Skip to main content
Therapeutic Advances in Endocrinology and Metabolism logoLink to Therapeutic Advances in Endocrinology and Metabolism
. 2022 Oct 28;13:20420188221129946. doi: 10.1177/20420188221129946

Menopause, androgens, and cardiovascular ageing: a narrative review

Eleni Armeni 1,, Irene Lambrinoudaki 2
PMCID: PMC9619256  PMID: 36325501

Abstract

Cardiovascular disease is the leading cause of death worldwide; however, women tend to be less affected than men during their reproductive years. The female cardiovascular risk increases significantly around the time of the menopausal transition. The loss of the protective action of ovarian oestrogens and the circulating androgens has been implicated in possibly inducing subclinical and overt changes in the cardiovascular system after the menopausal transition. In vitro studies performed in human or animal cell lines demonstrate an adverse effect of testosterone on endothelial cell function and nitric oxide bioavailability. Cohort studies evaluating associations between testosterone and/or dehydroepiandrosterone and subclinical vascular disease and clinical cardiovascular events show an increased risk for women with more pronounced androgenicity. However, a mediating effect of insulin resistance is possible. Data on cardiovascular implications following low-dose testosterone treatment in middle-aged women or high-dose testosterone supplementation for gender affirmatory purposes remain primarily inconsistent. It is prudent to consider the possible adverse association between testosterone and endothelial function during the decision-making process of the most appropriate treatment for a postmenopausal woman.

Keywords: androgens, cardiovascular disease, endothelial function, menopause, oxidative stress

Introduction

Cardiovascular disease (CVD) remains the leading cause of death worldwide, accounting for 30.3% of women’s all-cause mortality in the United States in 2018.1,2 Although incidence and mortality of CVD, in general, are lower in women compared to men, estimated as 25% lower risk for incident CVD and 38% lower risk for all-cause mortality, this trend tends to reverse after midlife.3 Data from the NHANES (National Health and Nutrition Examination Survey) from 2015 to 2018 show that CVD prevalence is estimated at 75.4% for women aged 60–79 years and up to 90.8% for women aged more than 80 years, incorporating hypertension as a primary cardiovascular outcome.2 Interestingly, coronary heart disease in women notably increases during midlife,2,4 coinciding with the menopausal transition.5

The age of ovarian senescence appears as an additional important determinant of overall cardiovascular risk. In a pooled analysis of 203,767 postmenopausal women,6 retrieved from observational studies, an inverse-graded association was observed between younger age at natural or surgical menopause and rates of incident CVD. The risk has been estimated as 1.59-times higher in women experiencing menopause at an age less than 35 years as opposed to women experiencing menopause at the age of 50–54 years.6 In addition, women experiencing early menopause or premature ovarian insufficiency had a higher relative risk for prevalent coronary heart disease and CVD mortality, estimated as 1.5-times and 1.19-times higher, respectively, compared with women experiencing spontaneous menopause at an age more than 45 years.7 Similarly, women experiencing menopause at the age of 50–54 years had a 13% lower risk of fatal coronary heart disease than women experiencing menopause at 50 years.7

According to the latest statement of the American Heart Association, the menopausal transition is an essential window of accelerated cardiovascular risk suggesting the acquisition of primary prevention strategies.5 Recent evidence highlighted the importance of both the traditional risk factors and additional sex-specific factors, including the possible effect of sex hormones in determining the overall cardiovascular risk.8 A large amount of data support a possible association between hormonal changes associated with menopause and age-related alterations of the cardiovascular system, further contributing to the sharp increase of CVD observed at midlife.5,8 However, the available literature seems to be characterized by heterogeneity, determined by the study design.

The aim of this review is to summarize the evidence on the association between androgens and subclinical as well as clinical CVD in ageing women.

Cardiovascular ageing: the effect of menopause

The process of cardiovascular ageing is comprised of a series of changes affecting both the left ventricle as well as the overall arterial tree. The effect of ageing on the left ventricle seems to range from concentric hypertrophy with or without diastolic dysfunction, aortic valvulopathy, reduced variability of the heart rate, and annular calcification of the mitral valve as well as conduction abnormalities.9 The arterial tree itself is characterized by progressively enlarging vascular lumen as well as thickening of the vessel wall.9,10 Simultaneously, early stages of vasodilation become apparent in the aorta, the large branches proximal to the myocardium.9,10 The raised aortic stiffness results in a further rise in reflected waves and aortic pulse wave velocity, which became apparent as elevated systolic blood pressure and increased left ventricular afterload.10 Diastolic blood pressure, on the contrary, tends to decline with age.10 The raised systolic blood pressure in the context of low diastolic blood pressure results in increased oxygen demand by the myocardium, the supply of which is likely to be compromised in the context of advancing atherosclerosis of coronary arteries.10

At a cellular level, ageing endothelial cells and cardiomyocytes produce increasing amounts of reactive oxygen species (ROS).1113 This process takes place principally in the mitochondria, which constitute up to 45% of the cardiac cell volume.14,15 Ultimately, ageing-mediated increased production of ROS compromises vasorelaxation, increases vascular stiffness and impairs endothelial cell turnover and release of nitric oxide.16,17 At a cellular level, oxidative stress and marked production of ROS result in the following changes, which contribute to the development of heart failure, valvular degeneration, and atrial fibrillation: (1) epigenetic alterations and DNA damage, (2) increased expression of tumour necrosis factor-alpha and matrix metalloproteinase’s, (3) impaired function of the sarcoplasmic/endoplasmic reticulum calcium-ATPase and consequently insufficient reuptake of ionized calcium, (4) impaired bioavailability of nitric oxide and suppressed function of endothelial nitric oxide synthase.16,17

Menopause-associated hormone changes contribute to the age-related alterations evident in the cardiovascular system. Oestrogen deficiency is implicated in endothelial dysfunction and reduced NO bioavailability.18 The postmenopausal ovary continues to produce testosterone, the levels of which decline progressively with ageing.19,20 The adrenal glands contribute to the pool of testosterone through an ongoing secretion of dehydroepiandrosterone sulphate (DHEAS), the levels of which also decline with ageing, as well as androstenedione.19,20 Androgen production during the female reproductive life is evident as follows:20 (1) in the ovarian theca cells, which are secreting up to 20% of circulating DHEAS, (2) in the ovarian stroma, which is producing 50% of the circulating androstenedione and up to 25% of the circulating testosterone, (3) the adrenal zona reticularis which is secreting up to 50% of circulating DHEAS, androstenedione and up to 25% of circulating testosterone. The remaining portion of testosterone is produced with the direct conversion from circulating androstenedione.20

Increasing age, as well as the menopause-related hormonal changes, result in changes that directly or indirectly contribute to increase in cardiovascular risk21 (Figure 1). Following ovarian senescence, the production of oestrogen decreases rapidly.22 Simultaneously, levels of sex hormone-binding globulin also decrease due to the subsequent reduction in oestrogen-liver interaction,23 a change that results in an increase in circulating levels of sex hormones.24 The rate of androgen production declines to a lesser extent, given the ongoing contribution of the adrenal glands to the pool of circulating sex hormones.20 Consequently, the androgen-to-oestrogen ratio increases, resulting in an environment of relative androgenicity.25 These hormonal alterations, as well as ageing per se, are known to induce pathophysiological manifestations, which consist of impaired fibrinolysis,26 insulin resistance,27 and visceral adiposity.28 In this context, impaired fibrinolysis contributes to the origin and progression of atherosclerotic changes.29,30 Hyperandrogenism, visceral adiposity, and insulin resistance contribute to metabolic dysfunction,31,32 generation of oxidative stress and low-grade inflammation,33,34 which have been linked with endothelial dysfunction.35 As described by experimental data, the early stages of the atherosclerotic process, as well as hyperandrogenemia, have also been demonstrated to contribute to endothelial dysfunction,36,37 which eventually results in more advanced atherosclerotic changes.38 Endothelial dysfunction is a recognized factor promoting the development of atherosclerosis.39

Figure 1.

Figure 1.

Pathophysiology of menopausal hormone changes and the related effects on the vascular endothelium.

Ovarian senescence is associated with a sharp decrease in levels of oestrogen and sex hormone binding globulin, secondary to the reduced oestrogen-liver interaction. Androgen production declines at a less steep rate compared to oestrogen production, resulting into a higher androgen to oestrogen ratio, an environment of relative androgenicity. The hormonal alterations together with ageing per-se are linked with a series of pathophysiological alterations, which overall increase cardiovascular risk. Impaired fibrinolysis contributes to atherosclerotic changes. Hyperandrogenism, visceral adiposity, and insulin resistance contribute to metabolic dysfunction, generation of oxidative stress, and low-grade inflammation, which result into endothelial dysfunction. Hyperandrogenemia has also been demonstrated to contribute to endothelial dysfunction, which eventually result into more advanced atherosclerotic changes.

The effect of androgens on cardiovascular tissues: preclinical studies

Androgen receptors are widely distributed in different cell types of the cardiovascular tissue; their presence has been confirmed in endothelial cells, cardiomyocytes and vascular smooth muscle cells (VSMCs).40 The interaction between androgens and endothelial cells is of particular significance, as these represent key players in cardiovascular pathophysiology, with a fundamental role in cardiovascular health and disease.41

Emerging data from in vitro studies suggest cardiovascular implications of testosterone to cells of the cardiovascular system (Table 1). Overall, animal studies support that testosterone improves endothelial mediated vasodilation,42,47 increases ROS generation and oxidative stress,46,60 and promotes apoptosis and migration of VSMCs.48,49 Data on angiogenesis are conflicting, with evidence supporting a positive effect of testosterone in male animals only, with no effect in females.45 The results on a possible link between testosterone and VSMC calcification remain conflicting, with studies supporting stimulation of VSMCs migration,48 induction of VSMCs-apoptosis,49 or suppression50,51 of VSMCs-calcification. Studies addressing the effect of testosterone on cardiomyocytes report protection from apoptosis,52 but also induction of apoptosis,54 suppression of oxidative stress55 and increased intracellular calcium ion signalling.53 Moreover, preclinical studies describe the effect of the synergistic action of estradiol and testosterone on cells of the cardiovascular system. Accordingly, spontaneously hypertensive ovariectomized rats treated with combined equine oestrogen (CEE) showed suppression of oxidative stress (ROS).56 This beneficial effect of CEE-treatment was reversed after the addition of testosterone, with induction of ROS.56,57 Others reported that the optimal estradiol to testosterone ratio following treatment with a combination of estradiol and testosterone is associated with a decrease in apoptosis,58,59 endothelial injury, vascular inflammation, coagulation, and formation of foam cells and vascular lipid lesions.59

Table 1.

