally via biological (i.e., prenatal fetal programming) and social (i.e., early life adversity) mechanisms. This hypothesis is in keeping with minority stress theory and the weathering hypothesis whereby chronic exposure to social and economic disadvantage leads to increased allostatic load and accelerated biological (and physiological) “wear and tear” on end organs causing inflammation and oxidative stress, hastening aging [226].

4.3 Racial Variation in the Gut-Heart Axis: Implications for CVD

This section will examine racial variation in the gut microbiome with consideration for how the systemic environment (i.e., structural racism) impacts the microbial environment to perpetuate cardiovascular health disparities. As introduced above, there is growing evidence that the social and environmental gradients which contribute to health inequities also predict gut microbiota traits [227]. Evidence shows that the human microbiome variation is linked to the incidence, prevalence, and mortality of many diseases and is associated with race and ethnicity in the US. To date, there have been several studies (discussed next) that have examined this outcome and have identified gut microbiota profiles shaped by host environments which affect host metabolic, immune, and neuroendocrine functions, making it an important pathway by which differences in experiences caused by social, political, and economic forces could contribute to health inequities.

It is thought that the gut microbiota is well established by the time a child is 4 years old, and there is strong evidence that maternal, and family socioeconomic status can influence gut microbiota. Several investigators have analyzed data from the Food and Microbiome Longitudinal Investigation (FAMiLI) study to obtain answers on how maternal family and SES influences the gut. FAMiLI is an ongoing multi-ethnic prospective study in the US that began in 2016 where participants complete demographic questionnaires and (optional) food frequency questionnaires and provide oral and stool samples. In 2020, Peters et al. [228], analyzed samples from 863 US residents, including US-born (315 White, 93 Black, 40 Hispanic) and foreign-born (105 Hispanic, 264 Korean). The authors determined dietary acculturation from dissimilarities based on food frequency questionnaires and used 16S rRNA gene sequencing to characterize the microbiome [228]. Their results showed a clear difference in gut microbiome composition across study groups. They found the largest differences in gut microbiota between foreign-born Koreans and US-born Whites, and significant differences were also observed between foreign-born and US-born Hispanics. Specifically, differences in sub-operational taxonomic unit (s-OTU) abundance between foreign-born and US-born groups tended to be distinct from differences between US-born groups. Bacteroides plebeius, a seaweed-degrading bacterium, was strongly enriched in foreign-born Koreans, while Prevotella copri and Bifidobacterium adolescentis were strongly enriched in foreign-born Koreans and Hispanics, compared with US-born Whites. Dietary acculturation in foreign-born participants was associated with specific s-OTUs, resembling abundance in US-born Whites; e.g., a Bacteroides plebeius s-OTU was depleted in highly diet-acculturated Koreans. The authors concluded that US nativity is a determinant of the gut microbiome in a US resident population.

The “sociobiome” was coined by Nobre and Alpuim Costa [229] to describe the microbiota composition occurring in residents of a neighborhood or geographic region due to similar socioeconomic exposures; socioeconomic status. Recently, Kwak et al. [230], using the FAMiLI cohort, investigated the sociobiome in a large, multi-ethnic sample. The cohort consisted of 825 adults (36.7% male), with a mean age of 59.6 years and racial and ethnic group composition consisting of 311 (37.7%) non-Hispanic White, 287 (34.8%) non-Hispanic Asian, 89 (10.8%) non-Hispanic Black, and 138 (16.7%) Hispanic participants and compared alpha-diversity, beta-diversity, and taxonomic and functional pathway abundance by SES. They showed that lower SES was significantly associated with greater α-diversity and compositional differences among groups, as measured by β-diversity. Several taxa related to low SES were identified, especially an increasing abundance of Prevotella copri and Catenibacterium sp000437715, and decreasing abundance of Dysosmobacter welbionis in terms of their high log-fold change differences. This is significant as Dysosmobacter welbionis was isolated from human commensal bacterium from samples provided by the Human Microbiome Project, American Gut Project, Flemish Gut Flora Project and Microbes4U projects. This bacterium was detected in 62.7%–69.8% of the healthy population and correlates negatively with body mass index, fasting glucose and glycated hemoglobin. In addition, Cani’s group using the humanized mouse model, taking human fecal samples/strains and putting them into a mouse, showed that Dysosmobacter welbionis prevented diet-induced obesity and metabolic disorders in mice by reducing fat mass gain, insulin resistance and white adipose tissue hypertrophy and inflammation [231]. In addition, live Dysosmobacter welbionis administration protected the mice from brown adipose tissue inflammation in association with increased mitochondria number and non-shivering thermogenesis. While this has yet to be translated to humans, the reduction of this bacteria in the human study coupled with its actions seen in animal studies suggest that the lack of this bacteria may place individuals at increased risk for metabolic disorders and adipose tissue dysfunction which could lead to adverse CVD outcomes.

Most recently, Mallott et al. [232], set out to determine the age at which microbiome variability emerges between race and ethnic groups. They used 8 datasets with 16S ribosomal RNA (rRNA) sequencing data and available race and ethnicity metadata for this study. Individuals between birth and 12 years of age, living in the US, with a caregiver-reported race of Black, White, or Asian/Pacific Islander, and with a caregiver-reported ethnicity of Hispanic or non-Hispanic were included in the analysis. They found that race and ethnicity did not significantly vary with gut microbiome alpha-diversity or beta-diversity in the early weeks and months of life, including the first week, 1 to 5.9 weeks, and 6 weeks to 2.9 months, however, at 3 to 11.9 and 12 to 35.9 months, gut microbiome composition varied slightly but significantly by both race and ethnicity. The group concluded that race and ethnicity are associated with gut microbiome composition and diversity beginning at 3 months of age, indicative of a narrow window of time when this variation emerges [232].

Finally, discrimination and stress have been found to contribute to changes in gut microbiota among racial and ethnic groups [233, 234]. A study by Dong et al. [235], examined 154 adults from the Los Angeles community and clinics. Participants self-reported race and ethnicity (Asian American, Black, Hispanic, or White) and discrimination was measured using the Everyday Discrimination Scale. Hispanic individuals self-reported the highest levels of early-life adversity, while Black individuals reported the highest levels of resilience. Microbiome and metabolite differences related to discrimination were only apparent when stratified by race/ethnicity. Results showed that Prevotella copri was the highest in Black and Hispanic individuals, who experienced high levels of discrimination, whereas White individuals reported low levels of discrimination. Isovalerate and valerate were significantly lower in Hispanic than in White individuals and fucosterol was significantly higher in Asian rather than White individuals. In a related study, Zhang et al. [236], investigated the impact of discrimination exposure on brain reactivity to food images and associated dysregulations in the brain–gut–microbiome axis. By employing multi-omics analyses of neuroimaging and fecal metabolite, they showed that discrimination is associated with increased food-cue reactivity in regions of the brain important for reward, motivation and executive control; altered glutamate-pathway metabolites involved in oxidative stress and inflammation as well as a preference for unhealthy foods. In addition, the relationship between discrimination-related brain and gut signatures was shifted towards unhealthy sweet foods after adjusting for age, diet, body mass index, race and SES. Given the extensive literature on diet, obesity and the gut microbiota, these results are significant in suggesting that individuals facing discrimination may prefer unhealthy foods (and/or may not have access to healthy foods) contributing to a more dysbiotic gut and thus adverse cardiometabolic health outcomes.

In conclusion, there are distinct gut microbiota profiles between racial and ethnic groups, which appear to be influenced by acculturation [237, 238, 239], discrimination and stress [233, 234], and diet [240], which may occur as early as 3 months of age. Where a person lives and the related neighborhood and environmental constraints, what stresses they are exposed to, and what a person eats (both what they choose to eat and what they have access to eat) may shape the gut microbiome more than race or ethnicity per se. Finally, these distinct gut microbial community structures can exacerbate CVD risk among minority racial and ethnic groups [241] (Fig. 3).

Fig. 3.

Fig. 3.

Working conceptual model. Race, ethnicity, gender, and sex interact (i.e., intersectionality) and are shaped by social determinants of health (SDoH) to moderate gut effects (dysbiosis, diversity, specific metabolites, gut “age”) on subclinical cardiovascular disease (CVD) (endothelial dysfunction, large artery stiffness) - driving CV health disparities and overt CVD (hypertension, coronary ischemia and vasospasm, myocardial infarction, heart failure). CV, cardiovascular.

4.4 Biological Sex, Gender and CVD

Another prejudice that has a profound impact on health and CVD risk is sexism [242]. Women, in general, have also been historically marginalized due to institutionalized patriarchy and a male-dominated social system. When considering the impact of sexism on CVD, we must first operationalize and contextualize differences (and overlap) between biological sex and gender. Sex, when considered biologically, comprises genetic differences related to chromosomes, gonadal structure and function, and hormonal sequela. We will conceptualize sex as referring to male, female, and intersex. Gender is a social construct based on sociocultural predetermined roles, relationships, and stereotypes (e.g., masculine versus feminine). Gender can be shaped by different power dynamics and how we interact with others around us based on ascribed gender and can vary based on regionality, nationality, and temporality (i.e., ideals can change over time). Gender also encompasses gender identity referring to a person’s inner sense of self as a man, woman, nonbinary person, or agender person among other identities. Sex and gender can be considered together to inform on both biological sex and self-identified gender. For example, a person who identifies as a cis-gender woman is a woman whose self-identified gender aligns with the biological sex assigned at birth.

In the context of CVD, biological sex and gender may converge to affect risk [243, 244]. Women are typically believed to be at lower risk for CVD owing to the biological effects of the gonadal hormone estrogen. Note here that we do not consider estrogen a sex hormone per se as both men and women produce estrogen (and testosterone), just in varying amounts. Just as low estrogen is associated with increased risk for coronary heart disease and CVD mortality in older men [245], low testosterone is associated with a greater risk of ischemic CVD and major adverse cardiovascular events in older women [246, 247]. Subsequently, CVD risk increases in women with advancing age, particularly post-menopause. With that said, it should be highlighted that CVD remains the leading cause of mortality in women of all ages, and hospitalizations and deaths attributed to CVD have witnessed an increase for younger and middle-aged women [248]. The reasons for these observations are likely multifactorial and may partly be related to societal sex- and gender-based discriminatory attitudes [249]. Not until the American Heart Association’s “Go Red” campaign has there been equitable education and promotion of CVD risk for women. As such, educational efforts on signs, symptoms, risk factors, and consequences of CVD in women were sparse. This may have contributed to increased CVD risk factor burden in women and women being less likely to seek timely medical care for signs and symptoms related to CVD. As cardiology is still a predominantly male workforce drawing from scientific literature where women are underrepresented, implicit bias may affect clinical decision-making. For example, signs of myocardial infarction are often categorized as “atypical” in women not because they are abnormal but because they are different from men, with male symptomology being construed as the norm. Some male physicians may also incorrectly assume that a younger/middle-aged woman presenting with chest pain cannot be having a myocardial infarction because that would go against the entrenched dogma that estrogen is cardioprotective. As a result, when seeking care, women have longer wait times when presenting with chest pain, are more likely to be misdiagnosed, more likely to have symptomology dismissed, and are less likely to be prescribed medications or treatments known to mitigate risk [250]. Women are also less likely to be referred to cardiac rehabilitation after a cardiac event [251, 252]. Together, all of these factors contribute to women having poorer outcomes after a cardiovascular event compared to men.

Women are more likely to develop concentric LV remodeling and heart failure with preserved ejection fraction than men [253]. The pathophysiology of coronary artery disease also differs by sex with women possibly having coronary endothelial dysfunction and microvascular defects compared to men, contributing to sexual dimorphism in acute coronary syndromes [254]. While premenopausal women may have better endothelial function than men [255], we and others have shown that women may have greater pressure from wave reflections increasing central hemodynamic load [256, 257, 258]. Sex differences in central hemodynamic burden may contribute to greater LV diastolic dysfunction and associations between arterial stiffness and LV mass/LV diastolic dysfunction may be greater in women compared to men [259, 260, 261]. Large artery stiffness increases disproportionately in postmenopausal women and the association between large artery stiffness and CVD mortality is almost twofold higher in women versus men [262]. As noted above, it is difficult to parse out how much CVD risk is attributable to sex and how much to gender. Some CVD risk in this setting has been suggested to be related to stature (e.g., smaller coronary arteries experiencing more shear stress, shorter aortic length contributing to greater pressure from wave reflections) [263, 264], which may be theorized to be biologically driven. Some CVD risk may be related to the physiological response to mental stress [265, 266, 267], which may be influenced by psychosocial determinants of health. Myocardial ischemia and peripheral microvascular endothelial dysfunction in response to mental stress are greater in women compared to men and associated with major adverse cardiovascular events in women only [268]. Taken together, CVD risk in women likely captures the interaction of both sex and gender on cardiovascular structure and function.

While traditional risk factors (age, lipids, glucose, smoking, blood pressure) affect CVD risk in women and men similarly, there are also sex-specific risk factors that are critically important to consider for women [269]. Sex-specific risk factors relate to biological variation in reproductive health factors and are uniquely ascribed to female biological sex [270]. Such risk factors may include adverse pregnancy outcomes (e.g., hypertensive disorders of pregnancy, gestational diabetes, fetal growth restriction, preterm delivery, and placental abruption), premature menarche, premature menopause and vasomotor symptoms, endometriosis and polycystic ovarian syndrome [270]. Additionally, there are other emerging CVD risk factors caused by other comorbidities and social factors that are more prevalent in women and may be influenced by both sex and gender. These factors include autoimmune disorders, migraine, fibromyalgia, postural orthostatic tachycardia syndrome, osteoporosis, breast cancer, irritable bowel syndrome, abuse, intimate partner violence, post-traumatic stress disorder, anxiety, and depression [270]. Each of the aforementioned female sex-specific and female sex-prevalent risk factors is associated with increased risk for hypertension, systemic inflammation and oxidative stress, subclinical atherosclerosis, and detrimental vascular remodeling (increased carotid intima-media thickness, coronary artery calcification, and large artery stiffness), target organ damage, myocardial infarction, heart failure, and stroke [271].

When considering intersectionality, Black and Hispanic women may encounter “double jeopardy” due to the combination of race and ethnicity bias, coupled with sex and gender bias [272]. Minority women experience additional ethnic, racial and gender constraints and risks including reduced health care access, possible language barriers, lower health literacy, racial discrimination, pressure to acculturate or conform to both a racial and culturally gendered identity, higher reports of depression and higher incidence of pregnancy complications (e.g., hypertensive disorders of pregnancy) [273, 274]. As stated above, these SDoH are also CVD risk factors and are as important and sometimes more important correlates of subclinical CVD in women [275, 276, 277, 278, 279, 280, 281]. As such, the prevalence of sex-specific CVD risk factors, coronary artery disease, heart failure, and stroke is highest among non-Hispanic Black women [282]. As stated by the American Heart Association, to understand and address the root causes of the prominent disparities in CVD outcomes between Black and White women and men in the United States, the intersectional aspects between race, sex, and gender must be considered [283]. Nearly 60% of Black women have CVD, contributing to a persistent life expectancy gap in the US [181]. Current life expectancy for Non-Hispanic Black women is 75 years on average compared with 80 years for non-Hispanic White women [269]. CVD is also the most prominent cause of mortality amongst Hispanic women, with approximately 42% of Hispanic women having CVD [181]. Paradoxically, despite a higher prevalence of such traditional CVD risk factors such as diabetes, obesity, and metabolic syndrome, CVD death rates in Hispanic women have remained 15% to 20% lower than in non-Hispanic White women - an observation commonly referred to as the Hispanic Paradox [284]. Interestingly, we have seen that young Hispanic women have better endothelial function and lower large artery stiffness compared to White women [285], suggesting that traditional CVD risk factors may not capture actual CVD risk in this population. It should be noted that this paradox is disappearing as Hispanic American individuals acculturate and adopt the high-fat, sedentary lifestyle of those with US nativity [286]. As noted above, sex differences in the vascular response to mental stress are a predictor of major adverse cardiovascular events in women. Endothelial dysfunction in response to mental stress is also a predictor of adverse CV outcomes in Black adults, explaining 69% of their excess risk [287]. Notable predictors of the development of transient endothelial dysfunction with mental stress beyond Black race include female gender, employment status, income, and a composite distress score derived from post-traumatic stress disorder, depression, anxiety, anger, perceived stress and racial discrimination [288, 289, 290, 291]. These findings highlight the importance of intersectionality and psychosocial determinants of vascular health impacting CVD risk in women, particularly Black women.

