Skip to main content
Molecular Metabolism logoLink to Molecular Metabolism
. 2018 May 16;15:45–55. doi: 10.1016/j.molmet.2018.05.008

Sex differences in lipid and lipoprotein metabolism

Brian T Palmisano 1,2, Lin Zhu 3, Robert H Eckel 4, John M Stafford 1,2,3,
PMCID: PMC6066747  PMID: 29858147

Abstract

Background

Endogenous sex hormones are important for metabolic health in men and women. Before menopause, women are protected from atherosclerotic cardiovascular disease (ASCVD) relative to men. Women have fewer cardiovascular complications of obesity compared to men with obesity. Endogenous estrogens have been proposed as a mechanism that lessens ASCVD risk, as risk of glucose and lipid abnormalities increases when endogenous estrogens decline with menopause. While baseline risk is higher in males than females, endogenously produced androgens are also protective against fatty liver, diabetes and ASCVD, as risk goes up with androgen deprivation and with the decline in androgens with age.

Scope of Review

In this review, we discuss evidence of how endogenous sex hormones and hormone treatment approaches impact fatty acid, triglyceride, and cholesterol metabolism to influence metabolic and cardiovascular risk. We also discuss potential reasons for why treatment strategies with estrogens and androgens in older individuals fail to fully recapitulate the effects of endogenous sex hormones.

Major Conclusions

The pathways that confer ASCVD protection for women are of potential therapeutic relevance. Despite protection relative to men, ASCVD is still the major cause of mortality in women. Additionally, diabetic women have similar ASCVD risk as diabetic men, suggesting that the presence of diabetes may offset the protective cardiovascular effects of being female through unknown mechanisms.

Keywords: Estrogen, Androgens, Sex differences, Cardiovascular disease, Lipid metabolism, Obesity

1. Introduction

Prior to menopause, women are protected from myocardial infarction (MI) compared to age-matched men. The age of onset for the first myocardial infarction is ∼10 years later for women compared to men. Furthermore, at any given age, women have one-third to one-half of the risk of cardiovascular disease relative to men [1], [2]. The mechanisms for this protection include the effects of sex hormones as well as hormone-independent effects of the sex-chromosomes in tissues throughout the body. The protective effect of endogenously produced estrogens with regard to ASCVD is supported in humans by the increase in risk when levels of naturally cycling estrogens and progestins decline with menopause. Despite lower risk than men, ASCVD is still the major cause of mortality in women, causing 1 in 3 deaths in women. Over the past two decades the prevalence of myocardial infarctions has increased in women midlife (ages 35–54 years), despite a decline in similarly aged men [3]. The increased prevalence diabetes in women partially offsets the protection conferred by being female [4], [5], [6].

The mechanisms conferring protection from atherosclerotic cardiovascular disease (ASCVD) in non-diabetic women have been of considerable research interest. Estrogens have effects in many organ systems that contribute to cardiovascular risk vs. protection, including regulation of liver lipid metabolism and serum lipoprotein levels. The primary movement of lipids between tissues occurs either as free fatty acids released by adipose tissue or in the form of lipoprotein carriers made primarily by the liver and gut (chylomicrons and very low density lipoprotein (VLDL) for triglyceride (TG), and low density lipoprotein (LDL) and high-density lipoprotein (HDL) for cholesterol). Many aspects of hepatic fatty acid, TG, and cholesterol biology are regulated by endogenous estrogens and androgens, but the physiologic control of these effects is distributed among different tissues, primarily adipose and liver, but increasingly it is recognized that CNS effects of sex hormones also contribute. More is known about estrogen control of lipid metabolism than androgen control of lipid metabolism. Estrogens mediate their effects through three receptors, Estrogen Receptor alpha (ERα), Estrogen Receptor beta (ERβ) and G-protein coupled Estrogen Receptor (GPER). The liver is an important site where fatty acids, TG, and cholesterol metabolism are coordinated to meet metabolic needs in normal physiology. Despite the protective effects of endogenous sex hormones in males and females, physicians are confronted with limited ability to recapitulate these physiologic roles with estrogen treatment approaches for women after menopause, or older men with androgen deficiency. Thus, there is an important therapeutic opportunity to develop tissue selective and pathway-preferential sex hormones in order to recapitulate this protective physiology in younger adults.

2. Mechanisms of estrogen signaling

Many aspects of sex-differences in physiology arise due to the mechanisms of the sex-hormones. Estrogen signaling pathways have pleotropic effects on many tissues and pathways that govern lipid and lipoprotein metabolism, but our understanding of these effects is complicated in that there are numerous endogenous estrogens and they differ between species [7]. 17β estradiol is the predominant endogenous estrogen in humans. It is made by the ovaries and circulates in plasma associated with sex-hormone binding globulin. Estrogens are lipophilic steroid hormones that are thought to passively diffuse through the plasma membrane into the cytoplasm and nucleus where they bind the steroid nuclear hormone receptors, ERα and ERβ [8]. Unliganded ERα and ERβ are kept inactive by association to Heat Shock Protein 90 (Hsp90) complexes. Binding of estrogens to ERα or ERβ promotes dissociation from Hsp90, dimerization, and translocation into the nucleus to activate gene transcription [8]. These genomic sequences are referred to as Estrogen Response Elements (EREs) and are commonly found in the promoter or enhancer regions of genes whose transcription is regulated by estrogens. The liver is a major tissue that impacts lipid metabolism in response to estrogen signaling. Over 1000 human liver genes display a sex-bias in their expression [9]. The top biological pathways are in lipid metabolism and genes related to ASCVD [9], [10]. Additionally, chromatin immunoprecipitation assay revealed 43 of the lipid genes are transcriptionally regulated by ERα [11]. In the mouse, scores of liver genes involved in TG and cholesterol metabolism vary with the four-day estrous cycle of the mouse in an ERα-dependent manner [12], demonstrating a tight coordination of liver lipid metabolism with reproductive needs.

Estrogens also regulate liver lipid metabolism by modifying signaling to estrogen receptors localized to the plasma membrane by palmitoylation of a serine residue and association with caveolin-1 [13], [14], [15], [16]. This membrane-localized estrogen signaling through ERα and ERβ activates ERK 1/2 and the PI3K pathways [14], [15], [16]. The benefits of the ERα agonist propyl-pyrazole-triol (PPT) with regard to liver lipid metabolism are largely accounted for by membrane-localized ERα [17]. Additionally, estrogens can activate the cell surface G-protein coupled Estrogen Receptor (GPER, also called Gpr30), which is expressed in multiple tissues including liver [18], [19], [20]. Activation of GPER by estrogens promotes increased cyclic AMP (cAMP) and intracellular Ca2+ [19]. Whole-body deletion of GPER promotes atherosclerosis and increases LDL cholesterol levels in mice [21]. The relative contributions of estrogen signaling through ERα, ERβ, or GPER with regard to lipid, lipoprotein metabolism, and ASCVD are not well defined.

3. Mechanisms of testosterone signaling

Testosterone is the major male sex hormone, dictates male sexual development, and maintains male sexual function throughout life after puberty. Testosterone can alter cell metabolism through effects on gene transcription through the androgen receptor (AR) and through non-genomic signaling mechanisms, similar to estrogen signaling through ERα and ERβ. AR is a classic steroid hormone receptor that enters the nucleus after binding of testosterone or dihydrotestosterone [22], [23]. Once in the nucleus, AR regulates gene transcription by classic hormone-receptor signaling to androgen-response elements (AREs) in promoter and enhancer regions of target genes [22], [23]. In addition to classic ARE-mediated transcription, AR activates several non-genomic signaling pathways, including the Mapk pathway and the PI3K/Akt pathway [24]. Membrane-associated AR can also mediate signaling via regulation of intracellular calcium [24]. In addition, other plasma membrane associated receptors, including the sex-hormone binding globulin receptor and epidermal growth factor receptor, can mediate effects of testosterone on cell signaling [25]. This modulation of cell signaling by testosterone not only changes intracellular signaling events but also influences transcription by both AR and non-AR transcription factors.

4. Sex-differences in body composition are due tissue-distributed actions of sex-hormones and chromosomal effects

A key aspect of sex-differences in lipid and lipoprotein metabolism is the differential ability of subcutaneous fat to expand and store excess nutrient calories as TGs in males vs. females. The concept of women as “pear-shaped”, with more subcutaneous fat, and men as “apple-shaped”, with more visceral fat, was put forth by Vague in 1947 [26]. Body fat distribution, as measured by waist-to-hip ratio, predicts risk of cardiovascular disease [27], [28], and women appear to be at lower risk of cardiovascular disease due to a more favorable body fat distribution in lower body, subcutaneous stores. Two large prospective studies confirmed that body fat distribution does indeed predict risk of future cardiovascular disease [27], [28], [29]. Exercise and weight loss can reduce waist to hip ratio and reduce risk of cardiovascular disease, but long-term weight loss in obese patients remains a clinical challenge due to weight regain. Furthermore, a pooled meta-analysis found that waist-to-hip ratio contributed to cardiovascular risk similarly between men and women [30]. In the intra-abdominal compartment, men have larger fat cell sizes than women, whereas in subcutaneous depots, women have larger fat cell sizes than men [31]. Overall, important differences between abdominal wall and subcutaneous adipose tissue sites is apparent and may have physiologic and pathophysiologic implications [32].

There may be racial differences in the distribution of adipose in upper body vs. lower-body compartments. Women of Caucasian background are more likely to gain weight with an upper-body distribution where adipocytes are less responsive to the stimulatory effects of insulin on glucose uptake, and less sensitive to the anti-lipolytic effects of insulin. However, for any abdominal circumference, women of African ancestry have relatively less intra-abdominal vs. subcutaneous fat distribution. Moreover, fat cells from upper body and lower body were equally sensitive to the effects of insulin to regulate nutrient uptake and lipolysis, which the authors state correlated with clinical data that upper-body obesity in Caucasian women but not in African American women is associated with insulin resistance and dyslipidemia [33], [34]. Conversely, testosterone seems to promote central adipose storage, as men with hypogonadism store a greater portion of dietary fatty acids in lower body subcutaneous fat (a female-like storage distribution) [35].