Experimental data on the effect of testosterone on cells of the cardiovascular system.

Ref. Target tissue Messenger or receptor involved Experimental response
Preclinical studies on the effect of testosterone administration
Endothelial cells
Arapa-Diaz et al.42 ECs derived from the coronary vascular bed of male hypertensive rats Bradykinin/NO-dependent pathway (↑) Endothelium-dependent relaxation
Campelo et al.43 ECs derived from aortic strips of female, sexually mature Wistar rats MAPK and PKC pathway mediating NO production Modulation of EC growth
Goglia et al.44 HUVECs from female and male foetuses
ECs from ovariectomized Wistar rats
Activation of eNOS
Regulation of t-PA and PAI-I expression
Modulation of ECs function
• Evident in rats administered physiological concentration of T
• Not evident in rats administered supraphysiological doses of T
• No association with DHT of either physiological or supraphysiological dose
Sieveking et al.45 HUVEC The effect of AR activation depends on the gender:
• Male ECs: antagonism of AR leads to reduced neovascularization, which is restored with T administration
• Female ECs: (↑) expression of AR resulted in androgen sensitivity and angiogenesis
(↑) neovascularization after ischemic injury in males only
Alves et al.46 Thoracic aorta derived from male 12 week-old NLRP3 knockout mice mROS generation and NLRP3 inflammasome activation (↑) contractile responses
(↓) endothelium-dependent vasodilation
Vascular smooth muscle cells
Montaño et al.47 Myocytes derived from thoracic aortas of adult male Wistar rats (↑) T concentration induces (↑) intracellular Ca2 + and cAMP production
(↓) T concentration results in L-VOCCs antagonism
(↑) vasodilation
Chignalia et al.48 VSMCs derived from Wistar-Kyoto rat and spontaneously hypertensive rat cSrc-dependent pathway and NADPH oxidase-derived ROS by genomic/non-genomic mechanisms • Induction of ROS generation
• Stimulation of VSMCs migration
Lopes et al.49 VSMCs derived from male Wistar rats Activation of MAPK and tyrosine kinase pathway
Modulation of the NADPH oxidase complex
VSMCs apoptosis via ROS generation
Zhu et al.50 VSMCs were derived from primary murine AR-ablated compared to WT-mice Non-specific alkaline phosphatase mRNA expression (↓) calcification in VSMCs from AR- vs WT-mice
Son et al.51 Human aortic smooth muscle cells AR signalling regulates Growth arrest-specific gene 6 transcription Inhibition of VSMCs calcification
Cardiomyocytes
Xiao et al.52 Cardiomyocytes derived from embryonic rat heart • AR and NF-κB signalling pathways
(↑) Akt activity and (↓) caspase-3 expression in cardiac myocytes
Protection of cardiomyocytes from superoxide injury and apoptosis
Vicencio et al.53 Cardiomyocytes derived from neonatal male Sprague Dawley rats PTX sensitive G protein – phospholipase C / inositol 1,4,5 triphosphate signalling pathway (↑) intracellular Ca2+ signalling
Akdis et al.54 Cardiomyocyte-based model derived from human stem cells in ARVC/D model Not specified (↑) T-induced cardiomyocyte apoptosis and lipid accumulation
Zhang et al.55 Cardiomyocytes derived from testicular feminized and castrated male mice AR-independent pathway
(↑) activities of SOD and GSH-Px enzyme
(↓) MDA levels
(↓) mtDNA mutations
suppression of oxidative stress
Preclinical studies on the effect of combined testosterone and oestrogen administration
Costa et al.56 Ovariectomized spontaneously hypertensive rats received CEE + T Phosphorylation of the cytosolic NADPH-oxidase subunit p47phox CEE + T vs CEE-only therapy resulted in (↑) ROS
Costa et al.57 Hypertensive ovariectomized female rats received CEE + T CYP4 F3/20-HETE pathway (↑) adrenergic vasoconstriction in isolated aorta
(↑) ROS generation in isolated aorta
Xu et al.58 Cardiomyocytes treated with estradiol (50 nM) and testosterone (10 nM) TGF-β1 pathway The optimal E2/T ratio is associated with (↓) apoptosis of cardiomyocytes
Dai et al.59 HUVECs derived from female mice were treated with 17b-estradiol (1 mg/day) ± testosterone (7 mg/day) for 120 days • PI3K/Akt pathway
• (↓) Expression of cleaved caspase 3, Bax, cleaved PARP
• (↑) Expression of Bcl-2
The optimal E2/T ratio is associated with
• (↓) Apoptosis of cardiomyocytes
• (↓) Lipid lesions, and formation of foam cells,
• (↓) Endothelial injury,
• Modulating the
coagulation system function
• (↓) Inflammation

A, androgens; AR, androgen receptor; cAMP, cyclic adenosine monophosphate; CEE, conjugated equine oestrogen; CP4 F3, cytochrome P450 4F3B; DHT, dihydrotestosterone; EC, endothelial cells; GSH-Px, glutathione peroxidase; HUVEC, human umbilical vein endothelial cells; iPSC-CMs, pluripotent stem cell-derived cardiomyocytes; L-VOCCs, L-type voltage operated Ca2+ channels; MAPK, mitogen-activated protein kinase; MDA, malondialdehyde; mtDNA, mitochondrial deoxyribonucleid acid; NADPH, Nicotinamide Adenine Dinucleotide Phosphate; PAI-I, plasminogen activator inhibitor I; PI3 K/Akt, phosphoinositide-3-kinase/protein kinase B; PTX, pertussis toxin; NLRP3, NOD-, LRR- and pyrin domain-containing protein 3; NO, nitric oxide; PKC, protein kinase C; ROS, Reactive oxygen species; SOD, superoxide dismutase; Src, regulator of redox-sensitive migration; T, testosterone; t-PA, tissue plasminogen activator; VSMC, vascular smooth muscle cell; (↑), tendency to increase; (↓), tendency to decrease; 20-HETE, 20-hydroxyeicosatetraenoic acid.

Data from preclinical studies describing the effect of dehydroepiandrosterone (DHEAS) on cardiovascular tissues are presented in Table 2. Experimental data on the effect of DHEAS administered to endothelial cells (ECs) describe that DHEAS-treatment is reducing the proliferation rate,61 or increasing the proliferation rate of ECs.67,69 DHEAS has, furthermore, been reported to induce endothelial cell necrosis.61,64 Others described reduction of vascular inflammation,62 exertion of antioxidative and anti-inflammatory effects,65,66 increased angiogenesis,67 and improved endothelial function.68 DHEAS treatment administered to VSMCs results in reduced inflammation and oxidative stress in healthy VSMCs.71 In studies describing balloon injury models, DHEAS treatment is linked with reduced VSMC migration and proliferation72,73 and increased apoptosis.72 The effect of DHEAS-treatment on cultures of cardiomyocytes consists of a tendency towards a lower chronotropic and hypertrophic response,76 reduced pulmonary artery pressure, prevention of pulmonary artery hyperreactivity and remodelling and prevention of the remodelling of the right ventricle.74

Table 2.

Experimental data on the effect of DHEAS on cardiovascular tissues.

Ref. Target tissue Messenger or receptor involved Experimental response
Endothelial cells
Gündoğan et al.61 Primary HUVECs were treated with DHEA (0–200 μM, 24–72 h) Not clearly described • Reduced proliferation rate of endothelial cells at 24 h, 48 h, 72 h
• Inhibition of proliferation and induction of necrosis
Altman et al.62 Human aortic ECs were treated with DHEAS (48 h) and then with TNF-alpha • TNF-alpha mediated activation of NF-kappaB
• PPARα inhibition
• Dose-dependent inhibition of the expression of VCAM-1
Curatola et al.63 Human coronary ECs from men and women • Increased expression of polysialylated NCAM • Inhibiting adhesion of monocytes
Liu et al.64 Bovine aortic ECs • Galphai-PI3K/Akt-Bcl-2 pathway • Protecting against cell ECs–apoptosis
Huerta-García et al.65 HUVEC exposed to PM and NPs and subsequently treated with DHEA Not clearly described • Anti-oxidative and anti-inflammatory effects
Huerta-García et al.66 HUVEC treated with a high concentration of DHEA and/or glucose • NF-κB • (↓) Oxidative stress
• (↓) Activation of endothelial cells
Liu et al.67 Bovine aorta ECs • PTX-sensitive G proteins
• ERK1/2
• Enhanced angiogenesis
• Increased proliferation of vascular ECs
Wang et al.68 In vivo: Ovariectomized rabbits
In vitro: Injured HUVECs due to oxidized LDL particles
• Not mediated through oestrogen receptor alpha or beta
• (↓) the lipopolysaccharide-induced NF-κB transcription
In vivo: (↑) EC-oestrogen receptor and (↑) serum NO
In vitro: (1) (↑) NO synthesis, (2) (↓) expression of adhesion molecules (VCAM-1, ICAM-1 and E-selectin); (3) (↓) expression of inflammatory molecules (MCP-1 and MDA)
Williams et al.69 Bovine aortic ECs derived from the aorta of young calves,
HUVEC
(↑) Production of eNOS
(↑) Expression of MAPK ERK 1 / 2 kinase
Effect not mediated by AR or ER
(↑) Poliferation of ECs
Vascular smooth muscle cells
Ochi et al.70 A7r5 aortic smooth muscle cells from the thoracic aorta of rat embryos were treated with DHEA. Cells lines were pretreated with 6-AN for competitive inhibition of G6PD • Voltage-independent inhibition: mediated by G6PD
• Voltage-dependent inhibition: open channel blocks (similar to DHP-CCBs)
• Voltage-dependent and -independent inhibition of ion-channels (L-type Ca2 +) and window current (IWD)
Chen et al.71 VSMC were retrieved from the thoracic aorta of SD rats • Inhibition of the ERK 1 / 2 signalling pathway
• Inhibition of the p38-MAPK/NF-κB signalling pathway
Reduction of inflammation and oxidative stress in VSMCs
Ii et al.72 Baloon injury of rabbit carotid artery in adult males treated with DHEAS or saline PPARα signalling Medial carotid layer cell death and apoptosis
Cyclin-dependent kinase inhibitor p16INK4a • (↓) VSMC migration
• (↓) VSMC proliferation
Urata et al.73 Thoracic aorta VSMC derived from embryonic rat models A7r5 • Upregulation of PPAR-α
• Inhibition of platelet-derived growth factor-β phosphorylation
• Up-regulation of glutathione/glutaredoxin-1 redox system
• Control of low molecular weight-protein tyrosine phosphatase
• No interaction with androgen or oestrogen receptor
• Suppression of VSMC phenotypic proliferation
Cardiomyocytes
Dumas de La Roque et al.74 Juvenile Wistar rat models of pulmonary hypertension were treated with DHEA or control (↑) The expression and activity of the large-conductance BKCa-inhibited KCL- and serotonin-induced VSMC contraction and proliferation • (↓) Pulmonary arterial pressure
• Preventing pulmonary artery hyper-reactivity and remodelling
• Preventing RV hypertrophy
Alzoubi et al.75 Adult chronically hypoxic, adult male Sprague-Dawley rats received monocrotaline injection followed by 5-week treatment with DHEA • (↓) NADPH levels in RV tissue • Preservation of contractile RV-function against fibrosis and apoptosis
Mannic et al.76 Ventricles retrieved from 1–2 day old Wistar rats; assessment of the effect of DHEAS following exposure to dexamethasone or aldosterone • Cellular pathways, likely including MAPK • (↓) Hypertrophic response of cardiomyocytes
• Non-genomic pathway: alteration of the activity of T-type calcium channel isoform, a1H
• Genomic pathway: unclear
• (↓) Chronotropic response of cardiomyocytes
Nakamura et al.77 In vivo: Human LV-tissue autopsy samples in patients with HF vs control
In vitro: Neonatal cardiocyte culture
Not clearly described • In vivo: DHEA levels in LV tissue are higher in control vs HF-patients
• In vitro: (1) (↓) Increase in the size of myocytes; (2) (↓) mRNA levels for BNP production