There is also emerging evidence that lesbian, gay, bisexual, transgender, and queer or questioning (LGBTQ+) adults, as a stigmatized and marginalized group, experience notable cardiovascular health disparities [292, 293]. According to the American Heart Association, people who are transgender and gender diverse may be at greater risk for CVD [294]. There is growing evidence that LGBTQ+ adults experience worse cardiovascular health relative to their cisgender heterosexual peers [292, 295]. For example, men who are transgender have a >2-fold and 4-fold increase in the prevalence of myocardial infarction compared with men who are cisgender and women who are cisgender, respectively. Conversely, women who are transgender have >2-fold increase in the prevalence of myocardial infarction compared with women who are cisgender. Moreover, compared to heterosexuals, sexual minorities are at a higher risk of hypertension and CVD and more likely to develop CVD at an earlier age [296, 297]. It should be underscored that the LGBTQ+ (intersexual, asexual, pansexual, two spirit) community is not a monolithic group [298]. Each has unique lived experiences that may subsequently shape CVD risk. Differences in CVD risk are partially, but not completely, explained by traditional CVD risk factors suggesting that SDoH plays a significant role. LGBTQ+ adults not only experience significantly higher discrimination from the broader community, but also specifically from healthcare professionals [299]. Additional psychosocial risk factors including self-stigma and internalized phobia, gender-related victimization, expectations of rejection, and concealment, all detrimentally impact mental health (anxiety, depression) and behavioral health (diet, sleep, physical activity, alcohol and tobacco/nicotine use) [300, 301]. Together, these factors may contribute to inflammation and oxidative stress, hastened vascular aging, subclinical atherosclerosis, target organ damage and overt CVD [302, 303].

Biological effects of gender-affirming hormone therapy (GAHT) may also have an impact on CVD risk [304, 305]. Use of GAHT in transgender and nonbinary individuals is perceived to improve cardiovascular health [306]. The association between GAHT and CVD risk is complex [307]. A higher blood concentration of testosterone among women who are transgender is associated with higher odds of having hypertension. Cross-sectional comparisons between men who are transgender receiving testosterone cypionate compared with age-matched women who are cisgender have found reduced endothelial function measured via brachial artery flow-mediated dilation [308]. In cross-sectional studies, carotid intima-media thickness, arterial stiffness and measured via brachial-ankle pulse wave velocity, and carotid augmentation index are higher in men transitioning (female to male) receiving testosterone than in men who are transgender not receiving hormone therapy [309, 310, 311]. Similarly, transgender men on long-term treatment with testosterone have higher aging-related aortic stiffening [312], suggesting accelerated vascular aging in transgender men receiving gender-affirming hormone treatment. This is supported by animal studies noting that female mice receiving dihydrotestosterone experience hastened rates of arterial stiffening and cardiovascular damage, mediated by decreased estrogen receptor expression [313]. Brachial artery flow-mediated dilation is higher in women who are transgender treated with estrogen than in age-matched men who are cisgender but is similar to women who are cisgender [314, 315]. Women who are transgender receiving estrogen also have a greater forearm blood flow response to acetylcholine, an endothelial-dependent vasodilator, than age-matched men who are cisgender [314]. In summary, GAHT is associated with an increased risk of subclinical atherosclerosis in transgender men but may have either neutral or beneficial effects in transgender women [316].

4.5 Biological Sex, Gender and the Gut-Heart Axis: Implications for CVD

This section will consider the mediating and moderating effects of sex, sex-specific CVD risk factors, and gender (operationalized as sexual orientation and gender identity) on the gut microbiome as an effector of CVD risk (Fig. 3). As stated above, there are notable sex differences in gut microbiota across a lifespan, and these differences may serve, in part, as the substrate for sex differences in CVD risk across a lifespan. The distribution of gut microbiota varies according to age (childhood, puberty, pregnancy, menopause, and old age) and sex. Also, as already established, this gut microbiota can contribute and is linked to CVD. It is critical to understand which gut microbiota and/or microbial derived metabolites may be linked to CVD in the sexes. To that end, Garcia-Fernandez et al. [317], analyzed gut microbiota data from the CORDIOPREV study, a clinical trial which involved 837 men and 165 women with CVD compared to their reference group of 375 individuals (270 men, 105 women) without CVD. They clearly demonstrated a sex-specific difference in beta diversity. Additional analysis showed there were sex-specific alterations in the gut microbiota linked to CVD. Women who have CVD show increased UBA1819 (Ruminococcaceae), Bilophila, Phascolarctobacterium, and Ruminococcaceae incertae sedis while men with CVD had a higher abundance of Subdoligranulum, and Barnesiellaceae. The authors concluded that the dysbiosis of the gut microbiota associated with coronary heart disease (CHD) seems to be partially sex-specific, which may influence the sexual dimorphism in its incidence particularly since the bacteria identified to be higher in CVD patients are linked to inflammation, intestinal barrier dysfunction, and CVD directly [317, 318].

The dysbiotic gut microbiome is associated with increased blood pressure and risk of hypertension [319]. Virwani et al. [320], specifically examined sex differences, gut microbiota and hypertension. Interestingly they reported that significant differences in beta-diversity and gut microbiota composition in hypertensive versus normotensive groups were only observed in women and not in men. Specifically, Ruminococcus gnavus, Clostridium bolteae, and Bacteroides ovatus were significantly more abundant in hypertensive women, whereas Dorea formicigenerans was more abundant in normotensive women. Furthermore, total plasma short-chain fatty acids and propionic acid were independent predictors of systolic and diastolic blood pressure in women but not men. Ruminococcus gnavus and Clostridium bolteae have been reported to induce inflammation and are pathogenic in humans. Gut microbial-derived metabolites are likely critical to affect the way gut microbiota influences systemic disease states. As noted above, butyrate may exacerbate hypertension, as propionate has also been demonstrated in this study [160]. However, the mechanisms by which this occurs are not elucidated, but need to be to fully understand the interactions of these SCFA and hypertension outcomes in women.

In addition to sex differences in gut microbiota and CVD, there are also sex differences in many of the risk factors associated with CVD of which most have associations with the gut microbiota including diabetes, hypertension and dyslipidemia, and obesity (see review by Ahmed and Spence [321]), which may be further exacerbated by race and ethnicity [322]. In addition, sex-specific CVD risk factors related to maternal health during pregnancy may also influence and be influenced by the gut microbiome. In 2023, Colonetti et al. [323], conducted a meta-analysis which included 6 studies, with 479 pregnant women. They reported a significantly lower gut microbiota alpha diversity in pregnant women with pre-eclampsia in comparison with healthy controls, while no significant differences were found in the relative abundance of Bacteroidota, Bacillota, Actinomycetota, and Pseudomonadota, despite significant differences being reported in the individual studies [323]. However, this could be due to a number of factors, most significantly the analytical techniques used to identify lower levels of taxonomic resolution that vary greatly between gut microbiota studies. A rodent study by Jama et al. [324], examined female C57BL/6J dams fed nutrient-matched high- or low-fiber diets during pregnancy and lactation, to understand how maternal fiber influences the gut microbiota. In addition, to evaluate long-term effects and predisposition to CVD, the authors exposed 6-week-old male offspring to saline or angiotensin II for 4 weeks to induce hypertension and organ damage. Results showed that male offspring from low-fiber-fed dams had significantly larger hearts relative to body weight, and echocardiography studies in the offspring demonstrated low-fiber offspring had increased LV posterior wall thickness, confirming hypertrophy, and reduced ejection fraction, showing reduced LV contraction [324]. Regarding the gut microbiota, offspring born to dams who received a low-fiber diet showed distinct gut microbial colonization that persisted into adulthood, with higher levels of several taxa, including Akkermansia species. Furthermore, the authors reported that they identified 174 microbial enzymatic pathway signatures enriched in low-fiber offspring with 154 of the identified enzyme signatures in low-fiber belonged to Akkermansia muciniphila. Akkermansia muciniphila-upregulated genes encoded for mucolytic enzymes that degrade the intestinal mucus, putting the colon at risk for inflammation [324]. In contrast, high-fiber offspring had only 5 grouped enzyme signatures, which belonged to Bacteroides ovatus, Escherichia coli, and Lactobacillus murinus; the latter of which has been known to reduce inflammatory pathways and blood pressure. The gut microbiota of women with hypertensive disorders of pregnancy is different from that of women with normotensive pregnancy [325]. Pregnant women with hypertensive disorders of pregnancy had a higher abundance of Rothia, Actinomyces, and Enterococcus and a lower abundance of Coprococcus than pregnant women with normotension [325]. Indeed, results from Mendelian randomization support a causal relationship between gut microbiota and hypertensive disorders of pregnancy [326]. Wu et al. [326] found causal associations of LachnospiraceaeUCG010, Olsenella, RuminococcaceaeUCG009, Ruminococcus2, Anaerotruncus, Bifidobacterium, and Intestinibacter with gestational hypertension, of Eubacterium (ruminantium group), Eubacterium (ventriosum group), Methanobrevibacter, RuminococcaceaeUCG002, and Tyzzerella3 with preeclampsia, and of Dorea and RuminococcaceaeUCG010 with eclampsia, respectively. These findings are supported by experimental studies whereby fecal microbiota transplantation from preeclamptic women into preeclamptic rats significantly exacerbated the phenotype whereas the gut microbiota of healthy pregnant women had significant protective effects [327]. Akkermansia muciniphila, propionate, or butyrate significantly alleviated the symptoms of preeclamptic rats whereas Akkermansia, Oscillibacter, and SCFAs could be used to accurately diagnose preeclampsia [327]. Taken together, recent findings support that gut dysbiosis is important in the etiology of preeclampsia, a significant sex-specific risk factor for CVD in women.

To date there are very few studies examining gut microbiota and gender (operationalized as sexual orientation and gender identity) hence research in this area is greatly needed. Rosendale et al. [328], recently published a cross-sectional study of 12,180 adults using 2007–2016 National Health and Nutrition Examination Survey data, Black, Hispanic, and White sexual minority female individuals with the primary outcome of overall cardiovascular health score. Results showed that Black, Hispanic, and White sexual minority female adults had lower overall cardiovascular health scores compared with their heterosexual counterparts. Furthermore, there were no differences in overall cardiovascular health scores for sexual minority male individuals of any race or ethnicity compared with White heterosexual male individuals [328]. It is important to mention that there are even fewer studies on GAHT and gut microbiota [329], and none to our knowledge which include CVD which is an area of research importance.

5. Future Directions

The mantra “exercise is medicine” is often touted as a solution to restore cardiovascular health and prevent disease. Indeed, as discussed above, exercise has a powerful effect on improving gut health, attenuating vascular aging, improving large artery compliance and systemic vascular endothelial function through its antioxidant effects, and preserving nitric oxide bioavailability - all reducing the risk for CVD. However, exercise (like medicine) is not accessible to all and exercise is not medicine for all. Black adults, Hispanic adults, and women in general are not meeting physical activity recommendations. Unique social barriers such as neighborhood dynamics (safety and cohesion) may contribute to disparities in physical activity engagement across different races and ethnicities [330, 331]. There is also considerable heterogeneity in the response to exercise across race and sex [332]. For example, while women may have a blunted cardiovascular physiological response to exercise training compared to men [333], women derive greater protection against CVD mortality from that same amount of exercise [334]. Indeed, the female athlete’s heart has a lower risk of experiencing exercise-induced coronary calcification, LV fibrosis, atrial fibrillation, lethal ventricular arrhythmias and sudden cardiac death. There is also racial variation in the cardiovascular response to acute exercise and exercise training [335, 336]. Some of the differences in cardiovascular responses to exercise may be related to the physiological impact of various psychosocial factors [337, 338]. For example, racial discrimination is associated with oxidative stress and endothelial damage [339, 340]. Future research is needed to explore racial variation and sex differences in the gut microbiome’s response to exercise. Can targeting the gut with diet (e.g., prebiotics), probiotics and/or exercise confer cardiovascular resilience? Additional research is also needed to examine the effect of the gut microbiome on cardiovascular responses to exercise training. Does underlying dysbiosis mediate or moderate heterogeneity in physiological adaptations to exercise training? Additional research will also be needed to understand the importance of intersectionality on the gut microbiome, considering race, ethnicity, sex and gender.

6. Conclusions

Studies continue to support that gut dysbiosis is a CVD risk factor, with numerous microbes impacting unique aspects of cardiovascular structure and function. The gut microbiome is shaped by biological sex, gender, race and ethnicity, potentially contributing to cardiovascular health disparities and sex differences in CVD. Psychosocial factors related to systemic racism, sexism, and discrimination impact the microbiome via effects on diet and food access. These same factors may also activate physiological stress systems, contributing to inflammation, oxidative stress, subclinical changes in vascular structure and function (i.e., EDD and arterial stiffening) and ultimately CVD.

To conclude, sociology impacts physiology and contributes to pathophysiology. Oppressive social factors experienced by minorities and women may shape the gut, in turn contributing to cardiovascular health disparities. Exercise remains a critical lifestyle and biobehavioral factor to promote gut resilience and foster cardioprotection.

Acknowledgment

The authors would like to thank Dr. Peter Kokkinos, FACSM, FAHA for the invitation to write this review.

Footnotes

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author Contributions

JG, KH and SC designed the study. All authors are involved in drafting the manuscript and revising it critically for important intellectual content. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References