The distribution of fat in subcutaneous depots in women is, in part, attributable to sex-hormone signaling in adipocytes. Loss of ovarian hormones with menopause is associated with a relative re-distribution of body fat from a more subcutaneous distribution to a more visceral distribution [36]. Estrogen signaling through ERα in pre-adipocytes drives differentiation of white adipocytes [37]. Female and male mice with selective deletion of ERα in adipocytes have less subcutaneous adiposity [37]. Part of the improved subcutaneous adipose tissue nutrient storage in women is because estrogen promotes insulin sensitivity and adiponectin action, two mediators of subcutaneous fat storage [38]. Castration of male mice enhances insulin sensitivity and increases lipolytic rates from adipocytes [39].

The physiology of subcutaneous and visceral adipose in women is quite distinct in humans, with over 2800 genes differentially expressed [32]. Using Affymetrix arrays to assess gene expression in four subcutaneous sites, nearly 3000/24,000 transcripts were differentially expressed between all sites. Major differences were found between the hip and flank compared to the lower and upper abdomen, but no genes were significantly different when the lower abdomen was compared to the upper abdomen and the hip to the flank. Genes involved in the complement and coagulation cascades and immune responses showed increased expression in the lower abdomen compared to the flank. Genes involved in basic biochemical metabolism including insulin signaling, the urea cycle, glutamate metabolism, arginine and proline metabolism and amino-sugar metabolism had higher expression in the lower abdomen compared to the hip [32].

Not all of estrogen-control of adiposity occurs at the level of adipocytes. Estrogen signaling in the CNS also contributes to body weight and adiposity, and thus at least indirectly to lipid and lipoprotein metabolism in animal models. Mice with hypothalamic deletions of ERα have hyperphagia and obesity [40], [41]. This CNS-mediated ERα effect seems to require lipid sensing through neuronal lipoprotein lipase [42]. Additionally, some of the contribution to body composition is genetic, as genome-wide association meta-analyses of traits related to waist and hip circumferences revealed 19 loci associated with increased hip fat distribution [43], which mirrored the findings of a meta-analysis of genetic studies [44]. Reue and colleagues have developed a mouse model, which they term “the four core genotypes”, which allows them to separate the hormonal and chromosomal contributions to biology. They found that the presence of two X chromosomes in females allows for expansion of adipose tissue independent of gonadal hormones [45]. Thus, the distribution of adiposity in men vs. women, and in women before vs. after menopause represents effects of gene regulation in multiple tissues, as well as hormonal effects of estrogens and androgens.

5. Sex-differences in free fatty acid metabolism, the relationship between adipose muscle and liver physiology

The sex-differences in adiposity distribution and TG storage capacity impact the flux of fatty acids that occurs in fasting and feeding. In response to obesity both men and women have increased fatty acid release into blood. Adipose in the visceral (or splanchnic) compartment has a higher contribution to fatty acid delivery to the liver compared to adipose from subcutaneous leg fat [46]. The liver takes up these fatty acids and assembles them into TGs, which are subsequently packaged into TG-rich VLDL particles for export from the liver. Obesity is associated with increased production of apoB-rich VLDL-TG particles by the liver to a greater degree in men than in women [47], [48]. Lower VLDL-TG levels produced by the liver are in part secondary to lower fatty acid delivery to the liver due to enhanced fatty acid clearance by muscle in women [49], [50], [51]. It is also known that in response to fatty acid delivery to the liver that women secrete VLDL particles that are more TG-rich [52], which would help the liver export liver TGs and prevent liver fat accumulation with obesity. In response to obesity, women do have increased VLDL production, especially when this obesity is abdominal [53]. Production of more TG-rich VLDL is matched with accelerated VLDL-TG clearance rates in women [54], which collectively contribute to less liver fat and lower plasma VLDL-TG levels with obesity in women.

Not all studies suggest a strong correlation between blood fatty acid levels and BMI. The strongest relationship between high fatty acid levels and BMI is at higher levels of BMI. Expressed per cell, women have higher lipoprotein lipase (LPL) activity than men, greater lipolysis in response to lipolytic stimuli of fasting, but greater suppression of lipolysis by insulin in the fed state [31], [39]. However, using tracers for FFA flux, Mittendorfer and colleagues found that males and females had a similar increase in the rate of appearance of free fatty acids (FFA-Ra) with increased adiposity. The FFA-Ra in relationship to fat-free mass (FFM) was greater in women than in men, but not related abdominal fat mass in this study [55]. Some studies show that blood levels of fatty acids are higher in women than men with obesity, but with a less severe impact on insulin resistance (reviewed in [56]). Thus, the appearance of free fatty acids in the blood has a complex association with obesity and appears to depend a great deal on adipocyte function.

Even with increased lipolysis associated with obese states, females are resistant to free fatty acid-induced insulin resistance. In experimental models, infusion of Intralipid plus heparin is a way to acutely elevate serum fatty acid levels and define fatty acid-mediated insulin resistance independent of diet or weight differences between groups. Female rodents are resistant to free fatty acid-induced insulin resistance and maintain normal insulin action in muscle and liver [57]. Female humans are also protected from insulin resistance due to Intralipid, and muscle is an important mediator of this protection [49]. For men, this accumulation of intramuscular TG associates with insulin resistance and impaired glucose disposal by muscle, whereas women are relatively protected from insulin resistance associated with intramuscular TG [58]. Thus, sex-differences fatty acid handling and intramuscular TG metabolism relate to sex-differences in risk for glucose tolerance and type-2 diabetes.

6. Sex differences in triglyceride metabolism

In the fasted state, most TGs circulate in the form of apoB100-containing VLDL made by the liver. As mentioned above, women secrete more TG-rich VLDL particles, which are matched by higher rates of LPL-mediated VLDL-TG clearance, contributing to overall lower blood TG levels in women with obesity. In the fed state, TGs circulate in the form of apoB48-containing chylomicrons. In response to both short and long-term high-fat feeding, women have better clearance of meal-related TGs and increased storage of those nutrients in subcutaneous gluteal fat, rather than abdominal fat, whereas storage is similar in abdominal vs. subcutaneous fat in men [54], [59], [60]. The increased TG clearance by subcutaneous fat is more pronounced with addition of high-carbohydrates to the diet in women [61]. Increased blood levels of TGs correlate with increased risk of cardiovascular disease, a correlation which is stronger in women in the Framingham Heart Study [62]. Increased ASCVD risk due to TGs is reduced when corrected for low HDL cholesterol in men, but the correlation remains even after correcting for low HDL cholesterol in women [63], [64]. The reason that high TGs correlate more strongly with ASCVD risk in women than in men remains unknown and is an extremely important uncertainty to answer.

Some of the sex-differences in VLDL-TG biology and fatty liver are due to liver estrogen signaling, which limits non-alcoholic fatty liver disease (NAFLD) with obesity. Loss of global estrogens after menopause, in experimental models, and due to estrogen antagonists lead to liver fat accumulation. Tamoxifen (TMX) is an anti-estrogen drug used for the treatment of hormone-sensitive breast cancer, which increases NAFLD and steatohepatitis [65], [66]. Rates of NAFLD increase as women transition to menopause [67], [68]. Aging is a natural risk factor for NAFLD, which may confound the impact of menopause on risk of NAFLD. In women undergoing surgical menopause, the risk of NAFLD is increased nearly two-fold [69]. This biology is modeled in animal models in which removal of the ovaries by ovariectomy leads to an accumulation of liver TG content [70], [71], [72]. Depletion of endogenous estrogens with 4-vinylcyclohexene diepoxide (VCD) also causes insulin resistance, fatty liver, and dyslipidemia [73]. Thus, absence of ovarian hormones leads to an increased risk of NAFLD that is at least partially reversible with estrogen treatment in rodents and postmenopausal women.

Loss of ovarian hormones also leads to weight gain, confounding definition of effects due to loss of estrogen signaling. To define tissue-specific contributions of estrogen signaling, several groups have created hepatocyte-specific ERα-knock out mice. The ability of estrogens to reduce liver steatosis is lost with deletion of hepatocyte ERα, suggesting that estrogens are acting directly in the liver to reduce TG content through ERα [12], [74], [75]. Loss of hepatocyte ERα results in loss of estrogen regulation of target genes [74], [75], increased expression of lipid synthesis genes [76], and impaired estrogen-regulation of other lipid metabolic target genes [77]. One proposed mechanism for ERα regulation of lipid synthesis targets involves estrogen-ERα regulation of the nuclear receptor Small Heterodimer Partner (SHP), a target gene of ERα [74], [78]. Additionally, estrogen-ERα regulation of liver lipid metabolism has been proposed to act via microRNA mir-125b [79].

The mechanisms by which estrogen signaling protects against hepatic steatosis includes reductions in de novo lipogenesis (hepatic synthesis of fatty acids) as reported by different laboratories. Using a combination of chromatin immunoprecipitation and tiled microarrays (ChIP-on-chip) approach, Gao et al. identified binding regions of ERα to DNA in intact chromatin in the liver [11]. This analysis revealed 19 gene ontology (GO) categories including lipid biosynthesis (GO 0008610) and fatty acid metabolism (GO 0006520) that are significantly enriched for genes that had ERα recruited to their promoter after 2 h of estradiol treatment [11]. Conventional ChIP followed by qPCR shows binding to ERα to promoter regions of lipogenic genes including STAT3 and SHP are consistently increased after treatment with estradiol or ERα agonist [11]. This report is consistent with their previous observation that estradiol treatment promotes ERα binding to STAT3 promoter and STAT3-Tyr phosphorylation, which subsequently suppresses Fasn, Scd1, Acaa1, and Gpam expression in the liver in ob/ob mice [80]. Many of these genes also vary in a tetradian-manner with the mouse's estrus cycle and are lower during the high-estrogen phase of the cycle, which correlates with lower liver fat content [12].

Estradiol treatment suppresses liver lipogenesis by maintaining ACC phosphorylation, which correlates with decreased tracer incorporation into hepatic lipid [75], [81], [82], [83]. This mechanism likely contributes to the correction of pathway-selective insulin resistance in the liver with estradiol treatment [75]. ACC phosphorylation is regulated by AMPKα phosphorylation in the liver. Estradiol induces signal transduction through ERα, which localizes to both the plasma membrane and nucleus. Activation of estrogen signaling by the ERα agonist PPT promotes AMPK phosphorylation via ERα localized exclusively at the plasma membrane, but not in ERα knockout mice. This study demonstrates that signaling changes mediated by membrane-localized ERα result in important metabolic effects independent of nuclear ERα [17]. Additionally, oral conjugated estrogens (CE) and the selective estrogen receptor modulator bazedoxifene (BZA) also promote AMPK phosphorylation via ERα in liver after ovariectomy [84]. In this study by Kim et al., oral CE and BZA reduce hepatic FAS expression and FAS activity and are associated with decreased liver TG accumulation in female mice after ovariectomy [84].