BKCa, Ca2+-activated potassium channel; BNP, brain natriuretic peptide; DHEA, dehydroepiandrosterone; DHP-CCBs, dihydropyridine Ca2+ channel blockers; EC, endothelial cells; ERK1/2, extracellular signal-regulated kinase 1/2; G6PD, glucose-6-phosphate dehydrogenase; HF, heart failure; HUVEC, human umbilical vein endothelial cells; ICAM, 1, intercellular adhesion molecule 1; LDL, low-density lipoprotein; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; mRNA, messenger rivonucleid acid; NCAM, neural cell adhesion molecule to monocyte; NO, nitric oxide; NPs, nanoparticles; PI3 K, phosphatidylinositol 3-kinase; PM, particulate matter; PTX, pertussis toxin; ROS, reactive oxygen species; RV, right ventricle; TNF, tumour necrosis factor; VCAM, vascular adhesion molecule; (↑), tending to increase; (↓), tending to decrease; 6-AN, 6-aminonicotinamide.

Pathophysiological mechanisms determining the interaction between menopause and endothelial cells

Endothelial cells are known to express both oestrogen (ER) and androgen receptors (AR), which are known to mediate their functional capacity.40 The binding of oestrogen on the ER has been linked with decreased expression of adhesion molecules (e.g. CD40, P-selectin, E-selectin, ICAM-1) as a response to atherogenic promoting factors, ensuing in increased NO release and VSMC relaxation.78 The chronic oestrogen deficiency following ovarian senescence contributes to endothelial dysfunction with a decrease in NO bioavailability, secondary to decreased NO synthesis and increased NO inactivation.79 Moreover, the postmenopausal environment of hypoestrogenemia induces an altered redox balance, contributing to oxidative stress,80 while also inducing the expression of homocysteine and cysteine molecules, with adverse effects on endothelial function.81

Focusing on the interactions mediated by androgens, testosterone has been linked with a dual effect on cardiovascular cells, both pro-oxidant and antioxidant.60 More specifically, the pro-oxidant effect consists of suppressed activation of the endothelial NO synthase (eNOS), resulting in impaired endothelial NO release in women.37,82 The antioxidant effect is maintained through conversion to estradiol, which also contributes to mediating high levels of antioxidant enzymes.60 Earlier data suggested that the degree of oxidative stress is an essential factor regulating testosterone action; thus, an environment of high oxidative stress is related to adverse testosterone effects and vice versa.55 These observations highlight the need for further research to determine factors driving oxidative stress during and after the menopausal transition.

Data on the cardiovascular alterations observed in women during the menopausal transition is rather sparse. Samargandy et al.83 evaluated 339 women retrieved from the Study of Women’s Health Across the Nation, aiming to measure the annual percentage change in carotid-femoral PWV in relation to the final menstrual period (FMP). This study83 highlighted the pattern of statistically significant changes in values of cfPWV around the FMP. Accordingly, cfPWV values increased at a steady rate up to 12 months prior to the FMP (0.9%, 95% CI: −0.6% to 2.3%), continued to increase more rapidly up to 1 year after the FMP (7.5%, 95% CI: 4.1% to 11.1%), and subsequently decreased at a steady rate (–1.0%, 95% CI: −2.8% to 0.8%).

Endogenous androgens and CVD: data from human cohort studies

Subclinical CVD

Data on cohort studies assessing the possible associations between endogenous androgens and markers of subclinical vascular disease are presented in Table 3.

Table 3.

Cohort studies exploring the possible associations between endogenous androgens and subclinical vascular disease.

Ref. Study sample and Design Primary outcome FU Results Discussion
Endothelial function
Georgiopoulos et al.84 180 postmenopausal women with no overt CVD or diabetes
Design: CS
New-onset HTN
FMD
Median: 29 m baseline FAI was associated with:
• New-onset HTN (OR 2.71, 95% CI: 1.14–6.41)
• change in FMD (–0.42% per SD of FAI)
(↑) Baseline FAI was associated with:
• new-onset HTN
• change in FMD
Arterial stiffness
Subramanya et al.85 Multi-ethnic study of atherosclerosis (2018)
457 postmenopausal women and 548 men aged 45–84 years
Design: CS and L
AAD
PWV
Median:
10 y
Baseline: Highest tertile vs lowest tertile of FT,
• AAD, b-coefficient = −0.10, 95% CI: −0.19 to −0.01
• PWV, b-coefficient = −0.04, 95% CI: −0.10 to 0.02
CS: (↑) FT was associated with (↓) AAD and (↓) PWV
L: no association
Lambrinoudaki et al.86 411 consecutively recruited healthy postmenopausal women
Design: CS
PWV
SI index
FMD
CAS
NA FAI was associated with
• PWV (beta = 0.149, p value = 0.014),
• SIindex (beta = 0.154, p value = 0.022),
• CAS (beta = 0.193, p value 0.02)
(↑) FAI was associated with
• (↑) PWV
• (↑) SIindex
• (↑) CAS
Georgiopoulos et al.84 180 postmenopausal women with no overt CVD or diabetes
Design: CS
PWV Median: 29 m Baseline FAI (high vs lower quartile) and transition into abnormal PWV: NRI = 53.4 ± 23.2%, p value = 0.021, across follow-up period (↑) FAI at baseline was associated with (↑) risk for abnormal PWV
Atherosclerosis
Kische et al.87 Study of Health in Pomerania (2016),
2,140 individuals (mean age: 60.8 years)
Design: CS and L
Carotid IMT Median: 5 year • Association between TT and IMT in women (beta per log unit increase: 0.02; 95% CI: 0.007–0.45)
• Association between SHBG and incident plaques (Q1 vs Q3: beta 1.35, 95% CI: 1.04–1.74)
• No consistent association between TT and subclinical CVD
CS: (↑) TT was associated with (↑) IMT; (↓) SHBG was associated with the incidence of atherosclerotic plaques
L: no consistent association between subclinical CVD and TT
Lee et al.88 The CARDIA study:
367 middle-aged women
Design: L
Carotid bulb IMT FU points: 15 and 20 years Carotid bulb IMT, multivariable adj. model
Cortisol to free testosterone ratio
• Tertile T3 vs T1: beta = 0.088, p = 0.006
(↑) Cortisol-to-free T ratio was associated with higher carotid bulb IMT
Calderon-Margalit et al.89 The CARDIA study
1629 women
Design: L
Carotid IMT 20 years Coronary – IMT, highest quartile:
• SHBG levels, Q4, OR = 0.56, 95% CI: 0.37–0.84, p for linear trend across quartiles 0.005
(↑) Carotid IMT was associated with (↓) SHBG
No association between carotid IMT and TT or FT
Coronary artery calcium
Subramanya et al.90 Multi-Ethnic Study of Atherosclerosis (2019)
2759 postmenopausal women (age 65 ± 9 years) free from baseline CVD
Design: CS and L
CAC Median 4.7 y (maximum 10 years) • (↑) FT and relative CAC progression ratio (2.16, 95% CI: 1.011–1.56)
• (↑) SHBG and CAC progression ratio (0.80, 95% CI: 0.64–0.99)
CS: No association between CAC and sex hormones
L: CAC progression ratio was associated with (↑) FT and (↓) SHBG
Calderon-Margalit et al.89 The CARDIA study
1629 women Design: L
CAC 20 years Incidence of CAC, multivariable adj. models
• SHBG above vs below median, adjusted OR = 0.59, 95% CI: 0.40–0.87
(↑) CAC-incidence was associated with (↓) SHBG but not with TT or FT
Lee et al.88 The CARDIA study
367 middle-aged women
Design: L
New-onset CAC FU points: 15 and 20 years Incident CAC,
Cortisol to free testosterone ratio
• Tertile T3 vs T1: OR = 3.45 (95% CI: 1.18–10.06)
(↑) CAC-incidence was associated with (↑) cortisol-to-FT ratio, but not with cortisol-to-TT ratio or with the cortisol-to-SHBG ratio

AAD, ascending aortic distensibility; CAC, coronary artery calcification; CARDIA, coronary artery risk development in young adults; CAS, cumulative marker combining pulse wave velocity and stiffness index; CI, confidence interval; CS, cross-sectional; CVD, cardiovascular disease; FAI, free androgen index; FT, free testosterone; FU, follow-up; IMT, intima media thickness; m, months; max, maximum; NA, non applicable; NRI, net reclassification index; NS, non-significant; OR, odds ratio; PWV, pulse wave velocity; SHBG, sex hormone binding globulin; SIindex, stiffness index; TT, total testosterone; y, years.