  • [1].Lindstrom M, DeCleene N, Dorsey H, Fuster V, Johnson CO, LeGrand KE, et al. Global Burden of Cardiovascular Diseases and Risks Collaboration, 1990-2021. Journal of the American College of Cardiology . 2022;80:2372–2425. doi: 10.1016/j.jacc.2022.11.001.. [DOI] [PubMed] [Google Scholar]
  • [2].Piercy KL, Troiano RP, Ballard RM, Carlson SA, Fulton JE, Galuska DA, et al. The Physical Activity Guidelines for Americans. JAMA . 2018;320:2020–2028. doi: 10.1001/jama.2018.14854.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Paffenbarger RS, Jr, Blair SN, Lee IM. A history of physical activity, cardiovascular health and longevity: the scientific contributions of Jeremy N Morris, DSc, DPH, FRCP. International Journal of Epidemiology . 2001;30:1184–1192. doi: 10.1093/ije/30.5.1184.. [DOI] [PubMed] [Google Scholar]
  • [4].MORRIS JN, RAFFLE PA. Coronary heart disease in transport workers; a progress report. British Journal of Industrial Medicine . 1954;11:260–264. doi: 10.1136/oem.11.4.260.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Guan Y, Yan Z. Molecular Mechanisms of Exercise and Healthspan. Cells . 2022;11:872. doi: 10.3390/cells11050872.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Myers J, Prakash M, Froelicher V, Do D, Partington S, Atwood JE. Exercise capacity and mortality among men referred for exercise testing. The New England Journal of Medicine . 2002;346:793–801. doi: 10.1056/NEJMoa011858.. [DOI] [PubMed] [Google Scholar]
  • [7].Wosinska L, Cotter PD, O’Sullivan O, Guinane C. The Potential Impact of Probiotics on the Gut Microbiome of Athletes. Nutrients . 2019;11:2270. doi: 10.3390/nu11102270.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Oren A, Garrity GM. Valid publication of the names of forty-two phyla of prokaryotes. International Journal of Systematic and Evolutionary Microbiology . 2021;71:10.1099/ijsem.0.005056. doi: 10.1099/ijsem.0.005056.. [DOI] [PubMed] [Google Scholar]
  • [9].Dalton A, Mermier C, Zuhl M. Exercise influence on the microbiome-gut-brain axis. Gut Microbes . 2019;10:555–568. doi: 10.1080/19490976.2018.1562268.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Campbell SC, Wisniewski PJ., 2nd Exercise is a Novel Promoter of Intestinal Health and Microbial Diversity. Exercise and Sport Sciences Reviews . 2017;45:41–47. doi: 10.1249/JES.0000000000000096.. [DOI] [PubMed] [Google Scholar]
  • [11].Allen JM, Berg Miller ME, Pence BD, Whitlock K, Nehra V, Gaskins HR, et al. Voluntary and forced exercise differentially alters the gut microbiome in C57BL/6J mice. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2015;118:1059–1066. doi: 10.1152/japplphysiol.01077.2014.. [DOI] [PubMed] [Google Scholar]
  • [12].Hsu YJ, Chiu CC, Li YP, Huang WC, Huang YT, Huang CC, et al. Effect of intestinal microbiota on exercise performance in mice. Journal of Strength and Conditioning Research . 2015;29:552–558. doi: 10.1519/JSC.0000000000000644.. [DOI] [PubMed] [Google Scholar]
  • [13].Allen JM, Mailing LJ, Cohrs J, Salmonson C, Fryer JD, Nehra V, et al. Exercise training-induced modification of the gut microbiota persists after microbiota colonization and attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes . 2018;9:115–130. doi: 10.1080/19490976.2017.1372077.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Barton W, Penney NC, Cronin O, Garcia-Perez I, Molloy MG, Holmes E, et al. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut . 2018;67:625–633. doi: 10.1136/gutjnl-2016-313627.. [DOI] [PubMed] [Google Scholar]
  • [15].Bressa C, Bailén-Andrino M, Pérez-Santiago J, González-Soltero R, Pérez M, Montalvo-Lominchar MG, et al. Differences in gut microbiota profile between women with active lifestyle and sedentary women. PloS One . 2017;12:e0171352. doi: 10.1371/journal.pone.0171352.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Langsetmo L, Johnson A, Demmer RT, Fino N, Orwoll ES, Ensrud KE, et al. The Association between Objectively Measured Physical Activity and the Gut Microbiome among Older Community Dwelling Men. The Journal of Nutrition, Health & Aging . 2019;23:538–546. doi: 10.1007/s12603-019-1194-x.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Bai J, Hu Y, Bruner DW. Composition of gut microbiota and its association with body mass index and lifestyle factors in a cohort of 7-18 years old children from the American Gut Project. Pediatric Obesity . 2019;14:e12480. doi: 10.1111/ijpo.12480.. [DOI] [PubMed] [Google Scholar]
  • [18].Campbell SC, Wisniewski PJ, Noji M, McGuinness LR, Häggblom MM, Lightfoot SA, et al. The Effect of Diet and Exercise on Intestinal Integrity and Microbial Diversity in Mice. PloS One . 2016;11:e0150502. doi: 10.1371/journal.pone.0150502.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Huang WC, Chen YH, Chuang HL, Chiu CC, Huang CC. Investigation of the Effects of Microbiota on Exercise Physiological Adaption, Performance, and Energy Utilization Using a Gnotobiotic Animal Model. Frontiers in Microbiology . 2019;10:1906. doi: 10.3389/fmicb.2019.01906.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Okamoto T, Morino K, Ugi S, Nakagawa F, Lemecha M, Ida S, et al. Microbiome potentiates endurance exercise through intestinal acetate production. American Journal of Physiology. Endocrinology and Metabolism . 2019;316:E956–E966. doi: 10.1152/ajpendo.00510.2018.. [DOI] [PubMed] [Google Scholar]
  • [21].Dowden RA, Wisniewski PJ, Longoria CR, Oydanich M, McNulty T, Rodriguez E, et al. Microbiota Mediate Enhanced Exercise Capacity Induced by Exercise Training. Medicine and Science in Sports and Exercise . 2023;55:1392–1400. doi: 10.1249/MSS.0000000000003170.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Nay K, Jollet M, Goustard B, Baati N, Vernus B, Pontones M, et al. Gut bacteria are critical for optimal muscle function: a potential link with glucose homeostasis. American Journal of Physiology. Endocrinology and Metabolism . 2019;317:E158–E171. doi: 10.1152/ajpendo.00521.2018.. [DOI] [PubMed] [Google Scholar]
  • [23].Hutchinson NT, Wang SS, Dokhanchi J, Johnson RW, Buford TW, Allen JM, et al. Effects of Broad-Spectrum Antibiotic Treatment or Germ-Free Status on Endurance Performance and Exercise Adaptations in Mice. Medicine and Science in Sports and Exercise . 2023;55:225–234. doi: 10.1249/MSS.0000000000003051.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Dohnalová L, Lundgren P, Carty JRE, Goldstein N, Wenski SL, Nanudorn P, et al. A microbiome-dependent gut-brain pathway regulates motivation for exercise. Nature . 2022;612:739–747. doi: 10.1038/s41586-022-05525-z.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Diduch BK. Gastrointestinal Conditions in the Female Athlete. Clinics in Sports Medicine . 2017;36:655–669. doi: 10.1016/j.csm.2017.06.001.. [DOI] [PubMed] [Google Scholar]
  • [26].Petersen LM, Bautista EJ, Nguyen H, Hanson BM, Chen L, Lek SH, et al. Community characteristics of the gut microbiomes of competitive cyclists. Microbiome . 2017;5:98. doi: 10.1186/s40168-017-0320-4.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Valentino TR, Vechetti IJ, Jr, Mobley CB, Dungan CM, Golden L, Goh J, et al. Dysbiosis of the gut microbiome impairs mouse skeletal muscle adaptation to exercise. The Journal of Physiology . 2021;599:4845–4863. doi: 10.1113/JP281788.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Lahiri S, Kim H, Garcia-Perez I, Reza MM, Martin KA, Kundu P, et al. The gut microbiota influences skeletal muscle mass and function in mice. Science Translational Medicine . 2019;11:eaan5662. doi: 10.1126/scitranslmed.aan5662.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Joyner MJ, Hunter SK, Lucia A, Jones AM. Physiology and fast marathons. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2019;128:1065–1068. doi: 10.1152/japplphysiol.00793.2019.. [DOI] [PubMed] [Google Scholar]
  • [30].Hunter SK, Joyner MJ, Jones AM. The two-hour marathon: What’s the equivalent for women? Journal of Applied Physiology (Bethesda, Md.: 1985) . 2014;118:1321–1323. doi: 10.1152/japplphysiol.00852.2014.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Joyner MJ. Physiological limits to endurance exercise performance: influence of sex. The Journal of Physiology . 2017;595:2949–2954. doi: 10.1113/JP272268.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].de Simone G, Devereux RB, Daniels SR, Meyer RA. Gender differences in left ventricular growth. Hypertension (Dallas, Tex.: 1979) . 1995;26:979–983. doi: 10.1161/01.hyp.26.6.979.. [DOI] [PubMed] [Google Scholar]
  • [33].Ogawa T, Spina RJ, Martin WH, 3rd, Kohrt WM, Schechtman KB, Holloszy JO, et al. Effects of aging, sex, and physical training on cardiovascular responses to exercise. Circulation . 1992;86:494–503. doi: 10.1161/01.cir.86.2.494.. [DOI] [PubMed] [Google Scholar]
  • [34].ASTRAND PO, CUDDY TE, SALTIN B, STENBERG J. CARDIAC OUTPUT DURING SUBMAXIMAL AND MAXIMAL WORK. Journal of Applied Physiology . 1964;19:268–274. doi: 10.1152/jappl.1964.19.2.268.. [DOI] [PubMed] [Google Scholar]
  • [35].Yilmaz DC, Buyukakilli B, Gurgul S, Rencuzogullari I. Adaptation of heart to training: a comparative study using echocardiography & impedance cardiography in male & female athletes. The Indian Journal of Medical Research . 2013;137:1111–1120. [PMC free article] [PubMed] [Google Scholar]
  • [36].Murphy WG. The sex difference in haemoglobin levels in adults - mechanisms, causes, and consequences. Blood Reviews . 2014;28:41–47. doi: 10.1016/j.blre.2013.12.003.. [DOI] [PubMed] [Google Scholar]
  • [37].Porter MM, Stuart S, Boij M, Lexell J. Capillary supply of the tibialis anterior muscle in young, healthy, and moderately active men and women. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2001;92:1451–1457. doi: 10.1152/japplphysiol.00744.2001.. [DOI] [PubMed] [Google Scholar]
  • [38].Hunter SK. The Relevance of Sex Differences in Performance Fatigability. Medicine and Science in Sports and Exercise . 2016;48:2247–2256. doi: 10.1249/MSS.0000000000000928.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Krause WC, Rodriguez R, Gegenhuber B, Matharu N, Rodriguez AN, Padilla-Roger AM, et al. Oestrogen engages brain MC4R signalling to drive physical activity in female mice. Nature . 2021;599:131–135. doi: 10.1038/s41586-021-04010-3.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Kannel WB, Hjortland MC, McNamara PM, Gordon T. Menopause and risk of cardiovascular disease: the Framingham study. Annals of Internal Medicine . 1976;85:447–452. doi: 10.7326/0003-4819-85-4-447.. [DOI] [PubMed] [Google Scholar]
  • [41].Jaggar M, Rea K, Spichak S, Dinan TG, Cryan JF. You’ve got male: Sex and the microbiota-gut-brain axis across the lifespan. Frontiers in Neuroendocrinology . 2020;56:100815. doi: 10.1016/j.yfrne.2019.100815.. [DOI] [PubMed] [Google Scholar]
  • [42].Valeri F, Endres K. How biological sex of the host shapes its gut microbiota. Frontiers in Neuroendocrinology . 2021;61:100912. doi: 10.1016/j.yfrne.2021.100912.. [DOI] [PubMed] [Google Scholar]
  • [43].Insenser M, Murri M, Del Campo R, Martínez-García MÁ, Fernández-Durán E, Escobar-Morreale HF. Gut Microbiota and the Polycystic Ovary Syndrome: Influence of Sex, Sex Hormones, and Obesity. The Journal of Clinical Endocrinology and Metabolism . 2018;103:2552–2562. doi: 10.1210/jc.2017-02799.. [DOI] [PubMed] [Google Scholar]
  • [44].Oki K, Toyama M, Banno T, Chonan O, Benno Y, Watanabe K. Comprehensive analysis of the fecal microbiota of healthy Japanese adults reveals a new bacterial lineage associated with a phenotype characterized by a high frequency of bowel movements and a lean body type. BMC Microbiology . 2016;16:284. doi: 10.1186/s12866-016-0898-x.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Koleva PT, Tun HM, Konya T, Guttman DS, Becker AB, Mandhane PJ, et al. Sex-specific impact of asthma during pregnancy on infant gut microbiota. The European Respiratory Journal . 2017;50:1700280. doi: 10.1183/13993003.00280-2017.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Aatsinki AK, Lahti L, Uusitupa HM, Munukka E, Keskitalo A, Nolvi S, et al. Gut microbiota composition is associated with temperament traits in infants. Brain, Behavior, and Immunity . 2019;80:849–858. doi: 10.1016/j.bbi.2019.05.035.. [DOI] [PubMed] [Google Scholar]
  • [47].Adlercreutz H, Järvenpää P. Assay of estrogens in human feces. Journal of Steroid Biochemistry . 1982;17:639–645. doi: 10.1016/0022-4731(82)90565-9.. [DOI] [PubMed] [Google Scholar]
  • [48].Mousavi SH, Mehrara S, Barzegari A, Ostadrahimi A. Correlation of Gut Microbiota Profile with Body Mass Index Among School Age Children. Iranian Red Crescent Medical Journal (IRCMJ) . 2024;20:e58049. [Google Scholar]
  • [49].Genazzani AR, Pintor C, Corda R. Plasma levels of gonadotropins, prolactin, thyroxine, and adrenal and gonadal steroids in obese prepubertal girls. The Journal of Clinical Endocrinology and Metabolism . 1978;47:974–979. doi: 10.1210/jcem-47-5-974.. [DOI] [PubMed] [Google Scholar]
  • [50].Markle JGM, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, et al. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science (New York, N.Y.) . 2013;339:1084–1088. doi: 10.1126/science.1233521.. [DOI] [PubMed] [Google Scholar]
  • [51].Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature . 2012;486:222–227. doi: 10.1038/nature11053.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Org E, Mehrabian M, Parks BW, Shipkova P, Liu X, Drake TA, et al. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes . 2016;7:313–322. doi: 10.1080/19490976.2016.1203502.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Yuan X, Chen R, Zhang Y, Lin X, Yang X. Gut microbiota: effect of pubertal status. BMC Microbiology . 2020;20:334. doi: 10.1186/s12866-020-02021-0.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, et al. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome . 2018;6:205. doi: 10.1186/s40168-018-0587-0.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas . 2017;103:45–53. doi: 10.1016/j.maturitas.2017.06.025.. [DOI] [PubMed] [Google Scholar]
  • [56].Koren O, Goodrich JK, Cullender TC, Spor A, Laitinen K, Bäckhed HK, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell . 2012;150:470–480. doi: 10.1016/j.cell.2012.07.008.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, et al. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. Journal of Translational Medicine . 2012;10:253. doi: 10.1186/1479-5876-10-253.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Mahnic A, Rupnik M. Different host factors are associated with patterns in bacterial and fungal gut microbiota in Slovenian healthy cohort. PloS One . 2018;13:e0209209. doi: 10.1371/journal.pone.0209209.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Santos-Marcos JA, Rangel-Zuñiga OA, Jimenez-Lucena R, Quintana-Navarro GM, Garcia-Carpintero S, Malagon MM, et al. Influence of gender and menopausal status on gut microbiota. Maturitas . 2018;116:43–53. doi: 10.1016/j.maturitas.2018.07.008.. [DOI] [PubMed] [Google Scholar]
  • [60].Lee SM, Kim N, Park JH, Nam RH, Yoon K, Lee DH. Comparative Analysis of Ileal and Cecal Microbiota in Aged Rats. Journal of Cancer Prevention . 2018;23:70–76. doi: 10.15430/JCP.2018.23.2.70.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Elgaddal N, Kramarow EA, Reuben C. Physical Activity Among Adults Aged 18 and Over: United States, 2020. NCHS Data Brief . 2022:1–8. doi: 10.15620/cdc:120213.. [DOI] [PubMed] [Google Scholar]
  • [62].Deschasaux M, Bouter KE, Prodan A, Levin E, Groen AK, Herrema H, et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nature Medicine . 2018;24:1526–1531. doi: 10.1038/s41591-018-0160-1.. [DOI] [PubMed] [Google Scholar]