Liver estrogen signaling also likely promotes fatty acid oxidation in liver. Levels of mRNA for CPT-1, a protein to transport fatty acid into mitochondrial for β-oxidation are induced with estradiol treatment after ovariectomy. Estradiol treatment after ovariectomy also increases oxygen consumption and liver ATP production along with changes in UCP2 expression in the liver [85]. Additionally, estradiol and CE increase production of FGF21 by the liver which may also increase hepatic fatty acid oxidation [84].

7. Sex-differences in cholesterol metabolism

There are important sex-differences in cholesterol metabolism which likely contribute to the large sex-differences in rates of ASCVD. Women have a nearly decade-long delay in first myocardial infarction compared to men, which may be largely driven by hormonal effects of estrogens on cholesterol metabolism before menopause [1], [2], [86]. Even after levels of ovarian hormones decline with menopause, women have lower risk of cardiovascular disease relative to men [1], [2], [87]. The prevalence of coronary heart disease (CHD) according to 2011–2014 NHANES data is 19.7% in men and 11% in women 60–79 years of age, and similar trends for older individuals [2]. Rates of heart attack are less than half among women in this high-risk age group [2]. It is not well known if these protective effects exhibited by women later in life are conferred by chromosomal effects, such as established by Reue and colleagues, or due to a different trajectory set by the pre-menopausal hormonal environment.

Liver estrogen signaling may contribute to sex-differences in atherosclerosis by promoting the hepatic steps of reverse cholesterol transport (RCT). RCT is the process of cholesterol removal from peripheral tissues culminating in delivery of cholesterol to the liver for conversion to bile acids, which are ultimately delivered into the feces (reviewed in [88]). Estrogen's role in the early steps of the RCT pathway is controversial in humans. The cholesterol efflux capacity of macrophages is enhanced by estradiol-esters present in HDL [89]; however, there were no sex-differences in macrophage to HDL efflux relative to men [90]. In premenopausal women, the concentration of estrogen in plasma is not associated with cholesterol efflux capacity [90]. In premenopausal women with polycystic ovary syndrome (PCOS), which is a state of reduced estrogen and increased androgens, cholesterol efflux capacity is reduced [91]. The estrogen deficiency of menopause, however, increases the cholesterol efflux capacity of HDL relative to premenopausal women, likely because of increased VLDL-TG levels after menopause [92]. In postmenopausal women, hormone treatment with CE-progestin increases cholesterol efflux capacity of HDL [93]. Thus, estrogen signaling pathways have been shown to have some effects on cholesterol efflux capacity, but there is not a consistent relationship between estrogen enhancing or impairing this initial step in RCT based on the literature in humans.

Estrogen signaling pathways have a more established role promoting the hepatic steps of RCT. Liver estrogen signaling through ERα has been shown to regulate hepatic cholesterol uptake and the efflux capacity of HDL from macrophages during the proestrus period when estrogen levels are high [77]. Female mice have increased total-body RCT compared to males fed a western diet [94]. In this study, liver deletion of ERα impaired total body RCT in female mice, suggesting that liver ERα is required for females to enhance total body RCT [94]. Estradiol and PPT treatment of mice both promote liver secretion of cholesterol into bile, a process that is prevented by concurrent treatment with an ERα antagonist [95]. The role of sex and estrogen on later stages in RCT is not well studied in humans.

8. Hormone treatment and the risk of cardiovascular disease in postmenopausal women

Despite decades of studies, treatment approaches with estrogen formulations in women after menopause have largely failed to recapitulate the protective physiology of a true replacement strategy. The results of treatments with estrogens with regard to glucose, lipids, and cardiovascular risk have varied based on the formulation of estrogen used, pairing with progestin, and route of delivery (reviewed in [96]). Two of the early randomized controlled trials of hormone treatment were the Women's Health Initiative (WHI) and the Heart and Estrogen/Progestin Replacement Study (HERS) [97], [98]. For both studies, hormone treatment consisted of conjugated estrogens plus a progestin if the women had an intact uterus. After 6.8 years of follow-up the HERS trial showed improvement in blood cholesterol levels and diabetes risk factors, but no improvement in cardiovascular disease [99]. After 5.6 years of follow-up in the WHI trial cardiovascular risk was higher in those with hormone treatment [98]. In the WHI trial where hormone treatment was largely initiated late after natural menopause, ASCVD risk was higher in women treated with hormones over 10 years after the onset of menopause. This effect led to the “timing hypothesis,” which suggests that hormone treatment is most beneficial if initiated soon after menopause, and potentially harmful if initiated late (>10 years) in menopause.

The timing hypothesis was tested in the Early versus Late Intervention Trial with Estradiol (ELITE) study, which randomized postmenopausal women to placebo or oral estradiol plus vaginal progesterone for 10 days per cycle [100]. Women were stratified into two groups –early menopause if menopause occurred in the last 6 years, and late menopause if menopause occurred at least 10 years prior to enrollment in the study. Estradiol treatment reduced the progression of carotid intima medial thickness (CIMT) in the early menopause group but failed to delay atherosclerosis in the late menopause group. This result supports the timing hypothesis of estrogen treatment.

Serum TG levels were increased with hormone treatment in the WHI trial. The cause of the TG-rich dyslipidemia with hormone treatment of postmenopausal women has been controversial. Variations in estrogen levels with a woman's menstrual cycle do not impact VLDL-TG or VLDL-apoB kinetics or concentrations [101]. Estrogen treatment, however, seems to increase serum TG in a manner that depends on the route and formulation. Oral delivery of micronized estradiol increased VLDL production rates by 80%, whereas transdermal estradiol had no effect on VLDL production rates in this study [102]. Another study with oral ethinyl estradiol increased VLDL apoB production over 100% [103], which is a similar result found to earlier studies with conjugated equine estrogens [104]. Progestins oppose the effect of estrogens by promoting VLDL clearance in both humans and animals ([105], [106] and reviewed in [38]). Transdermal preparations of estradiol have less effects on lowering LDL cholesterol and increasing HDL cholesterol [107], [108], [109], [110], [111] and do not seem to increase plasma TGs when compared to oral estrogen formulations [107], [108], [109], [110], [111], [112]. In fact, most studies demonstrate that transdermal estradiol reduces plasma TGs [107], [108], [111]. In a study examining VLDL-TG kinetics transdermal estradiol had no effect on VLDL-TG production but promoted VLDL-TG clearance [113]. The larger effect of oral estrogens on VLDL-TG production suggest that the liver is the primary organ responsible for hypertriglyceridemia with treatment.

9. Are male sex hormones mediators of increased ASCVD risk in men?

Testosterone has been hypothesized to contribute to a man's increased risk of ASCVD. The hypothesis that high testosterone increases ASCVD risk in men is controversial for several reasons. Firstly, the majority of cross-sectional studies examining the relationship between testosterone levels and ASCVD support an inverse relationship between testosterone and risk of cardiovascular disease [114], [115], [116], [117], [118], [119]. Certain studies, however, support a neutral [120], [121], [122], [123], positive or J-curve [124] relationship between testosterone and cardiovascular disease. In a meta-analysis of testosterone association with cardiovascular disease, testosterone correlated inversely with cardiovascular disease only when men above age 70 were included in the analysis [125]. This suggests that an age-related decline in testosterone [115] may be responsible for the inverse relationship between testosterone levels and risk of cardiovascular disease. Secondly, studies of testosterone deprivation show increased risk of cardiovascular disease [126], [127], [128]. This suggests that low testosterone increases cardiovascular disease risk. Thirdly, studies of testosterone therapy have different effects on risk of cardiovascular disease depending on testosterone status prior to treatment. For example, in hypogonadal men, testosterone treatment reduces risk of cardiovascular disease in men [129], [130]. In normal men, testosterone therapy seems to increase risk of cardiovascular disease in randomized controlled trials [131], [132], [133].

The unexpected conclusion that testosterone lowers risk of cardiovascular disease in men may partly be explained by the impact of low testosterone on risk of metabolic syndrome. Metabolic syndrome is associated with higher risk of cardiovascular disease [134], which may be an important confounder in understanding the cardiovascular disease risk associated with testosterone levels. Reduced testosterone levels are associated with increased fasting glucose, fasting insulin, and type 2 diabetes [135], [136], [137], [138], [139]. Testosterone treatment in men with low testosterone improves insulin sensitivity, reduces glucose and insulin, and reduces risk of type 2 diabetes [140], [141], [142]. In addition, testosterone treatment in hypogonadal men reduces obesity and improves lean muscle mass, both of which would contribute to reducing risk of type 2 diabetes [140]. Thus, the “benefit” of testosterone may be related more to improvements in muscle glucose metabolism and insulin sensitivity than improvements in cardiovascular disease, especially when considering the impact of testosterone in hypogonadal men.

The development of Androgen Receptor (AR) knockout (ARKO) models has allowed for a more precise definition of the contribution of androgen signaling to sex-differences in lipid metabolism and atherosclerosis. Mice with a global AR knockout (ARKO) had worse atherosclerosis relative to controls on an Apolipoprotein E knockout (ApoE−/−) background [143], [144]. Global ARKO mice had increased weight gain, increased plasma cholesterol and TGs, increased liver TG content, and impaired glucose metabolism. Additionally, 5α-dihydrotestosterone, a non-aromatizable AR agonist, reduced atherosclerosis, obesity, plasma cholesterol, and plasma insulin liver TG content and reduced atherosclerosis. These data suggest that AR signaling reduces atherosclerosis and improves glucose and lipid risk factors for cardiovascular disease. While these studies are informative about androgen signaling, they don't provide clear mechanisms of the increased risk of cardiovascular disease seen in men.