The possible association between endogenous androgens and endothelial function in middle-aged women has been investigated by one cohort study. Georgiopoulos et al.84 evaluated endothelial function in a total of 180 postmenopausal women with no overt CVD or diabetes during a follow-up period of up to 29 months. This study concluded that the pattern of change in %FMD levels during the follow-up period was associated with baseline levels of FAI: one SD increase in levels of baseline FAI was linked with a 0.42% decrease in mean FMD values, measured during follow-up.

The possible association between endogenous androgens and markers of arterial stiffness in middle-aged women has been investigated by three cohort studies. Subramanya et al.91 investigated 1345 postmenopausal women aged 45–84 years, retrieved from the MESA (multi-ethnic study in atherosclerosis), describing evidence of cross-sectional but not longitudinal associations between free testosterone and ascending aortic distensibility (AAD), during a follow-up of 10 years. In fact, at baseline, women with free testosterone at the highest tertile versus lowest tertiles had lower values of AAD, adjusting for risk factors (b-coefficient = −0.10, 95% CI: −0.19 to −0.01). This study91 could not demonstrate cross-sectional or longitudinal associations between free testosterone in tertiles and PWV. In an analysis of 411 consecutive middle-aged women, levels of FAI were directly associated with arterial stiffness in a study of non-causal design.86 More specifically, FAI was shown to directly predict PWV (beta = 0.149, p value = 0.014), stiffness index (beta = 0.154, p value = 0.022), combined measure of local and aortic arterial elastic properties (beta = 0.193, p value = 0.02). Using structural equation modelling analysis, this study also highlighted that the link between FAI and arterial stiffness is not mediated by FAI. The small cohort study by Georgiopoulos et al.84 reported that higher values of FAI at baseline are linked with a greater risk for transition into the category of abnormal PWV during a follow-up period of 29 months, taking cardiovascular risk factors into account. The results of longitudinal analyses seem to be supportive of a direct link between higher serum levels of testosterone and arterial stiffness in middle-aged women.

The possible link between serum levels of androgens and subclinical atherosclerosis was assessed by two cohort studies. Kische et al.92 provided cross-sectional and longitudinal associations between circulating androgens and carotid IMT as well as incident atherosclerotic plaque burden in a total of 2140 individuals aged on average 60.8 years and retrieved from the Study of Health in Pomerania. They showed a cross-sectional association between higher total testosterone and IMT, as well as a significant negative association between SHBG and incident plaque burden.92 However, they could not confirm any consistent longitudinal association between total testosterone and subclinical CVD.92 The earlier sub-analysis of the CARDIA (Coronary Artery Risk Development in Young Adults) study evaluated 1629 women who were followed up for 20 years.89 Coronary IMT measures within the highest quartile were associated inversely with levels of SHBG (Q4 levels, OR = 0.56, 95% CI: 0.37–0.84, p value = 0.005), while no associations were observed with total and free testosterone. Overall, longitudinal studies suggest that higher SHBG levels protect from the progression of carotid wall thickening, while there is no convincing evidence on the effect of total or free testosterone.

The possible interrelation between serum levels of circulating androgens and coronary artery calcium (CAC) has been investigated by three studies. The MESA study90 described a lack of cross-sectional association between prevalent CAC and serum androgen. The longitudinal analysis90 showed that the CAC-progression ratio increased in parallel with higher levels of free testosterone (2.16, 95% CI: 1.01–1.56), and the CAC-progression risk decreased with higher levels of SHBG (0.80, 95% CI: 0.64–0.99). Two sub-analyses of the CARDIA study tried to identify hormonal predictors of CAC.88,89 In a sample of 1629 women who were followed up for 20 years,89 the incidence of CAC was inversely associated with values of SHBG above rather than below the median (adjusted OR = 0.59, 95% CI: 0.40–0.86, p value = 0.008), but not with free or total testosterone. More recently, in another subanalysis of the CARDIA study,88 the incidence of CAC was found to be associated with values of the cortisol to free testosterone ratio within the highest tertile versus the lowest tertile (OR = 3.45, 95% CI: 1.18–10.06). Overall, evidence retrieved from longitudinal studies shows a protective effect of higher SHBG levels regarding the CAC progression. Evidence on the effect of testosterone levels with regards to CAC risk and progression is largely inconsistent.

Clinical CVD

Data on cohort studies assessing the possible associations between endogenous androgens and prevalent or incident rates of CVD is presented in Table 4.

Table 4.

Cohort studies exploring associations between endogenous androgens and cardiovascular events as well as all-cause mortality.

Ref. Study sample and design Primary outcome FU Results Discussion
Clinical cardiovascular disease
Di et al.93 Multi-ethnic study of atherosclerosis
N = 2834 postmenopausal women CVD-free at baseline
Design: L
CV risk Median 12.1 years Incident CV risk, per 1 SD of log-T/E2 ratio:
• For CVD, HR 1.19 (95% CI, 1.02–1.40)
• For CHD, HR 1.45 (95% CI, 1.19–1.78)
• For HF, HR 1.31 (95% CI, 1.01–1.70)
(↑) Log-T/E2 ratio was associated with (↑) incidence of CVD, CHD and HF
Zhao et al.94 Atherosclerosis risk in communities (ARIC) study
N = 4839 postmenopausal women, mean age 62.8 ± 5.5 years and 4107 men
Design: L
HF-risk Median
19.2 years
Incident HF events and log-transformed
• TT, HR 1.05 (95% CI: 0.99–1.13)
• DHEAS, HR 1.17 (95% CI: 1.09–1.24)
• SHBG, HR 0.93 (95% CI: 0.85–1.01)
(↑) HF incidence was associated with (↑) TT, DHEAS and (↓) SHBG
Wang et al.95 UK Biobank
N = 154,965 men and N = 93,314 postmenopausal women
Design: L
All-cause mortality
Cancer mortality
Median 8.9 years Levels of TT and:
• All-cause mortality (HR for Q5 vs Q1: 1.20; 95% CI: 1.06–1.37)
• Cancer mortality (p value 0.03)
(↑) TT was associated with (↑) All-cause- and cancer-related mortality
Jia et al.96 The atherosclerosis risk in communities study
N = 8143 individuals (3,650 men and 4,493 women) with prevalent CVD, mean age of 63 years
Design: L
Incident risk for HF-hospitalization or death in women DHEAS levels:
18 years
DHEAS change:
5.5 years
HF-hospitalization in women, DHEAS < 27.4 μg/dL (15th sex specific percentile): HR = 1.42, 95% CI: 1.13–1.179
Risk of death, DHEAS < 37.1 μg/dL (25th sex-specific percentile): HR = 1.19, 95% CI: 1.03–1.37
(↓) DHEAS was associated with:
• (↑) Risk for HF-hospitalization
• (↑) Risk of death
Schederecker et al.97 KORA F4 study
N = 1006 men and N = 709 peri- and postmenopausal women from a German population-based study
Design: L
All-cause or other disease-related mortality Median
8.7 years
All-cause mortality in women,
• SHBG, HR = 1.54, 95% CI: 1.16–2.04
• DHT, HR = 1.32, 95% CI: 1.00–1.73
Other disease-related mortality in women
• SHBG, HR = 1.86, 95% CI: 1.08–3.20
All-cause mortality was associated with (↑) SHBG and (↑) DHT.
Other disease-related mortality was associated with (↑) SHBG
Schaffrath et al.98 Study of Health in Pomerania
2129 middle-aged women with a mean age of 49 years (baseline)
Design: L
Incident CVD risk Median
10.9 years
Incident CVD and baseline sex hormones (multivariable adj models)
• TT, RR = 1.00 (95% CI: 0.96–1.06)
• SHBG, RR = 1.02 (95% CI: 0.97–1.07)
• Androstenedione, RR = 1.07 (95% CI: 0.93 to 1.09)
• FT, RR = 0.99 (95% CI: 0.95–1.05)
• FAI, RR = 0.99 (95% CI: 0.94–1.05)
Future CVD risk was not associated with baseline TT, SHBG, androstenedione, FT or FAI levels
Daka et al.99 N = 1109 men and women, aged at least 40 years (mean age 62 ± 12 years), with T2DM at baseline
Design: L
MI-risk Mean 14.1 ± 5.3 years MI–risk per change of FT levels (by 0.01 mmol/L), multivariable adjusted:
all subjects, HR 0.722, 95% CI: 0.52–1.00, p value = 0.046
T2DM, HR 0.579, 95% CI: 0.26–1.31
(↑) FT levels were associated with (↓) MI-risk in females
No association between MI-risk and androgens for T2DM females
Holmegard et al.100 Copenhagen City Heart Study
N = 4724 pre and perimenopausal women, N = 4615 men
Design: L and meta-analysis
Incident risk for ischemic stroke Median
29 years
Ischemic stroke according to T levels
T ⩽ 10th percentile vs 11th–90th percentile,
• premenopausal: HR 0.93, 95% CI: 0.28–3.08
• Postmenopausal, HR 0.98, 95% CI: 0.72–1.33
T > 91st to 100th percentile vs 11–90th percentile,
• Premenopausal women: HR 1.26, 95% CI: 0.48–3.27;
• Postmenopausal women: HR 1.22, 95% CI: 0.90–1.66
Incident risk for ischemic stroke was not associated with extremes of T levels
Laughlin et al.101 639 postmenopausal women (mean age 73.8 years)
Design: L
Incident risk for first time CHD Median
12.3 years
Incident first time CHD and TT, ⩽80 pg/mL vs > 80 pg/mL, HR 1.62, 95% CI: 1.10–2.39
Incident CHD and bioavailable T,
• 1st vs 3rd quintile: HR = 1.79, 95% CI: 1.01–3.16
• 5th vs 3rd quintile: HR = 1.96, 95% CI: 1.13–3.16
Incident risk for first time CHD was associated with extreme levels of bioavailable T (U-shaped association)
Wehr et al.102 N = 875 postmenopausal diabetic women
Design: L
Risk for all-cause mortality and CV-mortality Median
7.7 years
Mortality per FT Q4 vs Q1, multivariable adjusted:
• All-cause mortality, HR = 0.38, 95% CI: 0.08–0.90, p value = 0.025;
• CV-mortality, HR = 0.28, 95% CI: 0.08–0.90, p value = 0.032
(↓) FT was associated with (↑) all-cause and CV–mortality
Benn et al.103 Copenhagen City Heart Study
Nested study (N = 4,716 women) not on HRT or OC
Design: L
Risk for IHD or any death Max. ⩽ 30 y T > 95th percentile vs 10th–89th percentile (multifactorial adjusted)
• IHD, +68%, 95% CI: 34%–210%
• Any death, +36%, 95% CI: 18%–58%
(↑) T was associated with (↑) risk for IHD or risk for death of any cause

CHD, coronary heart disease; CV, cardiovascular; CVD, cardiovascular disease; DHEAS, dehydroepiandrosterone; DHT, dihydrotestosterone; FT, free testosterone; FU, follow-up; HF, heart failure; HR, hazard ratio; HRT, hormone replacement therapy; IHD, ischemic heart disease; MI, myocardial infarction; OC, oral contraceptives; RR, relative risk; SD, standard deviation; T, testosterone; T/E2, testosterone to oestrogen ratio; TT, total testosterone.