  • [63].Ross MC, Muzny DM, McCormick JB, Gibbs RA, Fisher-Hoch SP, Petrosino JF. 16S gut community of the Cameron County Hispanic Cohort. Microbiome . 2015;3 doi: 10.1186/s40168-015-0072-y.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Neff CP, Krueger O, Xiong K, Arif S, Nusbacher N, Schneider JM, et al. Fecal Microbiota Composition Drives Immune Activation in HIV-infected Individuals. EBioMedicine . 2018;30:192–202. doi: 10.1016/j.ebiom.2018.03.024.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Sankaranarayanan K, Ozga AT, Warinner C, Tito RY, Obregon-Tito AJ, Xu J, et al. Gut Microbiome Diversity among Cheyenne and Arapaho Individuals from Western Oklahoma. Current Biology: CB . 2015;25:3161–3169. doi: 10.1016/j.cub.2015.10.060.. CB. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].He Y, Wu W, Wu S, Zheng HM, Li P, Sheng HF, et al. Linking gut microbiota, metabolic syndrome and economic status based on a population-level analysis. Microbiome . 2018;6:172. doi: 10.1186/s40168-018-0557-6.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Miller GE, Engen PA, Gillevet PM, Shaikh M, Sikaroodi M, Forsyth CB, et al. Lower Neighborhood Socioeconomic Status Associated with Reduced Diversity of the Colonic Microbiota in Healthy Adults. PloS One . 2016;11:e0148952. doi: 10.1371/journal.pone.0148952.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Bowyer RCE, Jackson MA, Le Roy CI, Ni Lochlainn M, Spector TD, Dowd JB, et al. Socioeconomic Status and the Gut Microbiome: A TwinsUK Cohort Study. Microorganisms . 2019;7:17. doi: 10.3390/microorganisms7010017.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Amaruddin AI, Hamid F, Koopman JPR, Muhammad M, Brienen EA, van Lieshout L, et al. The Bacterial Gut Microbiota of Schoolchildren from High and Low Socioeconomic Status: A Study in an Urban Area of Makassar, Indonesia. Microorganisms . 2020;8:961. doi: 10.3390/microorganisms8060961.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Chong CW, Ahmad AF, Lim YAL, Teh CSJ, Yap IKS, Lee SC, et al. Effect of ethnicity and socioeconomic variation to the gut microbiota composition among pre-adolescent in Malaysia. Scientific Reports . 2015;5:13338. doi: 10.1038/srep13338.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Levin AM, Sitarik AR, Havstad SL, Fujimura KE, Wegienka G, Cassidy-Bushrow AE, et al. Joint effects of pregnancy, sociocultural, and environmental factors on early life gut microbiome structure and diversity. Scientific Reports . 2016;6:31775. doi: 10.1038/srep31775.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Galley JD, Bailey M, Kamp Dush C, Schoppe-Sullivan S, Christian LM. Maternal obesity is associated with alterations in the gut microbiome in toddlers. PloS One . 2014;9:e113026. doi: 10.1371/journal.pone.0113026.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Flannery JE, Stagaman K, Burns AR, Hickey RJ, Roos LE, Giuliano RJ, et al. Gut Feelings Begin in Childhood: the Gut Metagenome Correlates with Early Environment, Caregiving, and Behavior. mBio . 2020;11:2780. doi: 10.1128/mBio.02780-19.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Brooks AW, Priya S, Blekhman R, Bordenstein SR. Gut microbiota diversity across ethnicities in the United States. PLoS Biology . 2018;16:e2006842. doi: 10.1371/journal.pbio.2006842.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Stearns JC, Zulyniak MA, de Souza RJ, Campbell NC, Fontes M, Shaikh M, et al. Ethnic and diet-related differences in the healthy infant microbiome. Genome Medicine . 2017;9:32. doi: 10.1186/s13073-017-0421-5.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Sordillo JE, Zhou Y, McGeachie MJ, Ziniti J, Lange N, Laranjo N, et al. Factors influencing the infant gut microbiome at age 3-6 months: Findings from the ethnically diverse Vitamin D Antenatal Asthma Reduction Trial (VDAART) The Journal of Allergy and Clinical Immunology . 2017;139:482–491.e14. doi: 10.1016/j.jaci.2016.08.045.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Thompson PD, Franklin BA, Balady GJ, Blair SN, Corrado D, Estes NAM, 3rd, et al. Exercise and acute cardiovascular events placing the risks into perspective: a scientific statement from the American Heart Association Council on Nutrition, Physical Activity, and Metabolism and the Council on Clinical Cardiology. Circulation . 2007;115:2358–2368. doi: 10.1161/CIRCULATIONAHA.107.181485.. [DOI] [PubMed] [Google Scholar]
  • [78].Ekblom O, Ek A, Cider Å, Hambraeus K, Börjesson M. Increased Physical Activity Post-Myocardial Infarction Is Related to Reduced Mortality: Results From the SWEDEHEART Registry. Journal of the American Heart Association . 2018;7:e010108. doi: 10.1161/JAHA.118.010108.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Tan Y, Feng P, Feng L, Shi L, Song Y, Yang J, et al. Low-dose exercise protects the heart against established myocardial infarction via IGF-1-upregulated CTRP9 in male mice. MedComm . 2023;4:e411. doi: 10.1002/mco2.411.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Tian D, Meng J. Exercise for Prevention and Relief of Cardiovascular Disease: Prognoses, Mechanisms, and Approaches. Oxidative Medicine and Cellular Longevity . 2019;2019:3756750. doi: 10.1155/2019/3756750.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Guo S, Huang Y, Zhang Y, Huang H, Hong S, Liu T. Impacts of exercise interventions on different diseases and organ functions in mice. Journal of Sport and Health Science . 2020;9:53–73. doi: 10.1016/j.jshs.2019.07.004.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Myers J, Kokkinos P, Nyelin E. Physical Activity, Cardiorespiratory Fitness, and the Metabolic Syndrome. Nutrients . 2019;11:1652. doi: 10.3390/nu11071652.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Tucker WJ, Fegers-Wustrow I, Halle M, Haykowsky MJ, Chung EH, Kovacic JC. Exercise for Primary and Secondary Prevention of Cardiovascular Disease: JACC Focus Seminar 1/4. Journal of the American College of Cardiology . 2022;80:1091–1106. doi: 10.1016/j.jacc.2022.07.004.. [DOI] [PubMed] [Google Scholar]
  • [84].Kondamudi N, Thangada N, Patel KV, Ayers C, Chandra A, Berry JD, et al. Regional adiposity, cardiorespiratory fitness, and left ventricular strain: an analysis from the Dallas Heart Study. Journal of Cardiovascular Magnetic Resonance: Official Journal of the Society for Cardiovascular Magnetic Resonance . 2021;23:78. doi: 10.1186/s12968-021-00757-w.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Campos JC, Gomes KMS, Ferreira JCB. Impact of exercise training on redox signaling in cardiovascular diseases. Food and Chemical Toxicology: an International Journal Published for the British Industrial Biological Research Association . 2013;62:107–119. doi: 10.1016/j.fct.2013.08.035.. [DOI] [PubMed] [Google Scholar]
  • [86].Rubanyi GM. Endothelium-derived relaxing and contracting factors. Journal of Cellular Biochemistry . 1991;46:27–36. doi: 10.1002/jcb.240460106.. [DOI] [PubMed] [Google Scholar]
  • [87].Tamang HK, Timilsina U, Singh KP, Shrestha S, Raman RK, Panta P, et al. Apo B/Apo A-I Ratio is Statistically A Better Predictor of Cardiovascular Disease (CVD) than Conventional Lipid Profile: A Study from Kathmandu Valley, Nepal. Journal of Clinical and Diagnostic Research: JCDR . 2014;8:34–36. doi: 10.7860/JCDR/2014/7588.4000.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].DuPont JJ, Greaney JL, Wenner MM, Lennon-Edwards SL, Sanders PW, Farquhar WB, et al. High dietary sodium intake impairs endothelium-dependent dilation in healthy salt-resistant humans. Journal of Hypertension . 2013;31:530–536. doi: 10.1097/HJH.0b013e32835c6ca8.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circulation Research . 2000;87:840–844. doi: 10.1161/01.res.87.10.840.. [DOI] [PubMed] [Google Scholar]
  • [90].Guers JJ, Farquhar WB, Edwards DG, Lennon SL. Voluntary Wheel Running Attenuates Salt-Induced Vascular Stiffness Independent of Blood Pressure. American Journal of Hypertension . 2019;32:1162–1169. doi: 10.1093/ajh/hpz128.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Lesniewski LA, Zigler ML, Durrant JR, Nowlan MJ, Folian BJ, Donato AJ, et al. Aging compounds western diet-associated large artery endothelial dysfunction in mice: prevention by voluntary aerobic exercise. Experimental Gerontology . 2013;48:1218–1225. doi: 10.1016/j.exger.2013.08.001.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Roque FR, Briones AM, García-Redondo AB, Galán M, Martínez-Revelles S, Avendaño MS, et al. Aerobic exercise reduces oxidative stress and improves vascular changes of small mesenteric and coronary arteries in hypertension. British Journal of Pharmacology . 2013;168:686–703. doi: 10.1111/j.1476-5381.2012.02224.x.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Dinh QN, Drummond GR, Sobey CG, Chrissobolis S. Roles of inflammation, oxidative stress, and vascular dysfunction in hypertension. BioMed Research International . 2014;2014:406960. doi: 10.1155/2014/406960.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Birukov KG. Cyclic stretch, reactive oxygen species, and vascular remodeling. Antioxidants & Redox Signaling . 2009;11:1651–1667. doi: 10.1089/ars.2008.2390.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Guzik TJ, Sadowski J, Guzik B, Jopek A, Kapelak B, Przybylowski P, et al. Coronary artery superoxide production and nox isoform expression in human coronary artery disease. Arteriosclerosis, Thrombosis, and Vascular Biology . 2006;26:333–339. doi: 10.1161/01.ATV.0000196651.64776.51.. [DOI] [PubMed] [Google Scholar]
  • [96].Bowles DK, Farrar RP, Starnes JW. Exercise training improves cardiac function after ischemia in the isolated, working rat heart. The American Journal of Physiology . 1992;263:H804–H809. doi: 10.1152/ajpheart.1992.263.3.H804.. [DOI] [PubMed] [Google Scholar]
  • [97].Weeks KL, McMullen JR. The athlete’s heart vs. the failing heart: can signaling explain the two distinct outcomes? Physiology (Bethesda, Md.) . 2011;26:97–105. doi: 10.1152/physiol.00043.2010.. [DOI] [PubMed] [Google Scholar]
  • [98].Norheim F, Langleite TM, Hjorth M, Holen T, Kielland A, Stadheim HK, et al. The effects of acute and chronic exercise on PGC-1α, irisin and browning of subcutaneous adipose tissue in humans. The FEBS Journal . 2014;281:739–749. doi: 10.1111/febs.12619.. [DOI] [PubMed] [Google Scholar]
  • [99].Lira VA, Benton CR, Yan Z, Bonen A. PGC-1alpha regulation by exercise training and its influences on muscle function and insulin sensitivity. American Journal of Physiology. Endocrinology and Metabolism . 2010;299:E145–E161. doi: 10.1152/ajpendo.00755.2009.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Handschin C, Spiegelman BM. The role of exercise and PGC1alpha in inflammation and chronic disease. Nature . 2008;454:463–469. doi: 10.1038/nature07206.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Oppert JM, Ciangura C, Bellicha A. Physical activity and exercise for weight loss and maintenance in people living with obesity. Reviews in Endocrine & Metabolic Disorders . 2023;24:937–949. doi: 10.1007/s11154-023-09805-5.. [DOI] [PubMed] [Google Scholar]
  • [102].Goto C, Nishioka K, Umemura T, Jitsuiki D, Sakagutchi A, Kawamura M, et al. Acute moderate-intensity exercise induces vasodilation through an increase in nitric oxide bioavailiability in humans. American Journal of Hypertension . 2007;20:825–830. doi: 10.1016/j.amjhyper.2007.02.014.. [DOI] [PubMed] [Google Scholar]
  • [103].Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, et al. Voluntary wheel running restores endothelial function in conduit arteries of old mice: direct evidence for reduced oxidative stress, increased superoxide dismutase activity and down-regulation of NADPH oxidase. The Journal of Physiology . 2009;587:3271–3285. doi: 10.1113/jphysiol.2009.169771.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Moncada S, Higgs A. The L-arginine-nitric oxide pathway. The New England Journal of Medicine . 1993;329:2002–2012. doi: 10.1056/NEJM199312303292706.. [DOI] [PubMed] [Google Scholar]
  • [105].Heffernan KS, Fahs CA, Ranadive SM, Patvardhan EA. L-arginine as a nutritional prophylaxis against vascular endothelial dysfunction with aging. Journal of Cardiovascular Pharmacology and Therapeutics . 2010;15:17–23. doi: 10.1177/1074248409354599.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Tao X, Chen Y, Zhen K, Ren S, Lv Y, Yu L. Effect of continuous aerobic exercise on endothelial function: A systematic review and meta-analysis of randomized controlled trials. Frontiers in Physiology . 2023;14:1043108. doi: 10.3389/fphys.2023.1043108.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Sun D, Huang A, Koller A, Kaley G. Short-term daily exercise activity enhances endothelial NO synthesis in skeletal muscle arterioles of rats. Journal of Applied Physiology (Bethesda, Md . 1994;76:2241–2247. doi: 10.1152/jappl.1994.76.5.2241.. [DOI] [PubMed] [Google Scholar]
  • [108].Paula SM, Fernandes T, Couto GK, Jordão MT, Oliveira EM, Michelini LC, et al. Molecular Pathways Involved in Aerobic Exercise Training Enhance Vascular Relaxation. Medicine and Science in Sports and Exercise . 2020;52:2117–2126. doi: 10.1249/MSS.0000000000002355.. [DOI] [PubMed] [Google Scholar]
  • [109].Guers JJ, Kasecky-Lardner L, Farquhar WB, Edwards DG, Lennon SL. Voluntary wheel running prevents salt-induced endothelial dysfunction: role of oxidative stress. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2018;126:502–510. doi: 10.1152/japplphysiol.00421.2018.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Ying WZ, Sanders PW. Dietary salt increases endothelial nitric oxide synthase and TGF-beta1 in rat aortic endothelium. The American Journal of Physiology . 1999;277:H1293–H1298. doi: 10.1152/ajpheart.1999.277.4.H1293.. [DOI] [PubMed] [Google Scholar]
  • [111].Heitzer T, Krohn K, Albers S, Meinertz T. Tetrahydrobiopterin improves endothelium-dependent vasodilation by increasing nitric oxide activity in patients with Type II diabetes mellitus. Diabetologia . 2000;43:1435–1438. doi: 10.1007/s001250051551.. [DOI] [PubMed] [Google Scholar]
  • [112].Marino F, Scalise M, Cianflone E, Salerno L, Cappetta D, Salerno N, et al. Physical Exercise and Cardiac Repair: The Potential Role of Nitric Oxide in Boosting Stem Cell Regenerative Biology. Antioxidants (Basel, Switzerland) . 2021;10:1002. doi: 10.3390/antiox10071002.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Kuczmarski JM, Martens CR, Kim J, Lennon-Edwards SL, Edwards DG. Cardiac function is preserved following 4 weeks of voluntary wheel running in a rodent model of chronic kidney disease. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2014;117:482–491. doi: 10.1152/japplphysiol.00344.2014.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Sessa WC, Pritchard K, Seyedi N, Wang J, Hintze TH. Chronic exercise in dogs increases coronary vascular nitric oxide production and endothelial cell nitric oxide synthase gene expression. Circulation Research . 1994;74:349–353. doi: 10.1161/01.res.74.2.349.. [DOI] [PubMed] [Google Scholar]
  • [115].Tsukiyama Y, Ito T, Nagaoka K, Eguchi E, Ogino K. Effects of exercise training on nitric oxide, blood pressure and antioxidant enzymes. Journal of Clinical Biochemistry and Nutrition . 2017;60:180–186. doi: 10.3164/jcbn.16-108.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Shannon OM, Clifford T, Seals DR, Craighead DH, Rossman MJ. Nitric oxide, aging and aerobic exercise: Sedentary individuals to Master’s athletes. Nitric Oxide: Biology and Chemistry . 2022;125:31–39. doi: 10.1016/j.niox.2022.06.002.. [DOI] [PubMed] [Google Scholar]
  • [117].Tanaka H, Dinenno FA, Monahan KD, Clevenger CM, DeSouza CA, Seals DR. Aging, habitual exercise, and dynamic arterial compliance. Circulation . 2000;102:1270–1275. doi: 10.1161/01.cir.102.11.1270.. [DOI] [PubMed] [Google Scholar]
  • [118].Lewis TV, Dart AM, Chin-Dusting JP, Kingwell BA. Exercise training increases basal nitric oxide production from the forearm in hypercholesterolemic patients. Arteriosclerosis, Thrombosis, and Vascular Biology . 1999;19:2782–2787. doi: 10.1161/01.atv.19.11.2782.. [DOI] [PubMed] [Google Scholar]
  • [119].Aker A, Saliba W, Bahouth F, Naoum I, Zafrir B. Cardiorespiratory Fitness and Risk of Cardiovascular Events and Mortality in Middle Age Patients without Known Cardiovascular Disease. Journal of Clinical Medicine . 2023;12:7011. doi: 10.3390/jcm12227011.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Drexler H, Lu W. Endothelial dysfunction of hindquarter resistance vessels in experimental heart failure. The American Journal of Physiology . 1992;262:H1640–H1645. doi: 10.1152/ajpheart.1992.262.6.H1640.. [DOI] [PubMed] [Google Scholar]
  • [121].Piña IL, Apstein CS, Balady GJ, Belardinelli R, Chaitman BR, Duscha BD, et al. Exercise and heart failure: A statement from the American Heart Association Committee on exercise, rehabilitation, and prevention. Circulation . 2003;107:1210–1225. doi: 10.1161/01.cir.0000055013.92097.40.. [DOI] [PubMed] [Google Scholar]
  • [122].Hirooka Y, Imaizumi T, Tagawa T, Shiramoto M, Endo T, Ando S, et al. Effects of L-arginine on impaired acetylcholine-induced and ischemic vasodilation of the forearm in patients with heart failure. Circulation . 1994;90:658–668. doi: 10.1161/01.cir.90.2.658.. [DOI] [PubMed] [Google Scholar]