10. Sex-specific considerations for the treatment of dyslipidemia and ASCVD

The historically lower risk of cardiovascular disease in women than in men, in some ways has biased physicians and scientists from understanding cardiovascular disease and its treatment in women. In women, death from cardiovascular causes is higher than from breast and ovarian cancers combined. Although most studies suggest that cholesterol lowering approaches with statins are equally effective in men and women, trials from the major statin studies had only 15–30% women [145]. The study with the largest percentage of women was the MEGA study with pravastatin which had 68% women, for which pravastatin did not reach statistical significance compared to diet alone [146]. PCSK9 degrades the LDLR, increasing blood LDL-C levels. In men but not in women, PCSK9 levels correlate with LDL cholesterol [147]. Deletion of PCSK9 in mice produces dramatic upregulation of LDLR only in male mice [148]. However, it is not conclusive if treatment with PCSK9 inhibitors aliorocumab and evolocumab is equally effective in men and women [149]. In a study looking at short-term mortality after myocardial infarction, among patients less than 50 years of age, the mortality rate for the women was more than twice that for the men [150], [151]. Women are less likely to be treated with reperfusion therapy and beta blockers [150]. After stent placement women have increased risk of subsequent major adverse cardiovascular events compared to men [152], [153]. Compared to men, women receive less cholesterol screening and fewer lipid-lowering therapies. Even for LDL of 160 women are less likely to be treated with a high-potency statin [154], [155]. Women are more likely to have myopathy and discontinue statin therapy, a biology likely related to sex-differences in statin metabolism. Older women may have less benefit from angiotensin converting-enzyme inhibitor use than men [156], and potentially less benefit from low-dose aspirin for primary prevention [157], although guidelines still support the use of both in clinically-indicated populations. Thus, in addition to understanding the physiology of sex-difference in lipid metabolism, it is also imperative to understand potential sex-differences in the efficacy of treatment and prevention strategies for ASCVD.

11. Conclusions and future directions

There are major sex differences in lipid and lipoprotein metabolism that contribute to sex-differences in ASCVD risk. The mechanisms for these sex differences are complex and involve hormonal effects that are distributed across tissues, as well as effects mediated by genes on the X-chromosome that escape the process of X-inactivation. It is important to note that the historically lower rates of ASCVD in women have created a false perception that ASCVD is less important in women than in men, which has certainly hindered both clinical and basic science discoveries. The British Heart Foundation states that the number of women living with ASCVD is now roughly the same as the number of men [158]. The lower perceived risk in women is unfortunate, because ASCVD is the major cause of death in women. This oversight is beginning to be corrected. We also don't understand well the interaction between diabetes and ASCVD in women, which may have a heightened contribution to risk compared to men [4], [5], [6]. This is of particular importance in certain groups of high-risk women including individuals of African and Hispanic descent for whom risk of diabetes is very high. Lastly, sex hormone treatment of older adults after endogenous hormone levels decline holds to promise to improve many aspects of cardiometabolic health, but physicians are largely unable to recapitulate the physiologic benefits of endogenous estrogens or androgens with treatment approaches in older adults. Much remains to be learned about mechanisms for these sex-differences. Gaining this knowledge would allow us to therapeutically target the relevant protective pathways as well as aid our ability to physiologically replace sex-hormones in older adults.

Funding sources

The Department of Veterans Affairs (BX002223) and NIH (R01DK109102) provided support to JMS. BP is supported by the Vanderbilt Medical Scientist Training Program (T32GM07347) and (F30DK104514). RH is supported by DK42266, DK4688, DK61668, and DK089309.

Conflicts of interest

None declared.