Five of the available cohort studies support a direct association between a more androgenic profile and cardiovascular and/or cerebrovascular events. The possible link between serum levels of androgens and clinical CVD has been investigated in 2834 postmenopausal women from the MESA.93 This study showed that the risk for future events associated with more pronounced androgenicity, reflected by the testosterone to oestrogen ratio as estimated at 12 years of follow-up (incident CVD, HR = 1.19, 95% CI: 1.02–1.40; incident CHD, HR = 1.45, 95% CI: 1.19–1.78; incident HF, HR = 1.31, 95% CI: 1.01–1.70). Similar results were presented by the Atherosclerosis Risk in Communities study (ARIC),94 which showed that the risk for incident HF was associated with higher levels of DHEAS (HR = 1.17, 95% CI: 1.09–1.24), but not with total testosterone (HR = 1.05, 95% CI: 0.99–1.13) or SHBG (HR = 0.93, 95% CI: 0.85–1.01). The UK biobank analysis 95 provided valuable insight into possible hormonal predictors of all-cause mortality and described in a total of 93,314 postmenopausal women that higher all-cause mortality was associated with higher total testosterone (HR for Q5 vs Q1: 1.20; 95% CI: 1.06–1.37). The German population-based study (KORA F4) evaluated 709 peri- and postmenopausal women as well as 1006 men for up to 8.7 years, aiming to describe possible links between serum androgens and mortality.97 This study concluded that female-specific all-cause mortality was associated with higher levels of SHBG and dihydrotestosterone (SHBG, HR = 1.54, 95% CI: 1.16–2.04; dihydrotestosterone, HR = 1.32, 95% CI: 1.00–1.73), while another disease-related mortality was associated with lower androgenicity (SHBG, HR = 1.86, 95% CI: 1.08–3.20). On the contrary, a nested prospective study of 4716 women not on oral contraceptives or hormone replacement therapy 103 reported that higher testosterone was associated with increased risk of ischemic heart disease or death.

The observed positive association between higher androgen levels and increased risk of CVD may be mediated by insulin resistance. Menopause and ageing are associated with lower levels of SHBG, which is a marker of both insulin resistance and of increased androgenicity104,105

On the contrary, three studies support an inverse association between levels of circulating androgens and the risk for cardiovascular and/or cerebrovascular events. One more analysis of the ARIC study96 demonstrated that the possible link between DHEAS and risk for hospitalizations due to heart failure or the risk of death was evaluated in a total of 8143 individuals with prevalent CVD. The incident risk for hospitalization due to HF was higher in women with DHEAS < 15th sex-specific percentile (HR = 1.42, 95% CI: 1.13–1.17).96 Similarly, the risk of death was higher for women with levels of DHEAS < 25th sex-specific percentile (HR = 1.19, 95% CI: 1.03–1.37).96 Investigating a sample of 1109 men and women aged at least 40 years, one more study reported a borderline significant association between low androgenicity and increasing risk of acute myocardial infarction, at least in women with type 2 diabetes,99 adjusting for cardiovascular risk factors. Similarly, a study of 875 postmenopausal women followed up for up to 7.7 years reported an inverse association between high free testosterone and low cardiovascular and all-cause mortality in postmenopausal women diagnosed with diabetes mellitus.102

Different results were presented in three more cohort studies. Interestingly, an earlier study101 described a U-shaped association between levels of testosterone and coronary heart disease in a sample of 639 postmenopausal women, followed up for up to 12.3 years. In conclusion, data retrieved from cohort studies seems to be supportive of a direct link between more pronounced androgenicity and the risk for clinically evident CVD. Finally, two cohort studies of 2129 middle-aged women98 and 4724 pre- and perimenopausal women100 could not find longitudinal associations between circulating androgens and clinically evident CVD.

Exogenous androgens and CVD

Trials of testosterone replacement in middle-aged women

Data retrieved from trials of testosterone replacement in middle-aged women are presented in Table 5. In an early trial,106 administration of a testosterone implant of 50 mg for a total of 6 weeks in postmenopausal women already on HRT described that testosterone administration resulted in improvement of endothelium-dependent and independent vasodilation (increase in FMD, 6.4% ± 0.7% to 9.1% ± 1.1, p value = 0.003; GTN induced, 14.9% ± 0.9 to 17.8 ± 1.2, p value = 0.03). One more trial107 estimated the impact of intramuscular hormone replacement with 2–5 mg of estradiol esters and 50–100 mg testosterone esters in postmenopausal women compared to never users of HRT. The study107 concluded that combined estradiol and testosterone treatment for at least 1 year was associated with a significantly higher risk of severe aortic calcification (OR = 3.1, 95% CI: 1.1–8.5). Penotti et al.108 described the effect on arterial resistance of treatment with testosterone undecanoate 40 mg/day in combination with sequential HRT for 8 months as opposed to HRT only in a small sample of postmenopausal women. The combined oestrogen and testosterone administration resulted in an increase in the pulsatility index of the middle cerebral artery as opposed to baseline. The effect of the oestrogen-testosterone combination was also significantly higher than the equivalent pulsatility index estimated in HRT-only users. One more intervention trial69 investigated the possible effect of DHEA administration of 100 mg per day in 36 healthy postmenopausal women compared to placebo and concluded that DHEA treatment was related to significant improvement in FMD values (intervention group, 8.4% ± 0.7% to 14.5%±1.1, p value < 0.05; placebo, 10.8% ± 1.1% to 10.9% ± 0.6, p value = NS), but had no effect on the GTN-induced vasodilation.

Table 5.

Studies exploring the possible associations between exogenous androgens and vascular disease.

Ref. Study sample and Design Treatment Primary outcome FU Results Discussion
Testosterone treatment
Worboys et al.106 postmenopausal women,
Cases (N = 33) HRT-users (>6 months) who also received T-treatment
Controls (N = 15) not on HRT
E2 ± progestin,
Exogenous T implant 50 mg
FMD
GTN-induced vasodilation
6 weeks Change in FMD from baseline:
• Cases, 6.4% ± 0.7% to 9.1% ± 1.1, p value = 0.003
• Control, 8.1% ± 1.4% to 5.6% ± 1.0, p value = 0.4
• Between group difference in FMD, p value = 0.02
GTN-induced vasodilation:
• Cases, 14.9% ± 0.9 vs 17.8% ± 1.2, p value = 0.03
T-treatment was associated with (↑) endothelium-dependent and independent vasodilation
Hak et al.107 Population-based study (N = 513) naturally postmenopausal women aged 54–67y;
Cases: hormone therapy users, Controls: never-users of hormone therapy
IM 2–5 mg estradiol esters,
IM 50–100 mg testosterone esters
Aortic atherosclerosis Min. 1 y Cases vs controls, adjusted for CV risk factors
• severe aortic atherosclerosis, OR = 3.1, 95% CI: 1.1–8.5
• mild aortic atherosclerosis, OR = 1.5, 95% CI: 0.5–4.6)
Hormone therapy use was associated with (↑) risk for aortic atherosclerosis
Penotti et al.108 Healthy postmenopausal women;
Cases: sequential HRT-users (N = 20), sequential HRT-users randomized to T-treatment (N = 20);
Controls: non-users of HRT (N = 20)
transdermal E2 50 μg/d + MPA 10 mg/d for 12 days every other month
Testosterone undecanoate 40 mg/day
PI index of middle cerebral artery 8 m Only HRT treatment, no significant difference between baseline vs 4 months vs 8 months
Combined HRT + T treatment,
• at 8 months vs baseline, 0.822 (95% CI: 0.740–1.065) vs 0.780 (95% CI: 0.6950.955), p value < 0.001
• at 8 months vs HRT-only, 0.822 (95% CI: 0.740 to 1.065) vs 0.777 (95% CI: 0.700–0.965), p value < 0.05
HRT + T-treatment was associated with a significant increase in the PI of the middle cerebral artery
Williams et al.69 Healthy postmenopausal women vs placebo
Cases: DHEA-treatment (N = 18)
Controls: placebo treatment (N = 18)
DHEA 100 mg/day or placebo FMD
GTN-induced vasodilation
3 m FMD:
• Cases: 8.4 ± 0.7% to 14.5 ± 1.1%, p value < 0.05
• Controls: 10.8 ± 1.1% to 10.9 ± 0.6%, p value = NS
GTN-induced vasodilation
• Cases: 16.4 ± 1.2% to 18.5 ± 1.1%, p value = NS
• Controls 19.2 ± 1.3% to 18.4 ± 1.4%, p value = NS
DHEAS treatment vs placebo was associated with (↑) FMD but had no effect on GTN induced vasodilation

CI, confidence interval; CV, cardiovascular; DHEA(S), dehydroepiandrosterone (sulphate); E + T, oestrogen and testosterone; FMD, flow-mediated dilation; FU, follow-up; GTN, glyceryl trinitrate; HRT, hormone replacement therapy; IM, intramuscular; m, months; MPA, medoxyprogesterone acetate; NS, not significant; OR, odds ratio; PI, pulsatility index; T, testosterone.