  • [123].Higashi Y, Noma K, Yoshizumi M, Kihara Y. Endothelial function and oxidative stress in cardiovascular diseases. Circulation Journal: Official Journal of the Japanese Circulation Society . 2009;73:411–418. doi: 10.1253/circj.cj-08-1102.. [DOI] [PubMed] [Google Scholar]
  • [124].Bachschmid MM, Schildknecht S, Matsui R, Zee R, Haeussler D, Cohen RA, et al. Vascular aging: chronic oxidative stress and impairment of redox signaling-consequences for vascular homeostasis and disease. Annals of Medicine . 2013;45:17–36. doi: 10.3109/07853890.2011.645498.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Li H, Horke S, Förstermann U. Vascular oxidative stress, nitric oxide and atherosclerosis. Atherosclerosis . 2014;237:208–219. doi: 10.1016/j.atherosclerosis.2014.09.001.. [DOI] [PubMed] [Google Scholar]
  • [126].Kalinina N, Agrotis A, Tararak E, Antropova Y, Kanellakis P, Ilyinskaya O, et al. Cytochrome b558-dependent NAD(P)H oxidase-phox units in smooth muscle and macrophages of atherosclerotic lesions. Arteriosclerosis, Thrombosis, and Vascular Biology . 2002;22:2037–2043. doi: 10.1161/01.atv.0000040222.02255.0f.. [DOI] [PubMed] [Google Scholar]
  • [127].Dikalov SI, Dikalova AE. Contribution of mitochondrial oxidative stress to hypertension. Current Opinion in Nephrology and Hypertension . 2016;25:73–80. doi: 10.1097/MNH.0000000000000198.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Hill MF, Singal PK. Right and left myocardial antioxidant responses during heart failure subsequent to myocardial infarction. Circulation . 1997;96:2414–2420. doi: 10.1161/01.cir.96.7.2414.. [DOI] [PubMed] [Google Scholar]
  • [129].Belch JJ, Bridges AB, Scott N, Chopra M. Oxygen free radicals and congestive heart failure. British Heart Journal . 1991;65:245–248. doi: 10.1136/hrt.65.5.245.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Vaitkevicius PV, Fleg JL, Engel JH, O’Connor FC, Wright JG, Lakatta LE, et al. Effects of age and aerobic capacity on arterial stiffness in healthy adults. Circulation . 1993;88:1456–1462. doi: 10.1161/01.cir.88.4.1456.. [DOI] [PubMed] [Google Scholar]
  • [131].Fleenor BS, Seals DR, Zigler ML, Sindler AL. Superoxide-lowering therapy with TEMPOL reverses arterial dysfunction with aging in mice. Aging Cell . 2012;11:269–276. doi: 10.1111/j.1474-9726.2011.00783.x.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Gryglewski RJ, Moncada S, Palmer RM. Bioassay of prostacyclin and endothelium-derived relaxing factor (EDRF) from porcine aortic endothelial cells. British Journal of Pharmacology . 1986;87:685–694. doi: 10.1111/j.1476-5381.1986.tb14586.x.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Zhu J, Huang T, Lombard JH. Effect of high-salt diet on vascular relaxation and oxidative stress in mesenteric resistance arteries. Journal of Vascular Research . 2007;44:382–390. doi: 10.1159/000102955.. [DOI] [PubMed] [Google Scholar]
  • [134].Ravi Y, Selvendiran K, Naidu SK, Meduru S, Citro LA, Bognár B, et al. Pulmonary hypertension secondary to left-heart failure involves peroxynitrite-induced downregulation of PTEN in the lung. Hypertension (Dallas, Tex.: 1979) . 2013;61:593–601. doi: 10.1161/HYPERTENSIONAHA.111.00514.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Didion SP, Ryan MJ, Didion LA, Fegan PE, Sigmund CD, Faraci FM. Increased superoxide and vascular dysfunction in CuZnSOD-deficient mice. Circulation Research . 2002;91:938–944. doi: 10.1161/01.res.0000043280.65241.04.. [DOI] [PubMed] [Google Scholar]
  • [136].Fukai T, Ushio-Fukai M. Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxidants & Redox Signaling . 2011;15:1583–1606. doi: 10.1089/ars.2011.3999.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiological Reviews . 2007;87:315–424. doi: 10.1152/physrev.00029.2006.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Bajic VP, Van Neste C, Obradovic M, Zafirovic S, Radak D, Bajic VB, et al. Glutathione ”Redox Homeostasis” and Its Relation to Cardiovascular Disease. Oxidative Medicine and Cellular Longevity . 2019;2019:5028181. doi: 10.1155/2019/5028181.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Forgione MA, Weiss N, Heydrick S, Cap A, Klings ES, Bierl C, et al. Cellular glutathione peroxidase deficiency and endothelial dysfunction. American Journal of Physiology. Heart and Circulatory Physiology . 2002;282:H1255–H1261. doi: 10.1152/ajpheart.00598.2001.. [DOI] [PubMed] [Google Scholar]
  • [140].Nandi A, Yan LJ, Jana CK, Das N. Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases. Oxidative Medicine and Cellular Longevity . 2019;2019:9613090. doi: 10.1155/2019/9613090.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Powers SK, Radak Z, Ji LL, Jackson M. Reactive oxygen species promote endurance exercise-induced adaptations in skeletal muscles. Journal of Sport and Health Science . 2024;13:780–792. doi: 10.1016/j.jshs.2024.05.001.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Blacher J, Asmar R, Djane S, London GM, Safar ME. Aortic pulse wave velocity as a marker of cardiovascular risk in hypertensive patients. Hypertension (Dallas, Tex.: 1979) . 1999;33:1111–1117. doi: 10.1161/01.hyp.33.5.1111.. [DOI] [PubMed] [Google Scholar]
  • [143].Benetos A, Safar M, Rudnichi A, Smulyan H, Richard JL, Ducimetieère P, et al. Pulse pressure: a predictor of long-term cardiovascular mortality in a French male population. Hypertension (Dallas, Tex.: 1979) . 1997;30:1410–1415. doi: 10.1161/01.hyp.30.6.1410.. [DOI] [PubMed] [Google Scholar]
  • [144].Lyle AN, Raaz U. Killing Me Unsoftly: Causes and Mechanisms of Arterial Stiffness. Arteriosclerosis, Thrombosis, and Vascular Biology . 2017;37:e1–e11. doi: 10.1161/ATVBAHA.116.308563.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Safar ME, London GM, Plante GE. Arterial stiffness and kidney function. Hypertension (Dallas, Tex.: 1979) . 2004;43:163–168. doi: 10.1161/01.HYP.0000114571.75762.b0.. [DOI] [PubMed] [Google Scholar]
  • [146].Fleenor BS, Marshall KD, Durrant JR, Lesniewski LA, Seals DR. Arterial stiffening with ageing is associated with transforming growth factor-β1-related changes in adventitial collagen: reversal by aerobic exercise. The Journal of Physiology . 2010;588:3971–3982. doi: 10.1113/jphysiol.2010.194753.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Suwaidi JA, Hamasaki S, Higano ST, Nishimura RA, Holmes DR, Jr, Lerman A. Long-term follow-up of patients with mild coronary artery disease and endothelial dysfunction. Circulation . 2000;101:948–954. doi: 10.1161/01.cir.101.9.948.. [DOI] [PubMed] [Google Scholar]
  • [148].Saz-Lara A, Cavero-Redondo I, Álvarez-Bueno C, Notario-Pacheco B, Ruiz-Grao MC, Martínez-Vizcaíno V. The Acute Effect of Exercise on Arterial Stiffness in Healthy Subjects: A Meta-Analysis. Journal of Clinical Medicine . 2021;10:291. doi: 10.3390/jcm10020291.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Shi N, Xia J, Wang C, Zhou J, Huang J, Hu M, et al. Aerobic Exercise Prevents Arterial Stiffness and Attenuates Hyperexcitation of Sympathetic Nerves in Perivascular Adipose Tissue of Mice after Transverse Aortic Constriction. International Journal of Molecular Sciences . 2022;23:11189. doi: 10.3390/ijms231911189.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Guzik TJ, Touyz RM. Oxidative Stress, Inflammation, and Vascular Aging in Hypertension. Hypertension (Dallas, Tex.: 1979) . 2017;70:660–667. doi: 10.1161/HYPERTENSIONAHA.117.07802.. [DOI] [PubMed] [Google Scholar]
  • [151].Rossman MJ, Santos-Parker JR, Steward CAC, Bispham NZ, Cuevas LM, Rosenberg HL, et al. Chronic Supplementation With a Mitochondrial Antioxidant (MitoQ) Improves Vascular Function in Healthy Older Adults. Hypertension (Dallas, Tex.: 1979) . 2018;71:1056–1063. doi: 10.1161/HYPERTENSIONAHA.117.10787.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Ismaeel A, Brumberg RS, Kirk JS, Papoutsi E, Farmer PJ, Bohannon WT, et al. Oxidative Stress and Arterial Dysfunction in Peripheral Artery Disease. Antioxidants (Basel, Switzerland) . 2018;7:145. doi: 10.3390/antiox7100145.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Gioscia-Ryan RA, Battson ML, Cuevas LM, Eng JS, Murphy MP, Seals DR. Mitochondria-targeted antioxidant therapy with MitoQ ameliorates aortic stiffening in old mice. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2017;124:1194–1202. doi: 10.1152/japplphysiol.00670.2017.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Gioscia-Ryan RA, Clayton ZS, Fleenor BS, Eng JS, Johnson LC, Rossman MJ, et al. Late-life voluntary wheel running reverses age-related aortic stiffness in mice: a translational model for studying mechanisms of exercise-mediated arterial de-stiffening. GeroScience . 2021;43:423–432. doi: 10.1007/s11357-020-00212-1.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Longoria CR, Guers JJ, Campbell SC. The Interplay between Cardiovascular Disease, Exercise, and the Gut Microbiome. Reviews in Cardiovascular Medicine . 2022;23:365. doi: 10.31083/j.rcm2311365.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Nemet I, Li XS, Haghikia A, Li L, Wilcox J, Romano KA, et al. Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality. European Heart Journal . 2023;44:3085–3096. doi: 10.1093/eurheartj/ehad333.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Wang T, Shi Z, Ren H, Xu M, Lu J, Yang F, et al. Divergent age-associated and metabolism-associated gut microbiome signatures modulate cardiovascular disease risk. Nature Medicine . 2024;30:1722–1731. doi: 10.1038/s41591-024-03038-y.. [DOI] [PubMed] [Google Scholar]
  • [158].Liu Y, Ritchie SC, Teo SM, Ruuskanen MO, Kambur O, Zhu Q, et al. Integration of polygenic and gut metagenomic risk prediction for common diseases. Nature Aging . 2024;4:584–594. doi: 10.1038/s43587-024-00590-7.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Wang YC, Koay YC, Pan C, Zhou Z, Tang W, Wilcox J, et al. Indole-3-Propionic Acid Protects Against Heart Failure With Preserved Ejection Fraction. Circulation Research . 2024;134:371–389. doi: 10.1161/CIRCRESAHA.123.322381.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Verhaar BJH, Wijdeveld M, Wortelboer K, Rampanelli E, Levels JHM, Collard D, et al. Effects of Oral Butyrate on Blood Pressure in Patients With Hypertension: A Randomized, Placebo-Controlled Trial. Hypertension (Dallas, Tex.: 1979) . 2024;81:2124–2136. doi: 10.1161/HYPERTENSIONAHA.123.22437.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Molinaro A, Nemet I, Bel Lassen P, Chakaroun R, Nielsen T, Aron-Wisnewsky J, et al. Microbially Produced Imidazole Propionate Is Associated With Heart Failure and Mortality. JACC. Heart Failure . 2023;11:810–821. doi: 10.1016/j.jchf.2023.03.008.. [DOI] [PubMed] [Google Scholar]
  • [162].Okami Y, Arima H, Kondo K, Hexun Z, Yano Y, Kadota A, et al. The gut microbiota and coronary artery calcification in Japanese men. American Heart Journal . 2024;267:12–21. doi: 10.1016/j.ahj.2023.09.009.. [DOI] [PubMed] [Google Scholar]
  • [163].Sayols-Baixeras S, Dekkers KF, Baldanzi G, Jönsson D, Hammar U, Lin YT, et al. Streptococcus Species Abundance in the Gut Is Linked to Subclinical Coronary Atherosclerosis in 8973 Participants From the SCAPIS Cohort. Circulation . 2023;148:459–472. doi: 10.1161/CIRCULATIONAHA.123.063914.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Salvado R, Santos-Minguez S, Lugones-Sánchez C, Gonzalez-Sánchez S, Tamayo-Morales O, Quesada-Rico JA, et al. Gut microbiota and its relationship with early vascular ageing in a Spanish population (MIVAS study) European Journal of Clinical Investigation . 2024;54:e14228. doi: 10.1111/eci.14228.. [DOI] [PubMed] [Google Scholar]
  • [165].Feng Z, Long W, Hao B, Ding D, Ma X, Zhao L, et al. A human stool-derived Bilophila wadsworthia strain caused systemic inflammation in specific-pathogen-free mice. Gut Pathogens . 2017;9:59. doi: 10.1186/s13099-017-0208-7.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Guo J, Jia P, Gu Z, Tang W, Wang A, Sun Y, et al. Altered gut microbiota and metabolite profiles provide clues in understanding resistant hypertension. Journal of Hypertension . 2024;42:1212–1225. doi: 10.1097/HJH.0000000000003716.. [DOI] [PubMed] [Google Scholar]
  • [167].Wang M, Li XS, Wang Z, de Oliveira Otto MC, Lemaitre RN, Fretts A, et al. Trimethylamine N-oxide is associated with long-term mortality risk: the multi-ethnic study of atherosclerosis. European Heart Journal . 2023;44:1608–1618. doi: 10.1093/eurheartj/ehad089.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Zhu Y, Dwidar M, Nemet I, Buffa JA, Sangwan N, Li XS, et al. Two distinct gut microbial pathways contribute to meta-organismal production of phenylacetylglutamine with links to cardiovascular disease. Cell Host & Microbe . 2023;31:18–32.e9. doi: 10.1016/j.chom.2022.11.015.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Ottosson F, Brunkwall L, Smith E, Orho-Melander M, Nilsson PM, Fernandez C, et al. The gut microbiota-related metabolite phenylacetylglutamine associates with increased risk of incident coronary artery disease. Journal of Hypertension . 2020;38:2427–2434. doi: 10.1097/HJH.0000000000002569.. [DOI] [PubMed] [Google Scholar]
  • [170].Poesen R, Claes K, Evenepoel P, de Loor H, Augustijns P, Kuypers D, et al. Microbiota-Derived Phenylacetylglutamine Associates with Overall Mortality and Cardiovascular Disease in Patients with CKD. Journal of the American Society of Nephrology: JASN . 2016;27:3479–3487. doi: 10.1681/ASN.2015121302.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Liu Y, Liu S, Zhao Z, Song X, Qu H, Liu H. Phenylacetylglutamine is associated with the degree of coronary atherosclerotic severity assessed by coronary computed tomographic angiography in patients with suspected coronary artery disease. Atherosclerosis . 2021;333:75–82. doi: 10.1016/j.atherosclerosis.2021.08.029.. [DOI] [PubMed] [Google Scholar]
  • [172].Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, et al. A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors. Cell . 2020;180:862–877.e22. doi: 10.1016/j.cell.2020.02.016.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Saha PP, Gogonea V, Sweet W, Mohan ML, Singh KD, Anderson JT, et al. Gut microbe-generated phenylacetylglutamine is an endogenous allosteric modulator of β2-adrenergic receptors. Nature Communications . 2024;15:6696. doi: 10.1038/s41467-024-50855-3.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Romano KA, Nemet I, Prasad Saha P, Haghikia A, Li XS, Mohan ML, et al. Gut Microbiota-Generated Phenylacetylglutamine and Heart Failure. Circulation. Heart Failure . 2023;16:e009972. doi: 10.1161/CIRCHEARTFAILURE.122.009972.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Tang WHW, Nemet I, Li XS, Wu Y, Haghikia A, Witkowski M, et al. Prognostic value of gut microbe-generated metabolite phenylacetylglutamine in patients with heart failure. European Journal of Heart Failure . 2024;26:233–241. doi: 10.1002/ejhf.3111.. [DOI] [PubMed] [Google Scholar]
  • [176].Theofilis P, Vlachakis PK, Oikonomou E, Tsioufis K, Tousoulis D. Targeting the Gut Microbiome to Treat Cardiometabolic Disease. Current Atherosclerosis Reports . 2024;26:25–34. doi: 10.1007/s11883-023-01183-2.. [DOI] [PubMed] [Google Scholar]
  • [177].Vyas A, Desai R, Went TR, Wiltshire D, Priyadarshni S, Shalaby M, et al. Cardiovascular Disease Burden and Major Adverse Cardiac Events in Young Black Patients: A National Analysis of 2 Cohorts 10 Years Apart (2017 Versus 2007) Journal of the American Heart Association . 2023;12:e029895. doi: 10.1161/JAHA.123.029895.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Post WS, Watson KE, Hansen S, Folsom AR, Szklo M, Shea S, et al. Racial and Ethnic Differences in All-Cause and Cardiovascular Disease Mortality: The MESA Study. Circulation . 2022;146:229–239. doi: 10.1161/CIRCULATIONAHA.122.059174.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Kyalwazi AN, Loccoh EC, Brewer LC, Ofili EO, Xu J, Song Y, et al. Disparities in Cardiovascular Mortality Between Black and White Adults in the United States, 1999 to 2019. Circulation . 2022;146:211–228. doi: 10.1161/CIRCULATIONAHA.122.060199.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Minhas AMK, Talha KM, Abramov D, Johnson HM, Antoine S, Rodriguez F, et al. Racial and ethnic disparities in cardiovascular disease - analysis across major US national databases. Journal of the National Medical Association . 2024;116:258–270. doi: 10.1016/j.jnma.2024.01.022.. [DOI] [PubMed] [Google Scholar]