References

  • 1.Wilmot K.A., O'Flaherty M., Capewell S., Ford E.S., Vaccarino V. Coronary heart disease mortality declines in the United States from 1979 through 2011: evidence for stagnation in young adults, especially women. Circulation. 2015;132:997–1002. doi: 10.1161/CIRCULATIONAHA.115.015293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Benjamin E.J., Blaha M.J., Chiuve S.E., Cushman M., Das S.R., Deo R. Heart disease and stroke statistics—2017 update: a report from the American Heart Association. Circulation. 2017;137:e1–e458. doi: 10.1161/CIR.0000000000000485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Towfighi A., Zheng L., Ovbiagele B. Sex-specific trends in midlife coronary heart disease risk and prevalence. Archives of Internal Medicine. 2009;169:1762–1766. doi: 10.1001/archinternmed.2009.318. [DOI] [PubMed] [Google Scholar]
  • 4.Hu G., Jousilahti P., Qiao Q., Katoh S., Tuomilehto J. Sex differences in cardiovascular and total mortality among diabetic and non-diabetic individuals with or without history of myocardial infarction. Diabetologia. 2005;48:856–861. doi: 10.1007/s00125-005-1730-6. [DOI] [PubMed] [Google Scholar]
  • 5.Regensteiner J.G., Golden S., Huebschmann A.G., Barrett-Connor E., Chang A.Y., Chyun D. Sex differences in the cardiovascular consequences of diabetes mellitus: a scientific statement from the American heart association. Circulation. 2015;132:2424–2447. doi: 10.1161/CIR.0000000000000343. [DOI] [PubMed] [Google Scholar]
  • 6.Barrett-Connor E.L., Cohn B.A., Wingard D.L., Edelstein S.L. Why is diabetes mellitus a stronger risk factor for fatal ischemic heart disease in women than in men? The Rancho Bernardo Study. Journal of the American Medical Association. 1991;265:627–631. [PubMed] [Google Scholar]
  • 7.Matthews J., Celius T., Halgren R., Zacharewski T. Differential estrogen receptor binding of estrogenic substances: a species comparison. The Journal of Steroid Biochemistry and Molecular Biology. 2000;74:223–234. doi: 10.1016/s0960-0760(00)00126-6. [DOI] [PubMed] [Google Scholar]
  • 8.Osborne C.K., Schiff R. Estrogen-receptor biology: continuing progress and therapeutic implications. Journal of Clinical Oncology. 2005;23:1616–1622. doi: 10.1200/JCO.2005.10.036. [DOI] [PubMed] [Google Scholar]
  • 9.Zhang Y., Klein K., Sugathan A., Nassery N., Dombkowski A., Zanger U.M. Transcriptional profiling of human liver identifies sex-biased genes associated with polygenic dyslipidemia and coronary artery disease. PLoS One. 2011;6:e23506. doi: 10.1371/journal.pone.0023506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Parks B.W., Sallam T., Mehrabian M., Psychogios N., Hui S.T., Norheim F. Genetic architecture of insulin resistance in the mouse. Cell Metabolism. 2015;21:334–346. doi: 10.1016/j.cmet.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gao H., Falt S., Sandelin A., Gustafsson J.A., Dahlman-Wright K. Genome-wide identification of estrogen receptor alpha-binding sites in mouse liver. Molecular Endocrinology. 2008;22:10–22. doi: 10.1210/me.2007-0121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Villa A., Della Torre S., Stell A., Cook J., Brown M., Maggi A. Tetradian oscillation of estrogen receptor alpha is necessary to prevent liver lipid deposition. Proceedings of the National Academy of Sciences of the U S A. 2012;109:11806–11811. doi: 10.1073/pnas.1205797109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Park C.J., Zhao Z., Glidewell-Kenney C., Lazic M., Chambon P., Krust A. Genetic rescue of nonclassical ERalpha signaling normalizes energy balance in obese Eralpha-null mutant mice. Journal of Clinical Investigation. 2011;121:604–612. doi: 10.1172/JCI41702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bjornstrom L., Sjoberg M. Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on target genes. Molecular Endocrinology. 2005;19:833–842. doi: 10.1210/me.2004-0486. [DOI] [PubMed] [Google Scholar]
  • 15.Marino M., Galluzzo P., Ascenzi P. Estrogen signaling multiple pathways to impact gene transcription. Current Genomics. 2006;7:497–508. doi: 10.2174/138920206779315737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Levin E.R. Plasma membrane estrogen receptors. Trends in Endocrinology and Metabolism. 2009;20:477–482. doi: 10.1016/j.tem.2009.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Pedram A., Razandi M., O'Mahony F., Harvey H., Harvey B.J., Levin E.R. Estrogen reduces lipid content in the liver exclusively from membrane receptor signaling. Science Signaling. 2013;6:ra36. doi: 10.1126/scisignal.2004013. [DOI] [PubMed] [Google Scholar]
  • 18.Sharma G., Mauvais-Jarvis F., Prossnitz E.R. Roles of G protein-coupled estrogen receptor GPER in metabolic regulation. The Journal of Steroid Biochemistry and Molecular Biology. 2018 Feb;176:31–37. doi: 10.1016/j.jsbmb.2017.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nilsson B.O., Olde B., Leeb-Lundberg L.M. G protein-coupled oestrogen receptor 1 (GPER1)/GPR30: a new player in cardiovascular and metabolic oestrogenic signalling. British Journal of Pharmacology. 2011;163:1131–1139. doi: 10.1111/j.1476-5381.2011.01235.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Owman C., Blay P., Nilsson C., Lolait S.J. Cloning of human cDNA encoding a novel heptahelix receptor expressed in Burkitt's lymphoma and widely distributed in brain and peripheral tissues. Biochemical and Biophysical Research Communications. 1996;228:285–292. doi: 10.1006/bbrc.1996.1654. [DOI] [PubMed] [Google Scholar]
  • 21.Meyer M.R., Fredette N.C., Howard T.A., Hu C., Ramesh C., Daniel C. G protein-coupled estrogen receptor protects from atherosclerosis. Scientific Reports. 2014;4:7564. doi: 10.1038/srep07564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gelmann E.P. Molecular biology of the androgen receptor. Journal of Clinical Oncology. 2002;20:3001–3015. doi: 10.1200/JCO.2002.10.018. [DOI] [PubMed] [Google Scholar]
  • 23.Heinlein C.A., Chang C. Androgen receptor (AR) coregulators: an overview. Endocrine Reviews. 2002;23:175–200. doi: 10.1210/edrv.23.2.0460. [DOI] [PubMed] [Google Scholar]
  • 24.Liao R.S., Ma S., Miao L., Li R., Yin Y., Raj G.V. Androgen receptor-mediated non-genomic regulation of prostate cancer cell proliferation. Translational Andrology and Urology. 2013;2:187–196. doi: 10.3978/j.issn.2223-4683.2013.09.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gregory C.W., Fei X., Ponguta L.A., He B., Bill H.M., French F.S. Epidermal growth factor increases coactivation of the androgen receptor in recurrent prostate cancer. Journal of Biological Chemistry. 2004;279:7119–7130. doi: 10.1074/jbc.M307649200. [DOI] [PubMed] [Google Scholar]
  • 26.Vague J. La différenciation sexuelle; facteur déterminant des formes de l'obésité. Presse Medicale. 1947;55:339. [PubMed] [Google Scholar]
  • 27.Canoy D., Boekholdt S.M., Wareham N., Luben R., Welch A., Bingham S. Body fat distribution and risk of coronary heart disease in men and women in the European Prospective Investigation into Cancer and Nutrition in Norfolk cohort: a population-based prospective study. Circulation. 2007;116:2933–2943. doi: 10.1161/CIRCULATIONAHA.106.673756. [DOI] [PubMed] [Google Scholar]
  • 28.Yusuf S., Hawken S., Ounpuu S., Bautista L., Franzosi M.G., Commerford P. Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: a case-control study. Lancet. 2005;366:1640–1649. doi: 10.1016/S0140-6736(05)67663-5. [DOI] [PubMed] [Google Scholar]
  • 29.Fried S.K., Lee M.J., Karastergiou K. Shaping fat distribution: new insights into the molecular determinants of depot- and sex-dependent adipose biology. Obesity (Silver Spring) 2015;23:1345–1352. doi: 10.1002/oby.21133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.de Koning L., Merchant A.T., Pogue J., Anand S.S. Waist circumference and waist-to-hip ratio as predictors of cardiovascular events: meta-regression analysis of prospective studies. European Heart Journal. 2007;28:850–856. doi: 10.1093/eurheartj/ehm026. [DOI] [PubMed] [Google Scholar]
  • 31.Fried S.K., Kral J.G. Sex differences in regional distribution of fat cell size and lipoprotein lipase activity in morbidly obese patients. International Journal of Obesity. 1987;11:129–140. [PubMed] [Google Scholar]
  • 32.Rehrer C.W., Karimpour-Fard A., Hernandez T.L., Law C.K., Stob N.R., Hunter L.E. Regional differences in subcutaneous adipose tissue gene expression. Obesity (Silver Spring) 2012;20:2168–2173. doi: 10.1038/oby.2012.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dowling H.J., Fried S.K., Pi-Sunyer F.X. Insulin resistance in adipocytes of obese women: effects of body fat distribution and race. Metabolism. 1995;44:987–995. doi: 10.1016/0026-0495(95)90094-2. [DOI] [PubMed] [Google Scholar]
  • 34.Despres J.P., Couillard C., Gagnon J., Bergeron J., Leon A.S., Rao D.C. Race, visceral adipose tissue, plasma lipids, and lipoprotein lipase activity in men and women: the Health, Risk Factors, Exercise Training, and Genetics (HERITAGE) family study. Arteriosclerosis, Thrombosis, and Vascular Biology. 2000;20:1932–1938. doi: 10.1161/01.atv.20.8.1932. [DOI] [PubMed] [Google Scholar]
  • 35.Santosa S., Jensen M.D. Effects of male hypogonadism on regional adipose tissue fatty acid storage and lipogenic proteins. PLoS One. 2012;7:e31473. doi: 10.1371/journal.pone.0031473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Svendsen O.L., Hassager C., Christiansen C. Age- and menopause-associated variations in body composition and fat distribution in healthy women as measured by dual-energy X-ray absorptiometry. Metabolism. 1995;44:369–373. doi: 10.1016/0026-0495(95)90168-x. [DOI] [PubMed] [Google Scholar]
  • 37.Lapid K., Lim A., Clegg D.J., Zeve D., Graff J.M. Oestrogen signalling in white adipose progenitor cells inhibits differentiation into brown adipose and smooth muscle cells. Nature Communications. 2014;5:5196. doi: 10.1038/ncomms6196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Magkos F., Mittendorfer B. Gender differences in lipid metabolism and the effect of obesity. Obstetrics & Gynecology Clinics of North America. 2009;36:245–265. doi: 10.1016/j.ogc.2009.03.001. vii. [DOI] [PubMed] [Google Scholar]