Data on transgender people

Insight into the possible cardiovascular implications of testosterone treatment can be gained from studies on female-to-male transgender. The behavioural risk factor surveillance system (BRSS, 2014–2017)109 evaluated the risk of myocardial infarction in a large sample of transgender men and women as well as cisgender women (transgender men and cisgender women 410,828; transgender women 1788, cisgender men 306,046). This analysis109 showed that female-to-male transgenders had higher rates of myocardial infarction compared with cisgender men (OR 2.53, 95% CI: 1.14–5.63, p value = 0.02) and also compared with cisgender women (OR 4.9, 95% CI: 2.21–10.90, p value < 0.01). Additional data retrieved from the BRFSS analysis110 showed that the elevated crude risk for myocardial infarction observed in transgender men as opposed to cisgender women is largely affected by confounders and highlighted differences in health behaviours between female-to-male transgenders and cisgender women. Transgenders are more frequently overweight (female-to-male vs cisgender women, adjusted odds ratio 1.54, 95% CI: 1.07–2.24), smokers (female-to-male vs cisgender women, OR 1.64, 95% CI: 1.17–2.31), with likely lower educational attainment (combined rates, 30.5% high school graduates; 13.3% never graduated from high-school) and likely to face challenges on employment (5.8% unemployed and 5.8% not able to work).110 A recent study evaluated the incidence of cardiovascular events in a large transgender cohort,111 consisting of 2517 male-to-female and 1358 female-to-male (median age of 30 years and 23 years, respectively). Results in male-to-female transgender showed that the standardized incident ratios (SIR) for the development of acute cardiovascular events were higher for stroke, myocardial infarction, and venous thromboembolism (VTE) as opposed to the equivalent risk for cis women (standardized incidence ratio, stroke: 2.42, 95% CI: 1.65–3.42; MI, 2.64, 95% CI: 1.81–3.72; VTE, 5.52, 95% CI: 4.36–6.90).111 A recent observational analysis of 114 transgender and 964 cisgender individuals112 described a higher risk of VTE in male-to-female transgenders compared to cis women (adjusted OR 3.94, 95% CI: 1.24–12.51), but comparable rates of any CVD condition in transgender individuals vs cis women or cis men. A prospective cohort study assessed 2842 male-to-female transgenders (followed up for 4 years) and 2118 female-to-male transgenders (followed up for 3.6 years) and compared the cohort with a matched group of 48,686 cisgender men and 48,775 cisgender women.113 The results showed113 that the rates of VTE, ischemic stroke or myocardial infarction did not differ between the female-to-male transgenders and the cisgender women (adjusted HR 1.1, 95% CI: 0.6–2.1; 1.3, 95% CI: 0.7–2.5; 1.3, 95% CI: 0.5–3.9; for VTE, stroke and MI, respectively). An earlier meta-analysis of 16 studies, including 1471 male-to-female and 652 female-to-male individuals, showed a low incidence of cardiovascular events in the female-to-male transgender group (death, two cases; myocardial infarction, two cases; VTE, two cases; stroke, no case).114

Data on the cardiovascular implications of female-to-male transgenders remains limited, and a detailed review of studies on the topic is outside the scope of this study, mainly due to the methodological differences between women on gender-affirming hormone treatment and the peri- or postmenopausal population. The majority of studies on female-to-male transgenders were observational and usually consisted of a small sample size of younger individuals, while the effect of health behaviour possibly modifying the overall cardiovascular risk was not always accounted for. A recent meta-analysis115 consisting of 29 studies with moderate risk of bias reported that sex hormone treatment in transgender men is usually accompanied by higher rates of dyslipidaemia (at ⩾24 months, mean difference, low-density lipoprotein (LDL) cholesterol, +17.8 mg/dL; 95% CI: 3.5–32.1; serum triglycerides, +21.4 mg/dL, 95% CI: 0.14–42.6). These observations were also supported by a systematic review of 13 studies,116 which described the metabolic effects of parenteral testosterone treatment (undecanoate 1000 mg per 12 weeks) for up to 60 months in transgender men. The same study116 reported that transgender men on testosterone treatment have consistently higher levels of LDL-cholesterol and also increased body mass index (from 1.3% to 11.4%). In any case, data on the effect of testosterone treatment in transgender individuals should not be considered directly comparable with the effect of lower-dose testosterone replacement in middle-aged women, considering the difference in age, health behavior, and overall metabolic burden.

Conclusion

Data on the possible effect of testosterone on cardiovascular cells and tissues remain contradictory. Preclinical studies highlight a possibly beneficial effect on vascular cell lines, which fails to be translated into an actual clinical benefit. The results of a recent study on cardiomyocytes58 and human umbilical vein endothelial cells59 have revealed that the optimal oestrogen-to-testosterone ratio is ideal, as sex hormones at this level have been found to reduce apoptosis of cardiac myocytes and improve the overall metabolism. Even though the idea of an ‘optimal’ oestrogen-to-testosterone ratio sounds promising, this balance is extremely difficult to be achieved in clinical praxis due to the existence of other counterregulatory factors, which are not present at the preclinical setting. In this context, factors like weight gain and adipose tissue,117 metabolic syndrome, or even insulin resistance104 and diabetes mellitus are pathophysiologically interrelated with the oestrogen-to-testosterone ratio and, therefore, likely to modify the effect of sex hormones on the cardiovascular system.

This narrative review summarizes the interplay between the hormonal alterations of the menopausal transition and the cardiovascular system, focusing mainly on androgens. Data on the possible effect of testosterone on cardiovascular cells and tissues remain contradictory. Also, preclinical studies highlight a possible beneficial effect on vascular cell lines, which fails to be translated into an actual clinical benefit. Clinical studies exploring the associations between endogenous androgens and subclinical vascular disease or clinically evident cardiovascular events show an increased risk in women with more pronounced androgenicity, though a possible mediatory effect of insulin resistance cannot be ruled out. On the contrary, several clinical studies describe neutral or inconclusive participation of total and/or free testosterone on cardiovascular endpoints. The idea of an ‘optimal’ oestrogen-to-testosterone ratio sounds promising. Yet this balance is extremely difficult to be achieved in clinical praxis due to the existence of other counterregulatory factors, which are not present at the preclinical setting. In this context, factors like weight gain and adipose tissue, metabolic syndrome or even insulin resistance and diabetes mellitus are pathophysiologically interrelated with the oestrogen-to-testosterone ratio and are, therefore, likely to modify the effect of sex hormones on the cardiovascular system. Evidence on the effect of testosterone treatment remains insufficient to draw firm conclusions. In any case, cardiovascular risk stratification should be incorporated in the management of postmenopausal women, especially when indications for testosterone treatment are present, to minimize the possible future cardiovascular risk.

Acknowledgments

None.

Footnotes

ORCID iD: Irene Lambrinoudaki Inline graphic https://orcid.org/0000-0003-1488-2668

Contributor Information

Eleni Armeni, Second Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Ave., GR-11528, Athens, Greece.

Irene Lambrinoudaki, Second Department of Obstetrics and Gynecology, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece.

Declarations

Ethics approval and consent to participate: Not applicable.

Consent for publication: Not applicable.

Author contributions: Eleni Armeni: Conceptualization; Data curation; Investigation; Methodology; Visualization; Writing – original draft.

Irene Lambrinoudaki: Conceptualization; Project administration; Validation; Visualization; Writing – review & editing.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Availability of data and materials: Not applicable