  • [181].Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, et al. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation . 2024;149:e347–e913. doi: 10.1161/CIR.0000000000001209.. [DOI] [PubMed] [Google Scholar]
  • [182].Garcia M, Almuwaqqat Z, Moazzami K, Young A, Lima BB, Sullivan S, et al. Racial Disparities in Adverse Cardiovascular Outcomes After a Myocardial Infarction in Young or Middle-Aged Patients. Journal of the American Heart Association . 2021;10:e020828. doi: 10.1161/JAHA.121.020828.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Skelton TN, Andrew ME, Arnett DK, Burchfiel CM, Garrison RJ, Samdarshi TE, et al. Echocardiographic left ventricular mass in African-Americans: the Jackson cohort of the Atherosclerosis Risk in Communities Study. Echocardiography (Mount Kisco, N.Y.) . 2003;20:111–120. doi: 10.1046/j.1540-8175.2003.03000.x.. [DOI] [PubMed] [Google Scholar]
  • [184].Fuller-Rowell TE, Curtis DS, Klebanov PK, Brooks-Gunn J, Evans GW. Racial Disparities in Blood Pressure Trajectories of Preterm Children: The Role of Family and Neighborhood Socioeconomic Status. American Journal of Epidemiology . 2017;185:888–897. doi: 10.1093/aje/kww198.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Arnett DK, Rautaharju P, Crow R, Folsom AR, Ekelund LG, Hutchinson R, et al. Black-white differences in electrocardiographic left ventricular mass and its association with blood pressure (the ARIC study). Atherosclerosis Risk in Communities. The American Journal of Cardiology . 1994;74:247–252. doi: 10.1016/0002-9149(94)90365-4.. [DOI] [PubMed] [Google Scholar]
  • [186].Wang J, Chen W, Ruan L, Toprak A, Srinivasan SR, Berenson GS. Differential effect of elevated blood pressure on left ventricular geometry types in black and white young adults in a community (from the Bogalusa Heart Study) The American Journal of Cardiology . 2011;107:717–722. doi: 10.1016/j.amjcard.2010.10.053.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Heffernan KS, Jae SY, Wilund KR, Woods JA, Fernhall B. Racial differences in central blood pressure and vascular function in young men. American Journal of Physiology. Heart and Circulatory Physiology . 2008;295:H2380–H2387. doi: 10.1152/ajpheart.00902.2008.. [DOI] [PubMed] [Google Scholar]
  • [188].Morris AA, Patel RS, Binongo JNG, Poole J, Al Mheid I, Ahmed Y, et al. Racial differences in arterial stiffness and microcirculatory function between Black and White Americans. Journal of the American Heart Association . 2013;2:e002154. doi: 10.1161/JAHA.112.002154.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Brothers RM, Fadel PJ, Keller DM. Racial disparities in cardiovascular disease risk: mechanisms of vascular dysfunction. American Journal of Physiology. Heart and Circulatory Physiology . 2019;317:H777–H789. doi: 10.1152/ajpheart.00126.2019.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Heffernan KS, Lefferts WK, Atallah-Yunes NH, Glasgow AC, Gump BB. Racial Differences in Left Ventricular Mass and Wave Reflection Intensity in Children. Frontiers in Pediatrics . 2020;8:132. doi: 10.3389/fped.2020.00132.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Lefferts WK, Augustine JA, Spartano NL, Atallah-Yunes NH, Heffernan KS, Gump BB. Racial Differences in Aortic Stiffness in Children. The Journal of Pediatrics . 2017;180:62–67. doi: 10.1016/j.jpeds.2016.09.071.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Heffernan KS, Fernhall B. A systematic appraisal of ventricular–aortic load in African American men. Artery Research . 2009;3:65–72. [Google Scholar]
  • [193].DeLoach SS, Daskalakis C, Gidding S, Falkner B. Central blood pressures are associated with left ventricular mass index among African-American adolescents. American Journal of Hypertension . 2012;25:41–45. doi: 10.1038/ajh.2011.174.. [DOI] [PubMed] [Google Scholar]
  • [194].Schutte AE, Kruger R, Gafane-Matemane LF, Breet Y, Strauss-Kruger M, Cruickshank JK. Ethnicity and Arterial Stiffness. Arteriosclerosis, Thrombosis, and Vascular Biology . 2020;40:1044–1054. doi: 10.1161/ATVBAHA.120.313133.. [DOI] [PubMed] [Google Scholar]
  • [195].Patel PD, Velazquez JL, Arora RR. Endothelial dysfunction in African-Americans. International Journal of Cardiology . 2009;132:157–172. doi: 10.1016/j.ijcard.2008.10.007.. [DOI] [PubMed] [Google Scholar]
  • [196].Gibbons GH. Physiology, genetics, and cardiovascular disease: focus on African Americans. Journal of Clinical Hypertension (Greenwich, Conn.) . 2004;6:11–18. doi: 10.1111/j.1524-6175.2004.03562.x.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Vita JA. Nitric oxide and vascular reactivity in African American patients with hypertension. Journal of Cardiac Failure . 2003;9:S199–S204. doi: 10.1054/s1071-9164(03)00588-8.. [DOI] [PubMed] [Google Scholar]
  • [198].Murphy BS, Nam Y, McClelland RL, Acquah I, Cainzos-Achirica M, Nasir K, et al. Addition of Social Determinants of Health to Coronary Heart Disease Risk Prediction: The Multi-Ethnic Study of Atherosclerosis. Journal of the American Heart Association . 2024;13:e033651. doi: 10.1161/JAHA.123.033651.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Javed Z, Haisum Maqsood M, Yahya T, Amin Z, Acquah I, Valero-Elizondo J, et al. Race, Racism, and Cardiovascular Health: Applying a Social Determinants of Health Framework to Racial/Ethnic Disparities in Cardiovascular Disease. Circulation. Cardiovascular Quality and Outcomes . 2022;15:e007917. doi: 10.1161/CIRCOUTCOMES.121.007917.. [DOI] [PubMed] [Google Scholar]
  • [200].Dennis AC, Chung EO, Lodge EK, Martinez RA, Wilbur RE. Looking Back to Leap Forward: A Framework for Operationalizing the Structural Racism Construct in Minority Health Research. Ethnicity & Disease . 2021;31:301–310. doi: 10.18865/ed.31.S1.301.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Needham BL, Dokshina D, Ali T, Allgood KL, Douglas M, Dulin A, et al. Exposure to structural racism-related state laws is associated with worse cardiovascular health among US adults, BRFSS 2011 and 2013. American Journal of Epidemiology . 2024 doi: 10.1093/aje/kwae176.. (online ahead of print) [DOI] [PubMed] [Google Scholar]
  • [202].Lawrence WR, Jones GS, Johnson JA, Ferrell KP, Johnson JN, Shiels MS, et al. Discrimination Experiences and All-Cause and Cardiovascular Mortality: Multi-Ethnic Study of Atherosclerosis. Circulation. Cardiovascular Quality and Outcomes . 2023;16:e009697. doi: 10.1161/CIRCOUTCOMES.122.009697.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Forde AT, Sims M, Muntner P, Lewis T, Onwuka A, Moore K, et al. Discrimination and Hypertension Risk Among African Americans in the Jackson Heart Study. Discrimination and Hypertension Risk Among African Americans in the Jackson Heart Study . 2020;76:715–723. doi: 10.1161/HYPERTENSIONAHA.119.14492.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Lewis TT, Aiello AE, Leurgans S, Kelly J, Barnes LL. Self-reported experiences of everyday discrimination are associated with elevated C-reactive protein levels in older African-American adults. Brain, Behavior, and Immunity . 2010;24:438–443. doi: 10.1016/j.bbi.2009.11.011.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Cruickshank JK, Silva MJ, Molaodi OR, Enayat ZE, Cassidy A, Karamanos A, et al. Ethnic Differences in and Childhood Influences on Early Adult Pulse Wave Velocity: The Determinants of Adolescent, Now Young Adult, Social Wellbeing, and Health Longitudinal Study. Hypertension . 2016;67:1133–1141. doi: 10.1161/HYPERTENSIONAHA.115.07079.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Camelo LV, Machado AV, Chor D, Griep RH, Mill JG, Brant LCC, et al. Racial discrimination is associated with greater arterial stiffness and carotid intima-media thickness: the ELSA-Brasil study. Annals of Epidemiology . 2022;72:40–47. doi: 10.1016/j.annepidem.2022.03.009.. [DOI] [PubMed] [Google Scholar]
  • [207].Mutambudzi M, Boakye K, Green O, Heffernan K. Chronic Work Discrimination, Allostatic Load, and HbA1c in Older Workers. The Gerontologist . 2024;64:gnae094. doi: 10.1093/geront/gnae094.. [DOI] [PubMed] [Google Scholar]
  • [208].Williams A, Nolan TS, Brock G, Garner J, Brewer LC, Sanchez EJ, et al. Association of Socioeconomic Status With Life’s Essential 8 Varies by Race and Ethnicity. Journal of the American Heart Association . 2023;12:e029254. doi: 10.1161/JAHA.122.029254.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Shah NS, Huang X, Petito LC, Bancks MP, Ning H, Cameron NA, et al. Social and Psychosocial Determinants of Racial and Ethnic Differences in Cardiovascular Health in the United States Population. Circulation . 2023;147:190–200. doi: 10.1161/CIRCULATIONAHA.122.061991.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Johnson AE, Herbert BM, Stokes N, Brooks MM, Needham BL, Magnani JW. Educational Attainment, Race, and Ethnicity as Predictors for Ideal Cardiovascular Health: From the National Health and Nutrition Examination Survey. Journal of the American Heart Association . 2022;11:e023438. doi: 10.1161/JAHA.121.023438.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Sims M, Diez-Roux AV, Gebreab SY, Brenner A, Dubbert P, Wyatt S, et al. Perceived discrimination is associated with health behaviours among African-Americans in the Jackson Heart Study. Journal of Epidemiology and Community Health . 2016;70:187–194. doi: 10.1136/jech-2015-206390.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Hines AL, Albert MA, Blair JP, Crews DC, Cooper LA, Long DL, et al. Neighborhood Factors, Individual Stressors, and Cardiovascular Health Among Black and White Adults in the US: The Reasons for Geographic and Racial Differences in Stroke (REGARDS) Study. JAMA Network Open . 2023;6:e2336207. doi: 10.1001/jamanetworkopen.2023.36207.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Islam SJ, Kim JH, Baltrus P, Topel ML, Liu C, Ko YA, et al. Neighborhood characteristics and ideal cardiovascular health among Black adults: results from the Morehouse-Emory Cardiovascular (MECA) Center for Health Equity. Annals of Epidemiology . 2022;65:120.e1–120.e10. doi: 10.1016/j.annepidem.2020.11.009.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Kim K, Joyce BT, Zheng Y, Nannini DR, Wang J, Gordon-Larsen P, et al. Associations of Urban Blue and Green Spaces With Coronary Artery Calcification in Black Individuals and Disadvantaged Neighborhoods. Circulation . 2024;150:203–214. doi: 10.1161/CIRCULATIONAHA.123.067992.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Town M, Eke P, Zhao G, Thomas CW, Hsia J, Pierannunzi C, et al. Racial and Ethnic Differences in Social Determinants of Health and Health-Related Social Needs Among Adults - Behavioral Risk Factor Surveillance System, United States, 2022. MMWR. Morbidity and Mortality Weekly Report . 2024;73:204–208. doi: 10.15585/mmwr.mm7309a3.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Rivier CA, Renedo DB, Sunmonu NA, de Havenon A, Sheth KN, Falcone GJ. Neighborhood Deprivation, Race, Ethnicity, and Undiagnosed Hypertension: Results From the All of Us Research Program. Hypertension (Dallas, Tex.: 1979) . 2024;81:e10–e12. doi: 10.1161/HYPERTENSIONAHA.123.22055.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Reddy NM, Mayne SL, Pool LR, Gordon-Larsen P, Carr JJ, Terry JG, et al. Exposure to Neighborhood-Level Racial Residential Segregation in Young Adulthood to Midlife and Incident Subclinical Atherosclerosis in Black Adults: The Coronary Artery Risk Development in Young Adults Study. Circulation. Cardiovascular Quality and Outcomes . 2022;15:e007986. doi: 10.1161/CIRCOUTCOMES.121.007986.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Gao X, Kershaw KN, Barber S, Schreiner PJ, Do DP, Diez Roux AV, et al. Associations Between Residential Segregation and Incident Hypertension: The Multi-Ethnic Study of Atherosclerosis. Journal of the American Heart Association . 2022;11:e023084. doi: 10.1161/JAHA.121.023084.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Fields ND, Martin ZT, Hoggard LS, Erving CL, Udaipuria S, Blevins KM, et al. Does stress from incarceration of family and friends contribute to signs of early vascular ageing in African American women? Journal of Epidemiology and Community Health . 2024;78:745–751. doi: 10.1136/jech-2024-222227.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Thurston RC, Matthews KA. Racial and socioeconomic disparities in arterial stiffness and intima media thickness among adolescents. Social Science & Medicine (1982) . 2009;68:807–813. doi: 10.1016/j.socscimed.2008.12.029.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Islam SJ, Kim JH, Li X, Ko YA, Baltrus P, Mitchell GF, et al. Neighborhood characteristics and arterial stiffness among Black adults - Results from the Jackson Heart Study and Morehouse-Emory Cardiovascular Center for Health Equity. Vascular Medicine (London, England) . 2023;28:188–196. doi: 10.1177/1358863X221136163.. [DOI] [PubMed] [Google Scholar]
  • [222].Gump BB, Hruska B, Heffernan K, Brann LS, Voss M, Labrie-Cleary C, et al. Race, cortisol, and subclinical cardiovascular disease in 9- to 11-year-old children. Health Psychology: Official Journal of the Division of Health Psychology, American Psychological Association . 2023;42:657–667. doi: 10.1037/hea0001300.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Gump BB, Hill DT, Robinson M, Kannan K, Heffernan K, Atallah-Yunes NH, et al. Perfluoroalkyl substances (PFAS) and lead (Pb) as ”cardiovascular disruptors” in 9-11-year-old children living in Syracuse, New York, United States. Environmental Research . 2023;236:116758. doi: 10.1016/j.envres.2023.116758.. [DOI] [PubMed] [Google Scholar]
  • [224].Gump BB, Heffernan K, Brann LS, Hill DT, Labrie-Cleary C, Jandev V, et al. Exposure to Arsenic and Subclinical Cardiovascular Disease in 9- to 11-Year-Old Children, Syracuse, New York. JAMA Network Open . 2023;6:e2321379. doi: 10.1001/jamanetworkopen.2023.21379.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Hill DT, Petroni M, Larsen DA, Bendinskas K, Heffernan K, Atallah-Yunes N, et al. Linking metal (Pb, Hg, Cd) industrial air pollution risk to blood metal levels and cardiovascular functioning and structure among children in Syracuse, NY. Environmental Research . 2021;193:110557. doi: 10.1016/j.envres.2020.110557.. [DOI] [PubMed] [Google Scholar]
  • [226].Forde AT, Crookes DM, Suglia SF, Demmer RT. The weathering hypothesis as an explanation for racial disparities in health: a systematic review. Annals of Epidemiology . 2019;33:1–18.e3. doi: 10.1016/j.annepidem.2019.02.011.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Amato KR, Arrieta MC, Azad MB, Bailey MT, Broussard JL, Bruggeling CE, et al. The human gut microbiome and health inequities. Proceedings of the National Academy of Sciences of the United States of America . 2021;118:e2017947118. doi: 10.1073/pnas.2017947118.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Peters BA, Yi SS, Beasley JM, Cobbs EN, Choi HS, Beggs DB, et al. US nativity and dietary acculturation impact the gut microbiome in a diverse US population. The ISME Journal . 2020;14:1639–1650. doi: 10.1038/s41396-020-0630-6.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Nobre JG, Alpuim Costa D. “Sociobiome”: How do socioeconomic factors influence gut microbiota and enhance pathology susceptibility? - A mini-review. Frontiers in Gastroenterology . 2022;1. [Google Scholar]