  • 39.Macotela Y., Boucher J., Tran T.T., Kahn C.R. Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes. 2009;58:803–812. doi: 10.2337/db08-1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Xu Y., Nedungadi T.P., Zhu L., Sobhani N., Irani B.G., Davis K.E. Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction. Cell Metabolism. 2011;14:453–465. doi: 10.1016/j.cmet.2011.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Frank A., Brown L.M., Clegg D.J. The role of hypothalamic estrogen receptors in metabolic regulation. Frontiers in Neuroendocrinology. 2014;35:550–557. doi: 10.1016/j.yfrne.2014.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wang H., Wang Y., Taussig M.D., Eckel R.H. Sex differences in obesity development in pair-fed neuronal lipoprotein lipase deficient mice. Molecular Metabolism. 2016;5:1025–1032. doi: 10.1016/j.molmet.2016.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shungin D., Winkler T.W., Croteau-Chonka D.C., Ferreira T., Locke A.E., Magi R. New genetic loci link adipose and insulin biology to body fat distribution. Nature. 2015;518:187–196. doi: 10.1038/nature14132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Heid I.M., Jackson A.U., Randall J.C., Winkler T.W., Qi L., Steinthorsdottir V. Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nature Genetics. 2010;42:949–960. doi: 10.1038/ng.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zore T., Palafox M., Reue K. Sex differences in obesity, lipid metabolism, and inflammation–A role for the sex chromosomes? Molecular Metabolism. 2018;15:35–44. doi: 10.1016/j.molmet.2018.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Nielsen S., Guo Z., Johnson C.M., Hensrud D.D., Jensen M.D. Splanchnic lipolysis in human obesity. Journal of Clinical Investigation. 2004;113:1582–1588. doi: 10.1172/JCI21047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Reaven G.M., Bernstein R.M. Effect of obesity on the relationship between very low density lipoprotein production rate and plasma triglyceride concentration in normal and hypertriglyceridemic subjects. Metabolism. 1978;27:1047–1054. doi: 10.1016/0026-0495(78)90150-6. [DOI] [PubMed] [Google Scholar]
  • 48.Mittendorfer B., Patterson B.W., Klein S. Effect of sex and obesity on basal VLDL-triacylglycerol kinetics. American Journal of Clinical Nutrition. 2003;77:573–579. doi: 10.1093/ajcn/77.3.573. [DOI] [PubMed] [Google Scholar]
  • 49.Frias J.P., Macaraeg G.B., Ofrecio J., Yu J.G., Olefsky J.M., Kruszynska Y.T. Decreased susceptibility to fatty acid-induced peripheral tissue insulin resistance in women. Diabetes. 2001;50:1344–1350. doi: 10.2337/diabetes.50.6.1344. [DOI] [PubMed] [Google Scholar]
  • 50.Clegg D., Hevener A.L., Moreau K.L., Morselli E., Criollo A., Van Pelt R.E. Sex hormones and cardiometabolic health: role of estrogen and estrogen receptors. Endocrinology. 2017;158:1095–1105. doi: 10.1210/en.2016-1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ribas V., Drew B.G., Zhou Z., Phun J., Kalajian N.Y., Soleymani T. Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females. Science Translational Medicine. 2016;8 doi: 10.1126/scitranslmed.aad3815. 334ra354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Magkos F., Patterson B.W., Mohammed B.S., Klein S., Mittendorfer B. Women produce fewer but triglyceride-richer very low-density lipoproteins than men. The Journal of Clinical Endocrinology & Metabolism. 2007;92:1311–1318. doi: 10.1210/jc.2006-2215. [DOI] [PubMed] [Google Scholar]
  • 53.Hodson L., Banerjee R., Rial B., Arlt W., Adiels M., Boren J. Menopausal status and abdominal obesity are significant determinants of hepatic lipid metabolism in women. Journal of the American Heart Association. 2015;4:e002258. doi: 10.1161/JAHA.115.002258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Matthan N.R., Jalbert S.M., Barrett P.H., Dolnikowski G.G., Schaefer E.J., Lichtenstein A.H. Gender-specific differences in the kinetics of nonfasting TRL, IDL, and LDL apolipoprotein B-100 in men and premenopausal women. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28:1838–1843. doi: 10.1161/ATVBAHA.108.163931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mittendorfer B., Magkos F., Fabbrini E., Mohammed B.S., Klein S. Relationship between body fat mass and free fatty acid kinetics in men and women. Obesity (Silver Spring) 2009;17:1872–1877. doi: 10.1038/oby.2009.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Karpe F., Dickmann J.R., Frayn K.N. Fatty acids, obesity, and insulin resistance: time for a reevaluation. Diabetes. 2011;60:2441. doi: 10.2337/db11-0425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hevener A., Reichart D., Janez A., Olefsky J. Female rats do not exhibit free fatty acid-induced insulin resistance. Diabetes. 2002;51:1907–1912. doi: 10.2337/diabetes.51.6.1907. [DOI] [PubMed] [Google Scholar]
  • 58.Perreault L., Bergman B.C., Hunerdosse D.M., Eckel R.H. Altered intramuscular lipid metabolism relates to diminished insulin action in men, but not women, in progression to diabetes. Obesity (Silver Spring) 2010;18:2093–2100. doi: 10.1038/oby.2010.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Votruba S.B., Jensen M.D. Sex-specific differences in leg fat uptake are revealed with a high-fat meal. American Journal of Physiology. Endocrinology and Metabolism. 2006;291:E1115–E1123. doi: 10.1152/ajpendo.00196.2006. [DOI] [PubMed] [Google Scholar]
  • 60.Santosa S., Hensrud D.D., Votruba S.B., Jensen M.D. The influence of sex and obesity phenotype on meal fatty acid metabolism before and after weight loss. American Journal of Clinical Nutrition. 2008;88:1134–1141. doi: 10.1093/ajcn/88.4.1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yost T.J., Jensen D.R., Haugen B.R., Eckel R.H. Effect of dietary macronutrient composition on tissue-specific lipoprotein lipase activity and insulin action in normal-weight subjects. American Journal of Clinical Nutrition. 1998;68:296–302. doi: 10.1093/ajcn/68.2.296. [DOI] [PubMed] [Google Scholar]
  • 62.Castelli W.P. The triglyceride issue: a view from Framingham. American Heart Journal. 1986;112:432–437. doi: 10.1016/0002-8703(86)90296-6. [DOI] [PubMed] [Google Scholar]
  • 63.Serum triglycerides as a risk factor for cardiovascular diseases in the Asia-Pacific region. Circulation. 2004;110:2678. doi: 10.1161/01.CIR.0000145615.33955.83. [DOI] [PubMed] [Google Scholar]
  • 64.Hokanson J.E., Austin M.A. Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies. Journal of Cardiovascular Risk. 1996;3:213–219. [PubMed] [Google Scholar]
  • 65.Nishino M., Hayakawa K., Nakamura Y., Morimoto T., Mukaihara S. Effects of tamoxifen on hepatic fat content and the development of hepatic steatosis in patients with breast cancer: high frequency of involvement and rapid reversal after completion of tamoxifen therapy. American Journal of Roentgenology. 2003;180:129–134. doi: 10.2214/ajr.180.1.1800129. [DOI] [PubMed] [Google Scholar]
  • 66.Murata Y., Ogawa Y., Saibara T., Nishioka A., Fujiwara Y., Fukumoto M. Unrecognized hepatic steatosis and non-alcoholic steatohepatitis in adjuvant tamoxifen for breast cancer patients. Oncology Reports. 2000;7:1299–1304. doi: 10.3892/or.7.6.1299. [DOI] [PubMed] [Google Scholar]
  • 67.Ryu S., Suh B.S., Chang Y., Kwon M.J., Yun K.E., Jung H.S. Menopausal stages and non-alcoholic fatty liver disease in middle-aged women. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2015;190:65–70. doi: 10.1016/j.ejogrb.2015.04.017. [DOI] [PubMed] [Google Scholar]
  • 68.Yang J.D., Abdelmalek M.F., Pang H., Guy C.D., Smith A.D., Diehl A.M. Gender and menopause impact severity of fibrosis among patients with nonalcoholic steatohepatitis. Hepatology. 2014;59:1406–1414. doi: 10.1002/hep.26761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Matsuo K., Gualtieri M.R., Cahoon S.S., Jung C.E., Paulson R.J., Shoupe D. Surgical menopause and increased risk of nonalcoholic fatty liver disease in endometrial cancer. Menopause. 2016;23:189–196. doi: 10.1097/GME.0000000000000500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cote I., Yasari S., Pighon A., Barsalani R., Rabasa-Lhoret R., Prud'homme D. Liver fat accumulation may be dissociated from adiposity gain in ovariectomized rats. Climacteric. 2012;15:594–601. doi: 10.3109/13697137.2011.637650. [DOI] [PubMed] [Google Scholar]
  • 71.de Oliveira M.C., Gilglioni E.H., de Boer B.A., Runge J.H., de Waart D.R., Salgueiro C.L. Bile acid receptor agonists INT747 and INT777 decrease oestrogen deficiency-related postmenopausal obesity and hepatic steatosis in mice. Biochimica et Biophysica Acta. 2016;1862:2054–2062. doi: 10.1016/j.bbadis.2016.07.012. [DOI] [PubMed] [Google Scholar]
  • 72.Paquette A., Shinoda M., Rabasa Lhoret R., Prud'homme D., Lavoie J.M. Time course of liver lipid infiltration in ovariectomized rats: impact of a high-fat diet. Maturitas. 2007;58:182–190. doi: 10.1016/j.maturitas.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 73.Romero-Aleshire M.J., Diamond-Stanic M.K., Hasty A.H., Hoyer P.B., Brooks H.L. Loss of ovarian function in the VCD mouse-model of menopause leads to insulin resistance and a rapid progression into the metabolic syndrome. American Journal of Physiology – Regulatory, Integrative and Comparative Physiology. 2009;297:R587–R592. doi: 10.1152/ajpregu.90762.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Palmisano B.T., Le T.D., Zhu L., Lee Y.K., Stafford J.M. Cholesteryl ester transfer protein alters liver and plasma triglyceride metabolism through two liver networks in female mice. The Journal of Lipid Research. 2016;57:1541–1551. doi: 10.1194/jlr.M069013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhu L., Brown W.C., Cai Q., Krust A., Chambon P., McGuinness O.P. Estrogen treatment after ovariectomy protects against fatty liver and may improve pathway-selective insulin resistance. Diabetes. 2013;62:424–434. doi: 10.2337/db11-1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bryzgalova G., Gao H., Ahren B., Zierath J.R., Galuska D., Steiler T.L. Evidence that oestrogen receptor-alpha plays an important role in the regulation of glucose homeostasis in mice: insulin sensitivity in the liver. Diabetologia. 2006;49:588–597. doi: 10.1007/s00125-005-0105-3. [DOI] [PubMed] [Google Scholar]
  • 77.Della Torre S., Mitro N., Fontana R., Gomaraschi M., Favari E., Recordati C. An essential role for liver ERalpha in coupling hepatic metabolism to the reproductive cycle. Cell Reports. 2016;15:360–371. doi: 10.1016/j.celrep.2016.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wang X., Lu Y., Wang E., Zhang Z., Xiong X., Zhang H. Hepatic estrogen receptor alpha improves hepatosteatosis through upregulation of small heterodimer partner. Journal of Hepatology. 2015;63:183–190. doi: 10.1016/j.jhep.2015.02.029. [DOI] [PubMed] [Google Scholar]
  • 79.Zhang Z.C., Liu Y., Xiao L.L., Li S.F., Jiang J.H., Zhao Y. Upregulation of miR-125b by estrogen protects against non-alcoholic fatty liver in female mice. Journal of Hepatology. 2015;63:1466–1475. doi: 10.1016/j.jhep.2015.07.037. [DOI] [PubMed] [Google Scholar]
  • 80.Gao H., Bryzgalova G., Hedman E., Khan A., Efendic S., Gustafsson J.A. Long-term administration of estradiol decreases expression of hepatic lipogenic genes and improves insulin sensitivity in ob/ob mice: a possible mechanism is through direct regulation of signal transducer and activator of transcription 3. Molecular Endocrinology. 2006;20:1287–1299. doi: 10.1210/me.2006-0012. [DOI] [PubMed] [Google Scholar]
  • 81.Cole L.K., Jacobs R.L., Vance D.E. Tamoxifen induces triacylglycerol accumulation in the mouse liver by activation of fatty acid synthesis. Hepatology. 2010;52:1258–1265. doi: 10.1002/hep.23813. [DOI] [PubMed] [Google Scholar]