References

  • 1. Benjamin EJ, Muntner P, Alonso A, et al. Heart disease and stroke statistics-2019 update: a report from the American Heart Association. Circulation 2019; 139: e56–e528. [DOI] [PubMed] [Google Scholar]
  • 2. Virani SS, Alonso A, Aparicio HJ, et al. Heart disease and stroke statistics-2021 update: a report from the American Heart Association. Circulation 2021; 143: e254–e743. [DOI] [PubMed] [Google Scholar]
  • 3. Walli-Attaei M, Joseph P, Rosengren A, et al. Variations between women and men in risk factors, treatments, cardiovascular disease incidence, and death in 27 high-income, middle-income, and low-income countries (PURE): a prospective cohort study. Lancet 2020; 396: 97–109. [DOI] [PubMed] [Google Scholar]
  • 4. Kannel WB, Hjortland MC, McNamara PM, et al. Menopause and risk of cardiovascular disease: the Framingham study. Ann Intern Med 1976; 85: 447–452. [DOI] [PubMed] [Google Scholar]
  • 5. El Khoudary SR, Aggarwal B, Beckie TM, et al. Menopause transition and cardiovascular disease risk: implications for timing of early prevention: a scientific statement from the American Heart Association. Circulation 2020; 142: e506–e532. [DOI] [PubMed] [Google Scholar]
  • 6. Zhu D, Chung H-F, Dobson AJ, et al. Type of menopause, age of menopause and variations in the risk of incident cardiovascular disease: pooled analysis of individual data from 10 international studies. Hum Reprod 2020; 35: 1933–1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Muka T, Oliver-Williams C, Kunutsor S, et al. Association of age at onset of menopause and time since onset of menopause with cardiovascular outcomes, intermediate vascular traits, and all-cause mortality: a systematic review and meta-analysis. JAMA Cardiol 2016; 1: 767–776. [DOI] [PubMed] [Google Scholar]
  • 8. Connelly PJ, Azizi Z, Alipour P, et al. The importance of gender to understand sex differences in cardiovascular disease. Can J Cardiol 2021; 37: 699–710. [DOI] [PubMed] [Google Scholar]
  • 9. Karavidas A, Lazaros G, Tsiachris D, et al. Aging and the cardiovascular system. Hellenic J Cardiol 2010; 51: 421–427. [PubMed] [Google Scholar]
  • 10. McEniery CM, Wilkinson IB, Avolio AP. Age, hypertension and arterial function. Clin Exp Pharmacol Physiol 2007; 34: 665–671. [DOI] [PubMed] [Google Scholar]
  • 11. Liguori I, Russo G, Curcio F, et al. Oxidative stress, aging, and diseases. Clin Interv Aging 2018; 13: 757–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Cosentino F, Francia P, Camici GG, et al. Final common molecular pathways of aging and cardiovascular disease: role of the p66Shc protein. Arterioscler Thromb Vasc Biol 2008; 28: 622–628. [DOI] [PubMed] [Google Scholar]
  • 13. Papaconstantinou J. The role of signaling pathways of inflammation and oxidative stress in development of senescence and aging phenotypes in cardiovascular disease. Cells 2019; 8: 1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Kaasik A, Kuum M, Joubert F, et al. Mitochondria as a source of mechanical signals in cardiomyocytes. Cardiovasc Res 2010; 87: 83–91. [DOI] [PubMed] [Google Scholar]
  • 15. North BJ, Sinclair DA. The intersection between aging and cardiovascular disease. Circ Res 2012; 110: 1097–1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Izzo C, Carrizzo A, Alfano A, et al. The impact of aging on cardio and cerebrovascular diseases. Int J Mol Sci 2018; 19: 481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Paneni F, Diaz Cañestro C, Libby P, et al. The aging cardiovascular system: understanding it at the cellular and clinical levels. J Am Coll Cardiol 2017; 69: 1952–1967. [DOI] [PubMed] [Google Scholar]
  • 18. Pourbagher-Shahri AM, Farkhondeh T, Talebi M, et al. An overview of NO signaling pathways in aging. Molecules 2021; 26: 4533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Davis SR, Lambrinoudaki I, Lumsden M, et al. Menopause. Nat Rev Dis Prim 2015; 1: 15004. [DOI] [PubMed] [Google Scholar]
  • 20. Burger HG. Androgen production in women. Fertil Steril 2002; 77(Suppl. 4): S3–S5. [DOI] [PubMed] [Google Scholar]
  • 21. Monteleone P, Mascagni G, Giannini A, et al. Symptoms of menopause – global prevalence, physiology and implications. Nat Rev Endocrinol 2018; 14: 199–215. [DOI] [PubMed] [Google Scholar]
  • 22. Lambrinoudaki I, Paschou SA, Lumsden MA, et al. Premature ovarian insufficiency: a toolkit for the primary care physician. Maturitas 2021; 147: 53–63. [DOI] [PubMed] [Google Scholar]
  • 23. Hammond GL. Plasma steroid-binding proteins: primary gatekeepers of steroid hormone action. J Endocrinol 2016; 230: R13–R25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Laurent MR, Helsen C, Antonio L, et al. Effects of sex hormone-binding globulin (SHBG) on androgen bioactivity in vitro. Mol Cell Endocrinol 2016; 437: 280–291. [DOI] [PubMed] [Google Scholar]
  • 25. Sutton-Tyrrell K, Zhao X, Santoro N, et al. Reproductive hormones and obesity: 9 years of observation from the Study of Women’s Health Across the Nation. Am J Epidemiol 2010; 171: 1203–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Bucciarelli P, Mannucci PM. The hemostatic system through aging and menopause. Climacteric 2009; 12(Suppl. 1): 47–51. [DOI] [PubMed] [Google Scholar]
  • 27. Barbieri RL, Smith S, Ryan KJ. The role of hyperinsulinemia in the pathogenesis of ovarian hyperandrogenism. Fertil Steril 1988; 50: 197–212. [DOI] [PubMed] [Google Scholar]
  • 28. Karvonen-Gutierrez C, Kim C. Association of mid-life changes in body size, body composition and obesity status with the menopausal transition. Healthc 2016; 4: 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. El Khoudary SR, Shields KJ, Chen HY, et al. Menopause, complement, and hemostatic markers in women at midlife: the Study of Women’s Health Across the Nation. Atherosclerosis 2013; 231: 54–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Wang NC, Matthews KA, Barinas-Mitchell EJ, et al. Inflammatory/hemostatic biomarkers and coronary artery calcification in midlife women of African-American and White race/ethnicity: the Study of Women’s Health Across the Nation (SWAN) heart study. Menopause 2016; 23: 653–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Gaspard U. Hyperinsulinaemia, a key factor of the metabolic syndrome in postmenopausal women. Maturitas 2009; 62: 362–365. [DOI] [PubMed] [Google Scholar]
  • 32. Schmiegelow MD, Hedlin H, Mackey RH, et al. Race and ethnicity, obesity, metabolic health, and risk of cardiovascular disease in postmenopausal women. J Am Heart Assoc 2015; 4: e001695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Anklam CFV, Lissarassa YPS, Dos Santos AB, et al. Oxidative and cellular stress markers in postmenopause women with diabetes: the impact of years of menopause. J Diabetes Res 2021; 2021: 3314871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Pou KM, Massaro JM, Hoffmann U, et al. Visceral and subcutaneous adipose tissue volumes are cross-sectionally related to markers of inflammation and oxidative stress: the Framingham Heart Study. Circulation 2007; 116: 1234–1241. [DOI] [PubMed] [Google Scholar]
  • 35. Incalza MA, D’Oria R, Natalicchio A, et al. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul Pharmacol 2018; 100: 1–19. [DOI] [PubMed] [Google Scholar]
  • 36. Gimbrone MAJ, García-Cardeña G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res 2016; 118: 620–636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Stanhewicz AE, Wenner MM, Stachenfeld NS. Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. Am J Physiol Heart Circ Physiol 2018; 315: H1569–H1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Donato AJ, Morgan RG, Walker AE, et al. Cellular and molecular biology of aging endothelial cells. J Mol Cell Cardiol 2015; 89(Pt. B): 122–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Briasoulis A, Tousoulis D, Androulakis ES, et al. Endothelial dysfunction and atherosclerosis: focus on novel therapeutic approaches. Recent Pat Cardiovasc Drug Discov 2012; 7: 21–32. [DOI] [PubMed] [Google Scholar]
  • 40. Takov K, Wu J, Denvir MA, et al. The role of androgen receptors in atherosclerosis. Mol Cell Endocrinol 2018; 465: 82–91. [DOI] [PubMed] [Google Scholar]
  • 41. Herrera-Zelada N, Zuñiga-Cuevas U, Ramirez-Reyes A, et al. Targeting the endothelium to achieve cardioprotection. Front Pharmacol 2021; 12: 636134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Arapa-Diaz JC, Rouver WDN, Giesen JAS, et al. Testosterone increases bradykinin-induced relaxation in the coronary bed of hypertensive rats. J Mol Endocrinol 2020; 65: 125–134. [DOI] [PubMed] [Google Scholar]
  • 43. Campelo AE, Cutini PH, Massheimer VL. Testosterone modulates platelet aggregation and endothelial cell growth through nitric oxide pathway. J Endocrinol 2012; 213: 77–87. [DOI] [PubMed] [Google Scholar]
  • 44. Goglia L, Tosi V, Sanchez AM, et al. Endothelial regulation of eNOS, PAI-1 and t-PA by testosterone and dihydrotestosterone in vitro and in vivo. Mol Hum Reprod 2010; 16: 761–769. [DOI] [PubMed] [Google Scholar]
  • 45. Sieveking DP, Lim P, Chow RWY, et al. A sex-specific role for androgens in angiogenesis. J Exp Med 2010; 207: 345–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Alves JV, da Costa RM, Pereira CA, et al. Supraphysiological levels of testosterone induce vascular dysfunction via activation of the NLRP3 inflammasome. Front Immunol 2020; 11: 1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Montaño LM, Calixto E, Figueroa A, et al. Relaxation of androgens on rat thoracic aorta: testosterone concentration dependent agonist/antagonist L-type Ca2+ channel activity, and 5beta-dihydrotestosterone restricted to L-type Ca2+ channel blockade. Endocrinology 2008; 149: 2517–2526. [DOI] [PubMed] [Google Scholar]
  • 48. Chignalia AZ, Schuldt EZ, Camargo LL, et al. Testosterone induces vascular smooth muscle cell migration by NADPH oxidase and c-Src-dependent pathways. Hypertension 2012; 59: 1263–1271 [DOI] [PubMed] [Google Scholar]
  • 49. Lopes RAM, Neves KB, Pestana CR, et al. Testosterone induces apoptosis in vascular smooth muscle cells via extrinsic apoptotic pathway with mitochondria-generated reactive oxygen species involvement. Am J Physiol Heart Circ Physiol 2014; 306: H1485–H1494. [DOI] [PubMed] [Google Scholar]
  • 50. Zhu D, Hadoke PWF, Wu J, et al. Ablation of the androgen receptor from vascular smooth muscle cells demonstrates a role for testosterone in vascular calcification. Sci Rep 2016; 6: 24807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Son B-K, Akishita M, Iijima K, et al. Androgen receptor-dependent transactivation of growth arrest-specific gene 6 mediates inhibitory effects of testosterone on vascular calcification. J Biol Chem 2010; 285: 7537–7544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Xiao F-Y, Nheu L, Komesaroff P, et al. Testosterone protects cardiac myocytes from superoxide injury via NF-κB signalling pathways. Life Sci 2015; 133: 45–52. [DOI] [PubMed] [Google Scholar]
  • 53. Vicencio JM, Ibarra C, Estrada M, et al. Testosterone induces an intracellular calcium increase by a nongenomic mechanism in cultured rat cardiac myocytes. Endocrinology 2006; 147: 1386–1395. [DOI] [PubMed] [Google Scholar]
  • 54. Akdis D, Saguner AM, Shah K, et al. Sex hormones affect outcome in arrhythmogenic right ventricular cardiomyopathy/dysplasia: from a stem cell derived cardiomyocyte-based model to clinical biomarkers of disease outcome. Eur Heart J 2017; 38: 1498–1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Zhang L, Wu S, Ruan Y, et al. Testosterone suppresses oxidative stress via androgen receptor-independent pathway in murine cardiomyocytes. Mol Med Rep 2011; 4: 1183–1188. [DOI] [PubMed] [Google Scholar]