  • [230].Kwak S, Usyk M, Beggs D, Choi H, Ahdoot D, Wu F, et al. Sociobiome - Individual and neighborhood socioeconomic status influence the gut microbiome in a multi-ethnic population in the US. NPJ Biofilms and Microbiomes . 2024;10:19. doi: 10.1038/s41522-024-00491-y.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Le Roy T, Moens de Hase E, Van Hul M, Paquot A, Pelicaen R, Régnier M, et al. Dysosmobacter welbionis is a newly isolated human commensal bacterium preventing diet-induced obesity and metabolic disorders in mice. Gut . 2022;71:534–543. doi: 10.1136/gutjnl-2020-323778.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Mallott EK, Sitarik AR, Leve LD, Cioffi C, Camargo CA, Jr, Hasegawa K, et al. Human microbiome variation associated with race and ethnicity emerges as early as 3 months of age. PLoS Biology . 2023;21:e3002230. doi: 10.1371/journal.pbio.3002230.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Carson TL, Wang F, Cui X, Jackson BE, Van Der Pol WJ, Lefkowitz EJ, et al. Associations Between Race, Perceived Psychological Stress, and the Gut Microbiota in a Sample of Generally Healthy Black and White Women: A Pilot Study on the Role of Race and Perceived Psychological Stress. Psychosomatic Medicine . 2018;80:640–648. doi: 10.1097/PSY.0000000000000614.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Knight RO, Cedillo YE, Judd SE, Baker EH, Frugé AD, Moellering DR. A cross-sectional study observing the association of psychosocial stress and dietary intake with gut microbiota genera and alpha diversity among a young adult cohort of black and white women in Birmingham, Alabama. BMC Women’s Health . 2024;24:142. doi: 10.1186/s12905-024-02968-6.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Dong TS, Gee GC, Beltran-Sanchez H, Wang M, Osadchiy V, Kilpatrick LA, et al. How Discrimination Gets Under the Skin: Biological Determinants of Discrimination Associated With Dysregulation of the Brain-Gut Microbiome System and Psychological Symptoms. Biological Psychiatry . 2023;94:203–214. doi: 10.1016/j.biopsych.2022.10.011.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Zhang X, Wang H, Kilpatrick LA, Dong TS, Gee GC, Labus JS, et al. Discrimination exposure impacts unhealthy processing of food cues: crosstalk between the brain and gut. Nature. Mental Health . 2023;1:841–852. doi: 10.1038/s44220-023-00134-9.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Ang QY, Alba DL, Upadhyay V, Bisanz JE, Cai J, Lee HL, et al. The East Asian gut microbiome is distinct from colocalized White subjects and connected to metabolic health. eLife . 2021;10:e70349. doi: 10.7554/eLife.70349.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Therdtatha P, Shinoda A, Nakayama J. Crisis of the Asian gut: associations among diet, microbiota, and metabolic diseases. Bioscience of Microbiota, Food and Health . 2022;41:83–93. doi: 10.12938/bmfh.2021-085.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Wang Y, Chen GC, Wang Z, Luo K, Zhang Y, Li Y, et al. Dietary Acculturation Is Associated With Altered Gut Microbiome, Circulating Metabolites, and Cardiovascular Disease Risk in US Hispanics and Latinos: Results From HCHS/SOL. Circulation . 2024;150:215–229. doi: 10.1161/CIRCULATIONAHA.124.069824.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Borrello K, Lim U, Park SY, Monroe KR, Maskarinec G, Boushey CJ, et al. Dietary Intake Mediates Ethnic Differences in Gut Microbial Composition. Nutrients . 2022;14:660. doi: 10.3390/nu14030660.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Walejko JM, Kim S, Goel R, Handberg EM, Richards EM, Pepine CJ, et al. Gut microbiota and serum metabolite differences in African Americans and White Americans with high blood pressure. International Journal of Cardiology . 2018;271:336–339. doi: 10.1016/j.ijcard.2018.04.074.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Molix L. Sex differences in cardiovascular health: does sexism influence women’s health? The American Journal of the Medical Sciences . 2014;348:153–155. doi: 10.1097/MAJ.0000000000000300.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Usselman CW, Lindsey ML, Robinson AT, Habecker BA, Taylor CE, Merryman WD, et al. Guidelines on the use of sex and gender in cardiovascular research. American Journal of Physiology. Heart and Circulatory Physiology . 2024;326:H238–H255. doi: 10.1152/ajpheart.00535.2023.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].O’Neil A, Scovelle AJ, Milner AJ, Kavanagh A. Gender/Sex as a Social Determinant of Cardiovascular Risk. Circulation . 2018;137:854–864. doi: 10.1161/CIRCULATIONAHA.117.028595.. [DOI] [PubMed] [Google Scholar]
  • [245].Arnlöv J, Pencina MJ, Amin S, Nam BH, Benjamin EJ, Murabito JM, et al. Endogenous sex hormones and cardiovascular disease incidence in men. Annals of Internal Medicine . 2006;145:176–184. doi: 10.7326/0003-4819-145-3-200608010-00005.. [DOI] [PubMed] [Google Scholar]
  • [246].Islam RM, Bell RJ, Handelsman DJ, McNeil JJ, Nelson MR, Reid CM, et al. Associations between blood sex steroid concentrations and risk of major adverse cardiovascular events in healthy older women in Australia: a prospective cohort substudy of the ASPREE trial. Healthy Longevity . 2022;3:e109–e118. doi: 10.1016/S2666-7568(22)00001-0.. The Lancet. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Heinze-Milne S, Banga S, Howlett SE. Low testosterone concentrations and risk of ischaemic cardiovascular disease in ageing: not just a problem for older men. Healthy Longevity . 2022;3:e83–e84. doi: 10.1016/S2666-7568(22)00008-3.. The Lancet. [DOI] [PubMed] [Google Scholar]
  • [248].Gonuguntla K, Badu I, Duhan S, Sandhyavenu H, Chobufo MD, Taha A, et al. Sex and Racial Disparities in Proportionate Mortality of Premature Myocardial Infarction in the United States: 1999 to 2020. Journal of the American Heart Association . 2024;13:e033515. doi: 10.1161/JAHA.123.033515.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Regitz-Zagrosek V, Gebhard C. Gender medicine: effects of sex and gender on cardiovascular disease manifestation and outcomes. Nature Reviews. Cardiology . 2023;20:236–247. doi: 10.1038/s41569-022-00797-4.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Clayton JA, Gaugh MD. Sex as a Biological Variable in Cardiovascular Diseases: JACC Focus Seminar 1/7. Journal of the American College of Cardiology . 2022;79:1388–1397. doi: 10.1016/j.jacc.2021.10.050.. [DOI] [PubMed] [Google Scholar]
  • [251].Smith JR, Thomas RJ, Bonikowske AR, Hammer SM, Olson TP. Sex Differences in Cardiac Rehabilitation Outcomes. Circulation Research . 2022;130:552–565. doi: 10.1161/CIRCRESAHA.121.319894.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Li S, Fonarow GC, Mukamal K, Xu H, Matsouaka RA, Devore AD, et al. Sex and Racial Disparities in Cardiac Rehabilitation Referral at Hospital Discharge and Gaps in Long-Term Mortality. Journal of the American Heart Association . 2018;7:e008088. doi: 10.1161/JAHA.117.008088.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Khan SS, Beach LB, Yancy CW. Sex-Based Differences in Heart Failure: JACC Focus Seminar 7/7. Journal of the American College of Cardiology . 2022;79:1530–1541. doi: 10.1016/j.jacc.2022.02.013.. [DOI] [PubMed] [Google Scholar]
  • [254].DeFilippis EM, Van Spall HGC. Is it Time for Sex-Specific Guidelines for Cardiovascular Disease? Journal of the American College of Cardiology . 2021;78:189–192. doi: 10.1016/j.jacc.2021.05.012.. [DOI] [PubMed] [Google Scholar]
  • [255].Stanhewicz AE, Wenner MM, Stachenfeld NS. Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. American Journal of Physiology. Heart and Circulatory Physiology . 2018;315:H1569–H1588. doi: 10.1152/ajpheart.00396.2018.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Lefferts WK, DeBlois JP, Augustine JA, Keller AP, Heffernan KS. Age, sex, and the vascular contributors to cerebral pulsatility and pulsatile damping. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2020;129:1092–1101. doi: 10.1152/japplphysiol.00500.2020.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].William EH, Nicole LS, Wesley KL, Jaqueline AA, Kevin SH. Sex differences in noninvasive estimates of left ventricular pressure energetics but not myocardial oxygen demand in young adults. Artery Research . 2014;8:197–204. doi: 10.1016/j.artres.2014.07.003.. [DOI] [Google Scholar]
  • [258].Nichols WW, Denardo SJ, Davidson JB, Huo T, Bairey Merz CN, Pepine CJ. Association of aortic stiffness and wave reflections with coronary flow reserve in women without obstructive coronary artery disease: An ancillary study from the National Heart, Lung, and Blood Institute-sponsored Women’s Ischemia Syndrome Evaluation (WISE) American Heart Journal . 2015;170:1243–1254. doi: 10.1016/j.ahj.2015.08.019.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Mitchell GF, Rong J, Larson MG, Cooper LL, Xanthakis V, Benjamin EJ, et al. Longitudinal Hemodynamic Correlates of and Sex Differences in the Evolution of Blood Pressure Across the Adult Lifespan: The Framingham Heart Study. Journal of the American Heart Association . 2023;12:e027329. doi: 10.1161/JAHA.122.027329.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Shim CY, Park S, Choi D, Yang WI, Cho IJ, Choi EY, et al. Sex differences in central hemodynamics and their relationship to left ventricular diastolic function. Journal of the American College of Cardiology . 2011;57:1226–1233. doi: 10.1016/j.jacc.2010.09.067.. [DOI] [PubMed] [Google Scholar]
  • [261].Russo C, Jin Z, Palmieri V, Homma S, Rundek T, Elkind MSV, et al. Arterial stiffness and wave reflection: sex differences and relationship with left ventricular diastolic function. Hypertension (Dallas, Tex.: 1979) . 2012;60:362–368. doi: 10.1161/HYPERTENSIONAHA.112.191148.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].DuPont JJ, Kenney RM, Patel AR, Jaffe IZ. Sex differences in mechanisms of arterial stiffness. British Journal of Pharmacology . 2019;176:4208–4225. doi: 10.1111/bph.14624.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Kannam JP, Levy D, Larson M, Wilson PW. Short stature and risk for mortality and cardiovascular disease events. The Framingham Heart Study. Circulation . 1994;90:2241–2247. doi: 10.1161/01.cir.90.5.2241.. [DOI] [PubMed] [Google Scholar]
  • [264].Smulyan H, Marchais SJ, Pannier B, Guerin AP, Safar ME, London GM. Influence of body height on pulsatile arterial hemodynamic data. Journal of the American College of Cardiology . 1998;31:1103–1109. doi: 10.1016/s0735-1097(98)00056-4.. [DOI] [PubMed] [Google Scholar]
  • [265].Widmer RJ, Prasad M, Gomaa M, Sara JDS, Reriani MK, Lerman LO, et al. Vascular reactivity to mental stress is associated with poor cardiovascular disease outcomes in females following acute coronary syndrome. Coronary Artery Disease . 2020;31:300–305. doi: 10.1097/MCA.0000000000000831.. [DOI] [PubMed] [Google Scholar]
  • [266].Vaccarino V, Almuwaqqat Z, Kim JH, Hammadah M, Shah AJ, Ko YA, et al. Association of Mental Stress-Induced Myocardial Ischemia With Cardiovascular Events in Patients With Coronary Heart Disease. JAMA . 2021;326:1818–1828. doi: 10.1001/jama.2021.17649.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Vaccarino V, Sullivan S, Hammadah M, Wilmot K, Al Mheid I, Ramadan R, et al. Mental Stress-Induced-Myocardial Ischemia in Young Patients With Recent Myocardial Infarction: Sex Differences and Mechanisms. Circulation . 2018;137:794–805. doi: 10.1161/CIRCULATIONAHA.117.030849.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Sullivan S, Young A, Garcia M, Almuwaqqat Z, Moazzami K, Hammadah M, et al. Sex Differences in Vascular Response to Mental Stress and Adverse Cardiovascular Events Among Patients With Ischemic Heart Disease. Arteriosclerosis, Thrombosis, and Vascular Biology . 2023;43:e112–e120. doi: 10.1161/ATVBAHA.122.318576.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Mehta LS, Velarde GP, Lewey J, Sharma G, Bond RM, Navas-Acien A, et al. Cardiovascular Disease Risk Factors in Women: The Impact of Race and Ethnicity: A Scientific Statement From the American Heart Association. Circulation . 2023;147:1471–1487. doi: 10.1161/CIR.0000000000001139.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Agarwala A, Michos ED, Samad Z, Ballantyne CM, Virani SS. The Use of Sex-Specific Factors in the Assessment of Women’s Cardiovascular Risk. Circulation . 2020;141:592–599. doi: 10.1161/CIRCULATIONAHA.119.043429.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Rajendran A, Minhas AS, Kazzi B, Varma B, Choi E, Thakkar A, et al. Sex-specific differences in cardiovascular risk factors and implications for cardiovascular disease prevention in women. Atherosclerosis . 2023;384:117269. doi: 10.1016/j.atherosclerosis.2023.117269.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [272].Berdahl JL, Moore C. Workplace harassment: double jeopardy for minority women. The Journal of Applied Psychology . 2006;91:426–436. doi: 10.1037/0021-9010.91.2.426.. [DOI] [PubMed] [Google Scholar]
  • [273].Meyerovitz CV, Juraschek SP, Ayturk D, Moore Simas TA, Person SD, Lemon SC, et al. Social Determinants, Blood Pressure Control, and Racial Inequities in Childbearing Age Women With Hypertension, 2001 to 2018. Journal of the American Heart Association . 2023;12:e027169. doi: 10.1161/JAHA.122.027169.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Gaffey AE, Cavanagh CE, Rosman L, Wang K, Deng Y, Sims M, et al. Depressive Symptoms and Incident Heart Failure in the Jackson Heart Study: Differential Risk Among Black Men and Women. Journal of the American Heart Association . 2022;11:e022514. doi: 10.1161/JAHA.121.022514.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Joseph JJ, Williams A, Azap RA, Zhao S, Brock G, Kline D, et al. Role of Sex in the Association of Socioeconomic Status With Cardiovascular Health in Black Americans: The Jackson Heart Study. Journal of the American Heart Association . 2023;12:e030695. doi: 10.1161/JAHA.123.030695.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Beatty DL, Matthews KA, Bromberger JT, Brown C. Everyday Discrimination Prospectively Predicts Inflammation Across 7-Years in Racially Diverse Midlife Women: Study of Women’s Health Across the Nation. The Journal of Social Issues . 2014;70:298–314. doi: 10.1111/josi.12061.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Troxel WM, Matthews KA, Bromberger JT, Sutton-Tyrrell K. Chronic stress burden, discrimination, and subclinical carotid artery disease in African American and Caucasian women. Health Psychology: Official Journal of the Division of Health Psychology, American Psychological Association . 2003;22:300–309. doi: 10.1037/0278-6133.22.3.300.. [DOI] [PubMed] [Google Scholar]
  • [278].Lewis TT, Everson-Rose SA, Powell LH, Matthews KA, Brown C, Karavolos K, et al. Chronic exposure to everyday discrimination and coronary artery calcification in African-American women: the SWAN Heart Study. Psychosomatic Medicine . 2006;68:362–368. doi: 10.1097/01.psy.0000221360.94700.16.. [DOI] [PubMed] [Google Scholar]
  • [279].Treiber FA, Kapuku GK, Davis H, Pollock JS, Pollock DM. Plasma endothelin-1 release during acute stress: role of ethnicity and sex. Psychosomatic Medicine . 2002;64:707–713. doi: 10.1097/01.psy.0000021952.59258.1c.. [DOI] [PubMed] [Google Scholar]
  • [280].Sheehy S, Aparicio HJ, Palmer JR, Cozier Y, Lioutas VA, Shulman JG, et al. Perceived Interpersonal Racism and Incident Stroke Among US Black Women. JAMA Network Open . 2023;6:e2343203. doi: 10.1001/jamanetworkopen.2023.43203.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Sheehy S, Brock M, Palmer JR, Albert MA, Cozier YC, Rosenberg L. Perceived Interpersonal Racism in Relation to Incident Coronary Heart Disease Among Black Women. Circulation . 2024;149:521–528. doi: 10.1161/CIRCULATIONAHA.123.066471.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Nayak A, Hicks AJ, Morris AA. Understanding the Complexity of Heart Failure Risk and Treatment in Black Patients. Circulation. Heart Failure . 2020;13:e007264. doi: 10.1161/CIRCHEARTFAILURE.120.007264.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].Wenger NK, Lloyd-Jones DM, Elkind MSV, Fonarow GC, Warner JJ, Alger HM, et al. Call to Action for Cardiovascular Disease in Women: Epidemiology, Awareness, Access, and Delivery of Equitable Health Care: A Presidential Advisory From the American Heart Association. Circulation . 2022;145:e1059–e1071. doi: 10.1161/CIR.0000000000001071.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [284].Quesada O, Crousillat D, Rodriguez F, Bravo-Jaimes K, Briller J, Ogunniyi MO, et al. Cardiovascular Disease in Hispanic Women: JACC Review Topic of the Week. Journal of the American College of Cardiology . 2024;83:1702–1712. doi: 10.1016/j.jacc.2024.02.039.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Pagan Lassalle P, DeBlois JP, Keller A, Stoner L, Heffernan KS. Central Blood Pressure and Subclinical Atherosclerotic Risk in Young Hispanic American Women. Ethnicity & Disease . 2021;31:489–500. doi: 10.18865/ed.31.4.489.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [286].Briggs Early K, Valencia SI, Stendell-Hollis N, Klyve D, Gee DL. Hypertension Prevalence and Related Risk Factors Among Mexican American Adults Are Increasing: National Health and Nutrition Examination Survey 1999 to 2018. Journal of the American Heart Association . 2024;13:e030126. doi: 10.1161/JAHA.123.030126.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Okoh AK, Young A, Garcia M, Sullivan S, Almuwaqqat Z, Hu Y, et al. Racial Differences in Mental Stress-Induced Transient Endothelial Dysfunction and Its Association With Cardiovascular Outcomes. Psychosomatic Medicine . 2023;85:431–439. doi: 10.1097/PSY.0000000000001201.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [288].Wagner JA, Tennen H, Finan PH, Ghuman N, Burg MM. Self-reported racial discrimination and endothelial reactivity to acute stress in women. Stress and Health: Journal of the International Society for the Investigation of Stress . 2013;29:214–221. doi: 10.1002/smi.2449.. [DOI] [PubMed] [Google Scholar]