  • 82.Zhang H., Liu Y., Wang L., Li Z., Zhang H., Wu J. Differential effects of estrogen/androgen on the prevention of nonalcoholic fatty liver disease in the male rat. The Journal of Lipid Research. 2013;54:345–357. doi: 10.1194/jlr.M028969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhu L., Martinez M.N., Emfinger C.H., Palmisano B.T., Stafford J.M. Estrogen signaling prevents diet-induced hepatic insulin resistance in male mice with obesity. American Journal of Physiology. Endocrinology and Metabolism. 2014;306:E1188–E1197. doi: 10.1152/ajpendo.00579.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kim J.H., Meyers M.S., Khuder S.S., Abdallah S.L., Muturi H.T., Russo L. Tissue-selective estrogen complexes with bazedoxifene prevent metabolic dysfunction in female mice. Molecular Metabolism. 2014;3:177–190. doi: 10.1016/j.molmet.2013.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Camporez J.P., Jornayvaz F.R., Lee H.Y., Kanda S., Guigni B.A., Kahn M. Cellular mechanism by which estradiol protects female ovariectomized mice from high-fat diet-induced hepatic and muscle insulin resistance. Endocrinology. 2013;154:1021–1028. doi: 10.1210/en.2012-1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Freedman D.S., Otvos J.D., Jeyarajah E.J., Shalaurova I., Cupples L.A., Parise H. Sex and age differences in lipoprotein subclasses measured by nuclear magnetic resonance spectroscopy: the Framingham Study. Clinical Chemistry. 2004;50:1189–1200. doi: 10.1373/clinchem.2004.032763. [DOI] [PubMed] [Google Scholar]
  • 87.Wilmot K.A., O'Flaherty M., Capewell S., Ford E.S., Vaccarino V. Coronary heart disease mortality declines in the United States from 1979 through 2011. Circulation. 2015;132:997. doi: 10.1161/CIRCULATIONAHA.115.015293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Rosenson R.S., Brewer H.B., Jr., Davidson W.S., Fayad Z.A., Fuster V., Goldstein J. Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport. Circulation. 2012;125:1905–1919. doi: 10.1161/CIRCULATIONAHA.111.066589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Badeau R.M., Metso J., Wahala K., Tikkanen M.J., Jauhiainen M. Human macrophage cholesterol efflux potential is enhanced by HDL-associated 17beta-estradiol fatty acyl esters. The Journal of Steroid Biochemistry and Molecular Biology. 2009;116:44–49. doi: 10.1016/j.jsbmb.2009.04.008. [DOI] [PubMed] [Google Scholar]
  • 90.Badeau R.M., Metso J., Kovanen P.T., Lee-Rueckert M., Tikkanen M.J., Jauhiainen M. The impact of gender and serum estradiol levels on HDL-mediated reverse cholesterol transport. European Journal of Clinical Investigation. 2013;43:317–323. doi: 10.1111/eci.12044. [DOI] [PubMed] [Google Scholar]
  • 91.Roe A., Hillman J., Butts S., Smith M., Rader D., Playford M. Decreased cholesterol efflux capacity and atherogenic lipid profile in young women with PCOS. The Journal of Clinical Endocrinology & Metabolism. 2014;99:E841–E847. doi: 10.1210/jc.2013-3918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.El Khoudary S.R., Hutchins P.M., Matthews K.A., Brooks M.M., Orchard T.J., Ronsein G.E. Cholesterol efflux capacity and subclasses of HDL particles in healthy women transitioning through menopause. The Journal of Clinical Endocrinology & Metabolism. 2016;101:3419–3428. doi: 10.1210/jc.2016-2144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ulloa N., Arteaga E., Bustos P., Duran-Sandoval D., Schulze K., Castro G. Sequential estrogen-progestin replacement therapy in healthy postmenopausal women: effects on cholesterol efflux capacity and key proteins regulating high-density lipoprotein levels. Metabolism. 2002;51:1410–1417. doi: 10.1053/meta.2002.35580. [DOI] [PubMed] [Google Scholar]
  • 94.Zhu L., Shi J., Luu T.N., Neuman J.C., Trefts E., Yu S. Hepatocyte estrogen receptor alpha mediates estrogen action to promote reverse cholesterol transport during Western-type diet feeding. Molecular Metabolism. 2018;8:106–116. doi: 10.1016/j.molmet.2017.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang H.H., Afdhal N.H., Wang D.Q. Estrogen receptor alpha, but not beta, plays a major role in 17beta-estradiol-induced murine cholesterol gallstones. Gastroenterology. 2004;127:239–249. doi: 10.1053/j.gastro.2004.03.059. [DOI] [PubMed] [Google Scholar]
  • 96.Palmisano B.T., Zhu L., Stafford J.M. Role of estrogens in the regulation of liver lipid metabolism. In: Mauvais-Jarvis F., editor. Sex and gender factors affecting metabolic homeostasis, diabetes and obesity. Springer International Publishing; Cham: 2017. pp. 227–256. [Google Scholar]
  • 97.Hulley S., Grady D., Bush T., Furberg C., Herrington D., Riggs B. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and estrogen/progestin replacement study (HERS) research group. Journal of the American Medical Association. 1998;280:605–613. doi: 10.1001/jama.280.7.605. [DOI] [PubMed] [Google Scholar]
  • 98.Manson J.E., Hsia J., Johnson K.C., Rossouw J.E., Assaf A.R., Lasser N.L. Estrogen plus progestin and the risk of coronary heart disease. New England Journal of Medicine. 2003;349:523–534. doi: 10.1056/NEJMoa030808. [DOI] [PubMed] [Google Scholar]
  • 99.Grady D., Herrington D., Bittner V., Blumenthal R., Davidson M., Hlatky M. Cardiovascular disease outcomes during 6.8 years of hormone therapy: heart and Estrogen/progestin Replacement Study follow-up (HERS II) Journal of the American Medical Association. 2002;288:49–57. doi: 10.1001/jama.288.1.49. [DOI] [PubMed] [Google Scholar]
  • 100.Hodis H.N., Mack W.J., Henderson V.W., Shoupe D., Budoff M.J., Hwang-Levine J. Vascular effects of early versus late postmenopausal treatment with estradiol. New England Journal of Medicine. 2016;374:1221–1231. doi: 10.1056/NEJMoa1505241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Magkos F., Patterson B.W., Mittendorfer B. No effect of menstrual cycle phase on basal very-low-density lipoprotein triglyceride and apolipoprotein B-100 kinetics. American Journal of Physiology, Endocrinology and Metabolism. 2006;291:E1243–E1249. doi: 10.1152/ajpendo.00246.2006. [DOI] [PubMed] [Google Scholar]
  • 102.Walsh B.W., Schiff I., Rosner B., Greenberg L., Ravnikar V., Sacks F.M. Effects of postmenopausal estrogen replacement on the concentrations and metabolism of plasma lipoproteins. New England Journal of Medicine. 1991;325:1196–1204. doi: 10.1056/NEJM199110243251702. [DOI] [PubMed] [Google Scholar]
  • 103.Schaefer E.J., Foster D.M., Zech L.A., Lindgren F.T., Brewer H.B., Jr., Levy R.I. The effects of estrogen administration on plasma lipoprotein metabolism in premenopausal females. The Journal of Clinical Endocrinology & Metabolism. 1983;57:262–267. doi: 10.1210/jcem-57-2-262. [DOI] [PubMed] [Google Scholar]
  • 104.Glueck C.J., Fallat R.W., Scheel D. Effects of estrogenic compounds on triglyceride kinetics. Metabolism. 1975;24:537–545. doi: 10.1016/0026-0495(75)90078-5. [DOI] [PubMed] [Google Scholar]
  • 105.Kissebah A.H., Harrigan P., Wynn V. Mechanism of hypertriglyceridaemia associated with contraceptive steroids. Hormone and Metabolic Research. 1973;5:184–190. doi: 10.1055/s-0028-1093969. [DOI] [PubMed] [Google Scholar]
  • 106.Kim H.J., Kalkhoff R.K. Sex steroid influence on triglyceride metabolism. Journal of Clinical Investigation. 1975;56:888–896. doi: 10.1172/JCI108168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Baksu B., Davas I., Agar E., Akyol A., Uluocak A. Do different delivery systems of estrogen therapy influence serum lipids differently in surgically menopausal women? Journal of Obstetrics and Gynaecology Research. 2007;33:346–352. doi: 10.1111/j.1447-0756.2007.00534.x. [DOI] [PubMed] [Google Scholar]
  • 108.Chen F.P., Lee N., Soong Y.K., Huang K.E. Comparison of transdermal and oral estrogen-progestin replacement therapy: effects on cardiovascular risk factors. Menopause. 2001;8:347–352. doi: 10.1097/00042192-200109000-00009. [DOI] [PubMed] [Google Scholar]
  • 109.Sanada M., Tsuda M., Kodama I., Sakashita T., Nakagawa H., Ohama K. Substitution of transdermal estradiol during oral estrogen-progestin therapy in postmenopausal women: effects on hypertriglyceridemia. Menopause. 2004;11:331–336. doi: 10.1097/01.gme.0000094211.15096.b4. [DOI] [PubMed] [Google Scholar]
  • 110.Strandberg T.E., Ylikorkala O., Tikkanen M.J. Differing effects of oral and transdermal hormone replacement therapy on cardiovascular risk factors in healthy postmenopausal women. The American Journal of Cardiology. 2003;92:212–214. doi: 10.1016/s0002-9149(03)00542-3. [DOI] [PubMed] [Google Scholar]
  • 111.Zegura B., Guzic-Salobir B., Sebestjen M., Keber I. The effect of various menopausal hormone therapies on markers of inflammation, coagulation, fibrinolysis, lipids, and lipoproteins in healthy postmenopausal women. Menopause. 2006;13:643–650. doi: 10.1097/01.gme.0000198485.70703.7a. [DOI] [PubMed] [Google Scholar]
  • 112.O'Sullivan A.J., Crampton L.J., Freund J., Ho K.K. The route of estrogen replacement therapy confers divergent effects on substrate oxidation and body composition in postmenopausal women. Journal of Clinical Investigation. 1998;102:1035–1040. doi: 10.1172/JCI2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Smith G.I., Reeds D.N., Okunade A.L., Patterson B.W., Mittendorfer B. Systemic delivery of estradiol, but not testosterone or progesterone, alters very low density lipoprotein-triglyceride kinetics in postmenopausal women. The Journal of Clinical Endocrinology & Metabolism. 2014;99:E1306–E1310. doi: 10.1210/jc.2013-4470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Chute C.G., Baron J.A., Plymate S.R., Kiel D.P., Pavia A.T., Lozner E.C. Sex hormones and coronary artery disease. The American Journal of Medicine. 1987;83:853–859. doi: 10.1016/0002-9343(87)90642-5. [DOI] [PubMed] [Google Scholar]
  • 115.Gray A., Feldman H.A., McKinlay J.B., Longcope C. Age, disease, and changing sex hormone levels in middle-aged men: results of the Massachusetts male aging study. The Journal of Clinical Endocrinology & Metabolism. 1991;73:1016–1025. doi: 10.1210/jcem-73-5-1016. [DOI] [PubMed] [Google Scholar]
  • 116.Hamalainen E., Tikkanen H., Harkonen M., Naveri H., Adlercreutz H. Serum lipoproteins, sex hormones and sex hormone binding globulin in middle-aged men of different physical fitness and risk of coronary heart disease. Atherosclerosis. 1987;67:155–162. doi: 10.1016/0021-9150(87)90275-9. [DOI] [PubMed] [Google Scholar]
  • 117.Lichtenstein M.J., Yarnell J.W., Elwood P.C., Beswick A.D., Sweetnam P.M., Marks V. Sex hormones, insulin, lipids, and prevalent ischemic heart disease. American Journal of Epidemiology. 1987;126:647–657. doi: 10.1093/oxfordjournals.aje.a114704. [DOI] [PubMed] [Google Scholar]
  • 118.Phillips G.B., Pinkernell B.H., Jing T.Y. The association of hypotestosteronemia with coronary artery disease in men. Arteriosclerosis & Thrombosis. 1994;14:701–706. doi: 10.1161/01.atv.14.5.701. [DOI] [PubMed] [Google Scholar]
  • 119.Swartz C.M., Young M.A. Low serum testosterone and myocardial infarction in geriatric male inpatients. Journal of the American Geriatrics Society. 1987;35:39–44. doi: 10.1111/j.1532-5415.1987.tb01317.x. [DOI] [PubMed] [Google Scholar]
  • 120.Hautanen A., Manttari M., Manninen V., Tenkanen L., Huttunen J.K., Frick M.H. Adrenal androgens and testosterone as coronary risk factors in the Helsinki heart study. Atherosclerosis. 1994;105:191–200. doi: 10.1016/0021-9150(94)90049-3. [DOI] [PubMed] [Google Scholar]