  • [289].Guyll M, Matthews KA, Bromberger JT. Discrimination and unfair treatment: relationship to cardiovascular reactivity among African American and European American women. Health Psychology: Official Journal of the Division of Health Psychology, American Psychological Association . 2001;20:315–325. doi: 10.1037//0278-6133.20.5.315.. [DOI] [PubMed] [Google Scholar]
  • [290].Saban KL, Mathews HL, Bryant FB, Tell D, Joyce C, DeVon HA, et al. Perceived discrimination is associated with the inflammatory response to acute laboratory stress in women at risk for cardiovascular disease. Brain, Behavior, and Immunity . 2018;73:625–632. doi: 10.1016/j.bbi.2018.07.010.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [291].McKinnon II, Shah AJ, Lima B, Moazzami K, Young A, Sullivan S, et al. Everyday Discrimination and Mental Stress-Induced Myocardial Ischemia. Psychosomatic Medicine . 2021;83:432–439. doi: 10.1097/PSY.0000000000000941.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [292].Caceres BA, Streed CG, Jr, Corliss HL, Lloyd-Jones DM, Matthews PA, Mukherjee M, et al. Assessing and Addressing Cardiovascular Health in LGBTQ Adults: A Scientific Statement From the American Heart Association. Circulation . 2020;142:e321–e332. doi: 10.1161/CIR.0000000000000914.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [293].Kharlamov A, Sherriff N, Delles C, van der Harst P. Tackling cardiovascular healthcare disparities for LGBTQIA+ population: a call to action in the European Society of Cardiology. European Heart Journal . 2024;45:1291–1294. doi: 10.1093/eurheartj/ehad882.. [DOI] [PubMed] [Google Scholar]
  • [294].Streed CG, Jr, Beach LB, Caceres BA, Dowshen NL, Moreau KL, Mukherjee M, et al. Assessing and Addressing Cardiovascular Health in People Who Are Transgender and Gender Diverse: A Scientific Statement From the American Heart Association. Circulation . 2021;144:e136–e148. doi: 10.1161/CIR.0000000000001003.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Caceres BA, Sharma Y, Ravindranath R, Ensari I, Rosendale N, Doan D, et al. Differences in Ideal Cardiovascular Health Between Sexual Minority and Heterosexual Adults. JAMA Cardiology . 2023;8:335–346. doi: 10.1001/jamacardio.2022.5660.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Sharma Y, Bhargava A, Doan D, Caceres BA. Examination of Sexual Identity Differences in the Prevalence of Hypertension and Antihypertensive Medication Use Among US Adults: Findings From the Behavioral Risk Factor Surveillance System. Circulation. Cardiovascular Quality and Outcomes . 2022;15:e008999. doi: 10.1161/CIRCOUTCOMES.122.008999.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Everett B, Mollborn S. Differences in hypertension by sexual orientation among U.S. young adults. Journal of Community Health . 2013;38:588–596. doi: 10.1007/s10900-013-9655-3.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [298].Moreira JD, Haack K, White V, Bates ML, Gopal DM, Roepke TA. Importance of survey demographic questions to foster inclusion in medicine and research and reduce health inequities for LGBTQIA2S+ individuals. American Journal of Physiology. Heart and Circulatory Physiology . 2023;324:H856–H862. doi: 10.1152/ajpheart.00152.2023.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [299].Safer JD, Coleman E, Feldman J, Garofalo R, Hembree W, Radix A, et al. Barriers to healthcare for transgender individuals. Current Opinion in Endocrinology, Diabetes, and Obesity . 2016;23:168–171. doi: 10.1097/MED.0000000000000227.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Caceres BA, Sharma Y, Levine A, Wall MM, Hughes TL. Investigating the Associations of Sexual Minority Stressors and Incident Hypertension in a Community Sample of Sexual Minority Adults. Annals of Behavioral Medicine: a Publication of the Society of Behavioral Medicine . 2023;57:1004–1013. doi: 10.1093/abm/kaac073.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Juster RP, Rutherford C, Keyes K, Hatzenbuehler ML. Associations Between Structural Stigma and Allostatic Load Among Sexual Minorities: Results From a Population-Based Study. Psychosomatic Medicine . 2024;86:157–168. doi: 10.1097/PSY.0000000000001289.. [DOI] [PubMed] [Google Scholar]
  • [302].Maschino L, Cook S, Lucas T. Personal justice beliefs, everyday discrimination, and carotid intima media thickness in sexual minority men. Health Psychology: Official Journal of the Division of Health Psychology, American Psychological Association . 2024;43:1–6. doi: 10.1037/hea0001329.. [DOI] [PubMed] [Google Scholar]
  • [303].Taweh O, Moreira JD. Proposed mechanisms of hypertension and risk of adverse cardiovascular outcomes in LGBT communities. American Journal of Physiology. Heart and Circulatory Physiology . 2023;325:H522–H528. doi: 10.1152/ajpheart.00346.2023.. [DOI] [PubMed] [Google Scholar]
  • [304].Connelly PJ, Marie Freel E, Perry C, Ewan J, Touyz RM, Currie G, et al. Gender-Affirming Hormone Therapy, Vascular Health and Cardiovascular Disease in Transgender Adults. Hypertension (Dallas, Tex.: 1979) . 2019;74:1266–1274. doi: 10.1161/HYPERTENSIONAHA.119.13080.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [305].Murphy CN, Delles C, Davies E, Connelly PJ. Cardiovascular disease in transgender individuals. Atherosclerosis . 2023;384:117282. doi: 10.1016/j.atherosclerosis.2023.117282.. [DOI] [PubMed] [Google Scholar]
  • [306].Rytz CL, Pattar BSB, Mizen SJ, Lieb P, Parsons Leigh J, Saad N, et al. Transgender and Nonbinary Individuals’ Perceptions Regarding Gender-Affirming Hormone Therapy and Cardiovascular Health: A Qualitative Study. Circulation. Cardiovascular Quality and Outcomes . 2024;17:e011024. doi: 10.1161/CIRCOUTCOMES.124.011024.. [DOI] [PubMed] [Google Scholar]
  • [307].Rytz CL, Miranda KT, Ronksley PE, Saad N, Raj SR, Somayaji R, et al. Association between serum estradiol and cardiovascular health among transgender adults using gender-affirming estrogen therapy. American Journal of Physiology. Heart and Circulatory Physiology . 2024;327:H340–H348. doi: 10.1152/ajpheart.00151.2024.. [DOI] [PubMed] [Google Scholar]
  • [308].Gulanski BI, Flannery CA, Peter PR, Leone CA, Stachenfeld NS. Compromised endothelial function in transgender men taking testosterone. Clinical Endocrinology . 2020;92:138–144. doi: 10.1111/cen.14132.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [309].Aranda G, Mora M, Hanzu FA, Vera J, Ortega E, Halperin I. Effects of sex steroids on cardiovascular risk profile in transgender men under gender affirming hormone therapy. Endocrinologia, Diabetes Y Nutricion . 2019;66:385–392. doi: 10.1016/j.endinu.2018.11.004.. [DOI] [PubMed] [Google Scholar]
  • [310].Yamada M, Gam H, Ikegami N, Nishikawa Y, Ishikawa A, Funaki A, et al. Effects of acute aerobic exercise on arterial stiffness in transgender men. Frontiers in Physiology . 2023;14:1294284. doi: 10.3389/fphys.2023.1294284.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [311].Emi Y, Adachi M, Sasaki A, Nakamura Y, Nakatsuka M. Increased arterial stiffness in female-to-male transsexuals treated with androgen. The Journal of Obstetrics and Gynaecology Research . 2008;34:890–897. doi: 10.1111/j.1447-0756.2008.00857.x.. [DOI] [PubMed] [Google Scholar]
  • [312].Cunha FS, Bachega TASS, Costa EMF, Brito VN, Alvares LA, Costa-Hong VA, et al. Arterial Stiffness in Transgender Men Receiving Long-term Testosterone Therapy. Journal of the Endocrine Society . 2023;7:bvad040. doi: 10.1210/jendso/bvad040.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [313].Horton AC, Wilkinson MM, Kilanowski-Doroh I, Dong Z, Liu J, Ogola BO, et al. Dihydrotestosterone induces arterial stiffening in female mice. Biology of Sex Differences . 2024;15 doi: 10.1186/s13293-024-00586-3.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [314].New G, Duffy SJ, Harper RW, Meredith IT. Long-term oestrogen therapy is associated with improved endothelium-dependent vasodilation in the forearm resistance circulation of biological males. Clinical and Experimental Pharmacology & Physiology . 2000;27:25–33. doi: 10.1046/j.1440-1681.2000.03195.x.. [DOI] [PubMed] [Google Scholar]
  • [315].New G, Timmins KL, Duffy SJ, Tran BT, O’Brien RC, Harper RW, et al. Long-term estrogen therapy improves vascular function in male to female transsexuals. Journal of the American College of Cardiology . 1997;29:1437–1444. doi: 10.1016/s0735-1097(97)00080-6.. [DOI] [PubMed] [Google Scholar]
  • [316].Moreira Allgayer RMC, Borba GDS, Moraes RS, Ramos RB, Spritzer PM. The Effect of Gender-Affirming Hormone Therapy on the Risk of Subclinical Atherosclerosis in the Transgender Population: A Systematic Review. Endocrine Practice: Official Journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists . 2023;29:498–507. doi: 10.1016/j.eprac.2022.12.017.. [DOI] [PubMed] [Google Scholar]
  • [317].Garcia-Fernandez H, Arenas-de Larriva AP, Lopez-Moreno J, Gutierrez-Mariscal FM, Romero-Cabrera JL, Molina-Abril H, et al. Sex-specific differences in intestinal microbiota associated with cardiovascular diseases. Biology of Sex Differences . 2024;15 doi: 10.1186/s13293-024-00582-7.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [318].Zhang Y, Zhang X, Chen D, Lu J, Gong Q, Fang J, et al. Causal associations between gut microbiome and cardiovascular disease: A Mendelian randomization study. Frontiers in Cardiovascular Medicine . 2022;9:971376. doi: 10.3389/fcvm.2022.971376.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [319].Weiner CM, Khan SE, Leong C, Ranadive SM, Campbell SC, Howard JT, et al. Association of enterolactone with blood pressure and hypertension risk in NHANES. PloS One . 2024;19:e0302254. doi: 10.1371/journal.pone.0302254.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [320].Virwani PD, Qian G, Hsu MSS, Pijarnvanit TKKTS, Cheung CNM, Chow YH, et al. Sex Differences in Association Between Gut Microbiome and Essential Hypertension Based on Ambulatory Blood Pressure Monitoring. Hypertension (Dallas, Tex.: 1979) . 2023;80:1331–1342. doi: 10.1161/HYPERTENSIONAHA.122.20752.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [321].Ahmed S, Spence JD. Sex differences in the intestinal microbiome: interactions with risk factors for atherosclerosis and cardiovascular disease. Biology of Sex Differences . 2021;12:35. doi: 10.1186/s13293-021-00378-z.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [322].Price CA, Jospin G, Brownell K, Eisen JA, Laraia B, Epel ES. Differences in gut microbiome by insulin sensitivity status in Black and White women of the National Growth and Health Study (NGHS): A pilot study. PloS One . 2022;17:e0259889. doi: 10.1371/journal.pone.0259889.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [323].Colonetti T, Limas Carmo Teixeira D, Grande AJ, Rodrigues Uggioni ML, Generoso J, Harding S, et al. The role of intestinal microbiota on pre-eclampsia: Systematic review and meta-analysis. European Journal of Obstetrics, Gynecology, and Reproductive Biology . 2023;291:49–58. doi: 10.1016/j.ejogrb.2023.10.003.. [DOI] [PubMed] [Google Scholar]
  • [324].Jama HA, Dona MSI, Dinakis E, Nakai M, Paterson MR, Shihata WA, et al. Maternal Diet and Gut Microbiota Influence Predisposition to Cardiovascular Disease in Offspring. Circulation Research . 2024;135:537–539. doi: 10.1161/CIRCRESAHA.124.324614.. [DOI] [PubMed] [Google Scholar]
  • [325].Wu Z, Ge M, Liu J, Chen X, Cai Z, Huang H. The gut microbiota composition and metabolites are different in women with hypertensive disorders of pregnancy and normotension: A pilot study. The Journal of Obstetrics and Gynaecology Research . 2024;50:334–341. doi: 10.1111/jog.15844.. [DOI] [PubMed] [Google Scholar]
  • [326].Wu X, Li Q, Lin D, Cai J, Huang H, Tan H. Gut microbiota and hypertensive disorders in pregnancy: evidence from the Mendelian randomization study. Aging . 2023;15:9105–9127. doi: 10.18632/aging.205019.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].Jin J, Gao L, Zou X, Zhang Y, Zheng Z, Zhang X, et al. Gut Dysbiosis Promotes Preeclampsia by Regulating Macrophages and Trophoblasts. Circulation Research . 2022;131:492–506. doi: 10.1161/CIRCRESAHA.122.320771.. [DOI] [PubMed] [Google Scholar]
  • [328].Rosendale N, Wood AJ, Leung CW, Kim AS, Caceres BA. Differences in Cardiovascular Health at the Intersection of Race, Ethnicity, and Sexual Identity. JAMA Network Open . 2024;7:e249060. doi: 10.1001/jamanetworkopen.2024.9060.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [329].Pal S, Morgan X, Dar HY, Gacasan CA, Patil S, Stoica A, et al. Gender-affirming hormone therapy preserves skeletal maturation in young mice via the gut microbiome. The Journal of Clinical Investigation . 2024;134:e175410. doi: 10.1172/JCI175410.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [330].Joseph RP, Ainsworth BE, Keller C, Dodgson JE. Barriers to Physical Activity Among African American Women: An Integrative Review of the Literature. Women & Health . 2015;55:679–699. doi: 10.1080/03630242.2015.1039184.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [331].Hornbuckle LM. Running while Black: A distinctive safety concern and barrier to exercise in White neighborhoods. Preventive Medicine Reports . 2021;22:101378. doi: 10.1016/j.pmedr.2021.101378.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [332].Moreau KL, Clayton ZS, DuBose LE, Rosenberry R, Seals DR. Effects of regular exercise on vascular function with aging: Does sex matter? American Journal of Physiology. Heart and Circulatory Physiology . 2024;326:H123–H137. doi: 10.1152/ajpheart.00392.2023.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [333].Howden EJ, Perhonen M, Peshock RM, Zhang R, Arbab-Zadeh A, Adams-Huet B, et al. Females have a blunted cardiovascular response to one year of intensive supervised endurance training. Journal of Applied Physiology (Bethesda, Md.: 1985) . 2015;119:37–46. doi: 10.1152/japplphysiol.00092.2015.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [334].Ji H, Gulati M, Huang TY, Kwan AC, Ouyang D, Ebinger JE, et al. Sex Differences in Association of Physical Activity With All-Cause and Cardiovascular Mortality. Journal of the American College of Cardiology . 2024;83:783–793. doi: 10.1016/j.jacc.2023.12.019.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [335].Swift DL, Johannsen NM, Earnest CP, Newton RL, Jr, McGee JE, Church TS. Cardiorespiratory Fitness and Exercise Training in African Americans. Progress in Cardiovascular Diseases . 2017;60:96–102. doi: 10.1016/j.pcad.2017.06.001.. [DOI] [PubMed] [Google Scholar]
  • [336].Heffernan KS, Jae SY, Fernhall B. Racial differences in arterial stiffness after exercise in young men. American Journal of Hypertension . 2007;20:840–845. doi: 10.1016/j.amjhyper.2007.03.015.. [DOI] [PubMed] [Google Scholar]
  • [337].Pandey A, Park BD, Ayers C, Das SR, Lakoski S, Matulevicius S, et al. Determinants of Racial/Ethnic Differences in Cardiorespiratory Fitness (from the Dallas Heart Study) The American Journal of Cardiology . 2016;118:499–503. doi: 10.1016/j.amjcard.2016.05.043.. [DOI] [PubMed] [Google Scholar]
  • [338].Cooper Z, Rodriguez WA, Hardwick J, Arena R, Laddu DR. Cardiorespiratory fitness and physical activity in the lens of social justice - Reporting on the disparities that exist. Progress in Cardiovascular Diseases . 2024;83:92–96. doi: 10.1016/j.pcad.2024.02.007.. [DOI] [PubMed] [Google Scholar]
  • [339].Friedman EM, Williams DR, Singer BH, Ryff CD. Chronic discrimination predicts higher circulating levels of E-selectin in a national sample: the MIDUS study. Brain, Behavior, and Immunity . 2009;23:684–692. doi: 10.1016/j.bbi.2009.01.002.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [340].Szanton SL, Rifkind JM, Mohanty JG, Miller ER, 3rd, Thorpe RJ, Nagababu E, et al. Racial discrimination is associated with a measure of red blood cell oxidative stress: a potential pathway for racial health disparities. International Journal of Behavioral Medicine . 2012;19:489–495. doi: 10.1007/s12529-011-9188-z.. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Reviews in Cardiovascular Medicine are provided here courtesy of IMR Press

RESOURCES