  • 121.Luria M.H., Johnson M.W., Pego R., Seuc C.A., Manubens S.J., Wieland M.R. Relationship between sex hormones, myocardial infarction, and occlusive coronary disease. Archives of Internal Medicine. 1982;142:42–44. [PubMed] [Google Scholar]
  • 122.Marques-Vidal P., Sie P., Cambou J.P., Chap H., Perret B. Relationships of plasminogen activator inhibitor activity and lipoprotein(a) with insulin, testosterone, 17 beta-estradiol, and testosterone binding globulin in myocardial infarction patients and healthy controls. The Journal of Clinical Endocrinology & Metabolism. 1995;80:1794–1798. doi: 10.1210/jcem.80.6.7775625. [DOI] [PubMed] [Google Scholar]
  • 123.Small M., Lowe G.D., Beastall G.H., Beattie J.M., McEachern M., Hutton I. Serum oestradiol and ischaemic heart disease–relationship with myocardial infarction but not coronary atheroma or haemostasis. Quarterly Journal of Medicine. 1985;57:775–782. [PubMed] [Google Scholar]
  • 124.Soisson V., Brailly-Tabard S., Helmer C., Rouaud O., Ancelin M.L., Zerhouni C. A J-shaped association between plasma testosterone and risk of ischemic arterial event in elderly men: the French 3C cohort study. Maturitas. 2013;75:282–288. doi: 10.1016/j.maturitas.2013.04.012. [DOI] [PubMed] [Google Scholar]
  • 125.Ruige J.B., Mahmoud A.M., De Bacquer D., Kaufman J.M. Endogenous testosterone and cardiovascular disease in healthy men: a meta-analysis. Heart. 2011;97:870–875. doi: 10.1136/hrt.2010.210757. [DOI] [PubMed] [Google Scholar]
  • 126.Keating N.L., O'Malley A.J., Smith M.R. Diabetes and cardiovascular disease during androgen deprivation therapy for prostate cancer. Journal of Clinical Oncology. 2006;24:4448–4456. doi: 10.1200/JCO.2006.06.2497. [DOI] [PubMed] [Google Scholar]
  • 127.Morgia G., Russo G.I., Tubaro A., Bortolus R., Randone D., Gabriele P. Prevalence of cardiovascular disease and osteoporosis during androgen deprivation therapy prescription discordant to EAU guidelines: results from a multi-center cross-sectional analysis from the CHOsIng treatment for prostate canCEr (CHOICE) study. Urology. 2016 Oct;96:165–170. doi: 10.1016/j.urology.2016.06.024. [DOI] [PubMed] [Google Scholar]
  • 128.Tsai H.K., D'Amico A.V., Sadetsky N., Chen M.H., Carroll P.R. Androgen deprivation therapy for localized prostate cancer and the risk of cardiovascular mortality. Journal of the National Cancer Institute. 2007;99:1516–1524. doi: 10.1093/jnci/djm168. [DOI] [PubMed] [Google Scholar]
  • 129.Anderson J.L., May H.T., Lappe D.L., Bair T., Le V., Carlquist J.F. Impact of testosterone replacement therapy on myocardial infarction, stroke, and death in men with low testosterone concentrations in an integrated health care system. The American Journal of Cardiology. 2016;117:794–799. doi: 10.1016/j.amjcard.2015.11.063. [DOI] [PubMed] [Google Scholar]
  • 130.Malkin C.J., Pugh P.J., Morris P.D., Kerry K.E., Jones R.D., Jones T.H. Testosterone replacement in hypogonadal men with angina improves ischaemic threshold and quality of life. Heart. 2004;90:871–876. doi: 10.1136/hrt.2003.021121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Basaria S., Coviello A.D., Travison T.G., Storer T.W., Farwell W.R., Jette A.M. Adverse events associated with testosterone administration. New England Journal of Medicine. 2010;363:109–122. doi: 10.1056/NEJMoa1000485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Finkle W.D., Greenland S., Ridgeway G.K., Adams J.L., Frasco M.A., Cook M.B. Increased risk of non-fatal myocardial infarction following testosterone therapy prescription in men. PLoS One. 2014;9:e85805. doi: 10.1371/journal.pone.0085805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Xu L., Freeman G., Cowling B.J., Schooling C.M. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Medicine. 2013;11:108. doi: 10.1186/1741-7015-11-108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Galassi A., Reynolds K., He J. Metabolic syndrome and risk of cardiovascular disease: a meta-analysis. The American Journal of Medicine. 2006;119:812–819. doi: 10.1016/j.amjmed.2006.02.031. [DOI] [PubMed] [Google Scholar]
  • 135.Pasquali R., Casimirri F., Cantobelli S., Melchionda N., Morselli Labate A.M., Fabbri R. Effect of obesity and body fat distribution on sex hormones and insulin in men. Metabolism. 1991;40:101–104. doi: 10.1016/0026-0495(91)90199-7. [DOI] [PubMed] [Google Scholar]
  • 136.Zumoff B., Strain G.W., Miller L.K., Rosner W., Senie R., Seres D.S. Plasma free and non-sex-hormone-binding-globulin-bound testosterone are decreased in obese men in proportion to their degree of obesity. The Journal of Clinical Endocrinology & Metabolism. 1990;71:929–931. doi: 10.1210/jcem-71-4-929. [DOI] [PubMed] [Google Scholar]
  • 137.Simon D., Charles M.A., Nahoul K., Orssaud G., Kremski J., Hully V. Association between plasma total testosterone and cardiovascular risk factors in healthy adult men: the telecom study. The Journal of Clinical Endocrinology & Metabolism. 1997;82:682–685. doi: 10.1210/jcem.82.2.3766. [DOI] [PubMed] [Google Scholar]
  • 138.Haffner S.M., Karhapaa P., Mykkanen L., Laakso M. Insulin resistance, body fat distribution, and sex hormones in men. Diabetes. 1994;43:212–219. doi: 10.2337/diab.43.2.212. [DOI] [PubMed] [Google Scholar]
  • 139.Kapoor D., Aldred H., Clark S., Channer K.S., Jones T.H. Clinical and biochemical assessment of hypogonadism in men with type 2 diabetes: correlations with bioavailable testosterone and visceral adiposity. Diabetes Care. 2007;30:911–917. doi: 10.2337/dc06-1426. [DOI] [PubMed] [Google Scholar]
  • 140.Kapoor D., Goodwin E., Channer K.S., Jones T.H. Testosterone replacement therapy improves insulin resistance, glycaemic control, visceral adiposity and hypercholesterolaemia in hypogonadal men with type 2 diabetes. European Journal of Endocrinology. 2006;154:899–906. doi: 10.1530/eje.1.02166. [DOI] [PubMed] [Google Scholar]
  • 141.Muraleedharan V., Marsh H., Kapoor D., Channer K.S., Jones T.H. Testosterone deficiency is associated with increased risk of mortality and testosterone replacement improves survival in men with type 2 diabetes. European Journal of Endocrinology. 2013;169:725–733. doi: 10.1530/EJE-13-0321. [DOI] [PubMed] [Google Scholar]
  • 142.Corona G., Monami M., Rastrelli G., Aversa A., Tishova Y., Saad F. Testosterone and metabolic syndrome: a meta-analysis study. The Journal of Sexual Medicine. 2011;8:272–283. doi: 10.1111/j.1743-6109.2010.01991.x. [DOI] [PubMed] [Google Scholar]
  • 143.Ikeda Y., Aihara K., Yoshida S., Sato T., Yagi S., Iwase T. Androgen-androgen receptor system protects against angiotensin II-induced vascular remodeling. Endocrinology. 2009;150:2857–2864. doi: 10.1210/en.2008-1254. [DOI] [PubMed] [Google Scholar]
  • 144.Fagman J.B., Wilhelmson A.S., Motta B.M., Pirazzi C., Alexanderson C., De Gendt K. The androgen receptor confers protection against diet-induced atherosclerosis, obesity, and dyslipidemia in female mice. The FASEB Journal. 2015;29:1540–1550. doi: 10.1096/fj.14-259234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pavanello C., Mombelli G. Considering gender in prescribing statins: what do physicians need to know? Clinical Lipidology. 2015;10:499–512. [Google Scholar]
  • 146.Nakamura H., Arakawa K., Itakura H., Kitabatake A., Goto Y., Toyota T. Primary prevention of cardiovascular disease with pravastatin in Japan (MEGA Study): a prospective randomised controlled trial. The Lancet. 2006;368:1155–1163. doi: 10.1016/S0140-6736(06)69472-5. [DOI] [PubMed] [Google Scholar]
  • 147.Mayne J., Raymond A., Chaplin A., Cousins M., Kaefer N., Gyamera-Acheampong C. Plasma PCSK9 levels correlate with cholesterol in men but not in women. Biochemical and Biophysical Research Communications. 2007;361:451–456. doi: 10.1016/j.bbrc.2007.07.029. [DOI] [PubMed] [Google Scholar]
  • 148.Roubtsova A., Chamberland A., Marcinkiewicz J., Essalmani R., Fazel A., Bergeron J.J. PCSK9 deficiency unmasks a sex- and tissue-specific subcellular distribution of the LDL and VLDL receptors in mice. Journal of Lipid Research. 2015;56:2133–2142. doi: 10.1194/jlr.M061952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Friedrich D., Ziajka P., Ravilla S., Diaz J., Vicari R. PCSK-9i gender study. Journal of Clinical Lipidology. 2017;11:817. [Google Scholar]
  • 150.Vaccarino V., Rathore S.S., Wenger N.K., Frederick P.D., Abramson J.L., Barron H.V. Sex and racial differences in the management of acute myocardial infarction, 1994 through 2002. New England Journal of Medicine. 2005;353:671–682. doi: 10.1056/NEJMsa032214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Vaccarino V., Parsons L., Every N.R., Barron H.V., Krumholz H.M. Sex-based differences in early mortality after myocardial infarction. National registry of myocardial infarction 2 participants. New England Journal of Medicine. 1999;341:217–225. doi: 10.1056/NEJM199907223410401. [DOI] [PubMed] [Google Scholar]
  • 152.Chandrasekhar J., Baber U., Sartori S., Faggioni M., Aquino M., Kini A. Sex-related differences in outcomes among men and women under 55 years of age with acute coronary syndrome undergoing percutaneous coronary intervention: results from the PROMETHEUS study. Catheterization and Cardiovascular Interventions. 2017;89:629–637. doi: 10.1002/ccd.26606. [DOI] [PubMed] [Google Scholar]
  • 153.Epps K.C., Holper E.M., Selzer F., Vlachos H.A., Gualano S.K., Abbott J.D. Sex differences in outcomes following percutaneous coronary intervention according to age. Circulation of Cardiovascular Quality Outcomes. 2016;9:S16–S25. doi: 10.1161/CIRCOUTCOMES.115.002482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Rodriguez F., Olufade T.O., Ramey D.R., Friedman H.S., Navaratnam P., Heithoff K. Gender disparities in lipid-lowering therapy in cardiovascular disease: insights from a managed care population. Journal of Womens Health (Larchmt) 2016;25:697–706. doi: 10.1089/jwh.2015.5282. [DOI] [PubMed] [Google Scholar]
  • 155.Rodriguez F., Olufade T., Heithoff K., Friedman H.S., Navaratnam P., Foody J.M. Frequency of high-risk patients not receiving high-potency statin (from a large managed care database) The American Journal of Cardiology. 2015;115:190–195. doi: 10.1016/j.amjcard.2014.10.021. [DOI] [PubMed] [Google Scholar]
  • 156.Wing L.M.H., Reid C.M., Ryan P., Beilin L.J., Brown M.A., Jennings G.L.R. A comparison of outcomes with angiotensin-converting–enzyme inhibitors and diuretics for hypertension in the elderly. New England Journal of Medicine. 2003;348:583–592. doi: 10.1056/NEJMoa021716. [DOI] [PubMed] [Google Scholar]
  • 157.Ridker P.M., Cook N.R., Lee I.M., Gordon D., Gaziano J.M., Manson J.E. A randomized trial of low-dose aspirin in the primary prevention of cardiovascular disease in women. New England Journal of Medicine. 2005;352:1293–1304. doi: 10.1056/NEJMoa050613. [DOI] [PubMed] [Google Scholar]
  • 158.British Heart Foundation CVD statistics -BHF UK factsheeet. In.

Articles from Molecular Metabolism are provided here courtesy of Elsevier

RESOURCES