  • 56. Costa TJ, Ceravolo GS, dos Santos RA, et al. Association of testosterone with estrogen abolishes the beneficial effects of estrogen treatment by increasing ROS generation in aorta endothelial cells. Am J Physiol Hear Circ Physiol 2015; 308: H723–H732. [DOI] [PubMed] [Google Scholar]
  • 57. Costa TJ, Ceravolo GS, Echem C, et al. Detrimental effects of testosterone addition to estrogen therapy involve cytochrome P-450-induced 20-HETE synthesis in aorta of ovariectomized spontaneously hypertensive rat (SHR), a model of postmenopausal hypertension. Front Physiol 2018; 9: 490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Xu S, Dai W, Li J, et al. Synergistic effect of estradiol and testosterone protects against IL-6-inducedcardiomyocyte apoptosismediated by TGF-β1. Int J Clin Exp Pathol 2018; 11: 10–26. [PMC free article] [PubMed] [Google Scholar]
  • 59. Dai W, Ming W, Li Y, et al. Synergistic effect of a physiological ratio of estradiol and testosterone in the treatment of early-stage atherosclerosis. Arch Med Res 2015; 46: 619–629. [DOI] [PubMed] [Google Scholar]
  • 60. Cruz-Topete D, Dominic P, Stokes KY. Uncovering sex-specific mechanisms of action of testosterone and redox balance. Redox Biology 2020; 31: 101490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Gündoğan GI., Kıg C, Karacan M, et al. Investigation of physiological effects induced by dehydroepiandrosterone in human endothelial cells and ovarian cancer cell line. Turkish J Pharm Sci 2021; 18: 185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Altman R, Motton DD, Kota RS, et al. Inhibition of vascular inflammation by dehydroepiandrosterone sulfate in human aortic endothelial cells: roles of PPARalpha and NF-kappaB. Vascul Pharmacol 2008; 48: 76–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Curatola AM, Huang K, Naftolin F. Dehydroepiandrosterone (DHEA) inhibition of monocyte binding by vascular endothelium is associated with sialylation of neural cell adhesion molecule. Reprod Sci 2012; 19: 86–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Liu D, Si H, Reynolds KA, et al. Dehydroepiandrosterone protects vascular endothelial cells against apoptosis through a Galphai protein-dependent activation of phosphatidylinositol 3-kinase/Akt and regulation of antiapoptotic Bcl-2 expression. Endocrinology 2007; 148: 3068–3076. [DOI] [PubMed] [Google Scholar]
  • 65. Huerta-García E, Montiél-Dávalos A, Alfaro-Moreno E, et al. Dehydroepiandrosterone protects endothelial cells against inflammatory events induced by urban particulate matter and titanium dioxide nanoparticles. Biomed Res Int 2013; 2013: 382058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Huerta-García E, Ventura-Gallegos JL, Victoriano ME, et al. Dehydroepiandrosterone inhibits the activation and dysfunction of endothelial cells induced by high glucose concentration. Steroids 2012; 77: 233–240. [DOI] [PubMed] [Google Scholar]
  • 67. Liu D, Iruthayanathan M, Homan LL, et al. Dehydroepiandrosterone stimulates endothelial proliferation and angiogenesis through extracellular signal-regulated kinase 1/2-mediated mechanisms. Endocrinology 2008; 149: 889–898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wang L, Hao Q, Wang YD, et al. Protective effects of dehydroepiandrosterone on atherosclerosis in ovariectomized rabbits via alleviating inflammatory injury in endothelial cells. Atherosclerosis 2011; 214: 47–57. [DOI] [PubMed] [Google Scholar]
  • 69. Williams MR, Dawood T, Ling S, et al. Dehydroepiandrosterone increases endothelial cell proliferation in vitro and improves endothelial function in vivo by mechanisms independent of androgen and estrogen receptors. J Clin Endocrinol Metab 2004; 89: 4708–4715. [DOI] [PubMed] [Google Scholar]
  • 70. Ochi R, Chettimada S, Kizub I, et al. Dehydroepiandrosterone inhibits I(Ca, L) and its window current in voltage-dependent and -independent mechanisms in arterial smooth muscle cells. Am J Physiol Heart Circ Physiol 2018; 315: H1602–H1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Chen J, Xu L, Huang C. DHEA inhibits vascular remodeling following arterial injury: a possible role in suppression of inflammation and oxidative stress derived from vascular smooth muscle cells. Mol Cell Biochem 2014; 388: 75–84. [DOI] [PubMed] [Google Scholar]
  • 72. Ii M, Hoshiga M, Negoro N, et al. Adrenal androgen dehydroepiandrosterone sulfate inhibits vascular remodeling following arterial injury. Atherosclerosis 2009; 206: 77–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Urata Y, Goto S, Kawakatsu M, et al. DHEA attenuates PDGF-induced phenotypic proliferation of vascular smooth muscle A7r5 cells through redox regulation. Biochem Biophys Res Commun 2010; 396: 489–494. [DOI] [PubMed] [Google Scholar]
  • 74. Dumas de La Roque E, Bellance N, Rossignol R, et al. Dehydroepiandrosterone reverses chronic hypoxia/reoxygenation-induced right ventricular dysfunction in rats. Eur Respir J 2012; 40: 1420–1429. [DOI] [PubMed] [Google Scholar]
  • 75. Alzoubi A, Toba M, Abe K, et al. Dehydroepiandrosterone restores right ventricular structure and function in rats with severe pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2013; 304: H1708–H1718. [DOI] [PubMed] [Google Scholar]
  • 76. Mannic T, Mouffok M, Python M, et al. DHEA prevents mineralo- and glucocorticoid receptor-induced chronotropic and hypertrophic actions in isolated rat cardiomyocytes. Endocrinology 2013; 154: 1271–1281. [DOI] [PubMed] [Google Scholar]
  • 77. Nakamura S, Yoshimura M, Nakayama M, et al. Possible association of heart failure status with synthetic balance between aldosterone and dehydroepiandrosterone in human heart. Circulation 2004; 110: 1787–1793. [DOI] [PubMed] [Google Scholar]
  • 78. Boese AC, Kim SC, Yin K-J, et al. Sex differences in vascular physiology and pathophysiology: estrogen and androgen signaling in health and disease. Am J Physiol Heart Circ Physiol 2017; 313: H524–H545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Somani YB, Pawelczyk JA, De Souza MJ, et al. Aging women and their endothelium: probing the relative role of estrogen on vasodilator function. Am J Physiol Heart Circ Physiol 2019; 317: H395–H404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Moreau KL, Hildreth KL, Klawitter J, et al. Decline in endothelial function across the menopause transition in healthy women is related to decreased estradiol and increased oxidative stress. Geroscience 2020; 42: 1699–1714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Keller AC, Klawitter J, Hildreth KL, et al. Elevated plasma homocysteine and cysteine are associated with endothelial dysfunction across menopausal stages in healthy women. J Appl Physiol 2019; 126: 1533–1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Stone T, Stachenfeld NS. Pathophysiological effects of androgens on the female vascular system. Biol Sex Differ 2020; 11: 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Samargandy S, Matthews KA, Brooks MM, et al. Arterial stiffness accelerates within 1 year of the final menstrual period: the SWAN Heart Study. Arterioscler Thromb Vasc Biol 2020; 40: 1001–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Georgiopoulos GA, Lambrinoudaki I, Athanasouli F, et al. Free androgen index as a predictor of blood pressure progression and accelerated vascular aging in menopause. Atherosclerosis 2016; 247: 177–183. [DOI] [PubMed] [Google Scholar]
  • 85. Subramanya V, Ambale-Venkatesh B, Ohyama Y, et al. Relation of sex hormone levels with prevalent and 10-year change in aortic distensibility assessed by MRI: the multi-ethnic study of atherosclerosis. Am J Hypertens 2018; 31: 774–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Lambrinoudaki I, Georgiopoulos GA, Athanasouli F, et al. Free androgen index as a determinant of arterial stiffness in menopause: a mediation analysis. Menopause 2017; 24: 635–644. [DOI] [PubMed] [Google Scholar]
  • 87. Kische H, Gross S, Wallaschofski H, et al. Serum androgen concentrations and subclinical measures of cardiovascular disease in men and women. Atherosclerosis 2016; 247: 193–200. [DOI] [PubMed] [Google Scholar]
  • 88. Lee JM, Colangelo LA, Schwartz JE, et al. Associations of cortisol/testosterone and cortisol/sex hormone-binding globulin ratios with atherosclerosis in middle-age women. Atherosclerosis 2016; 248: 203–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Calderon-Margalit R, Schwartz SM, Wellons MF, et al. Prospective association of serum androgens and sex hormone-binding globulin with subclinical cardiovascular disease in young adult women: the ‘coronary artery risk development in young adults’ women’s study. J Clin Endocrinol Metab 2010; 95: 4424–4431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Subramanya V, Zhao D, Ouyang P, et al. Association of endogenous sex hormone levels with coronary artery calcium progression among post-menopausal women in the Multi-Ethnic Study of Atherosclerosis (MESA). J Cardiovasc Comput Tomogr 2019; 13: 41–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Subramanya V, Zhao D, Ouyang P, et al. Sex hormone levels and change in left ventricular structure among men and post-menopausal women: the Multi-Ethnic Study of Atherosclerosis (MESA). Maturitas 2018; 108: 37–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Kische H, Gross S, Wallaschofski H, et al. Clinical correlates of sex hormones in women: the study of health in Pomerania. Metabolism 2016; 65: 1286–1296. [DOI] [PubMed] [Google Scholar]
  • 93. Di Z, Eliseo G, Pamela O, et al. Endogenous sex hormones and incident cardiovascular disease in post-menopausal women. J Am Coll Cardiol 2018; 71: 2555–2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Zhao D, Guallar E, Ballantyne CM, et al. Sex hormones and incident heart failure in men and postmenopausal women: the atherosclerosis risk in communities study. J Clin Endocrinol Metab 2020; 105: e3798–e3807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Wang J, Fan X, Yang M, et al. Sex-specific associations of circulating testosterone levels with all-cause and cause-specific mortality. Eur J Endocrinol 2021; 184: 723–732. [DOI] [PubMed] [Google Scholar]
  • 96. Jia X, Sun C, Tang O, et al. Plasma dehydroepiandrosterone sulfate and cardiovascular disease risk in older men and women. J Clin Endocrinol Metab 2020; 105: e4304–e4327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Schederecker F, Cecil A, Prehn C, et al. Sex hormone-binding globulin, androgens and mortality: the KORA-F4 cohort study. Endocr Connect 2020; 9: 326–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Schaffrath G, Kische H, Gross S, et al. Association of sex hormones with incident 10-year cardiovascular disease and mortality in women. Maturitas 2015; 82: 424–430. [DOI] [PubMed] [Google Scholar]
  • 99. Daka B, Langer RD, Larsson CA, et al. Low concentrations of serum testosterone predict acute myocardial infarction in men with type 2 diabetes mellitus. BMC Endocr Disord 2015; 15: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Holmegard HN, Nordestgaard BG, Jensen GB, et al. Sex hormones and ischemic stroke: a prospective cohort study and meta-analyses. J Clin Endocrinol Metab 2016; 101: 69–78. [DOI] [PubMed] [Google Scholar]
  • 101. Laughlin GA, Goodell V, Barrett-Connor E. Extremes of endogenous testosterone are associated with increased risk of incident coronary events in older women. J Clin Endocrinol Metab 2010; 95: 740–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Wehr E, Pilz S, Boehm BO, et al. Low free testosterone levels are associated with all-cause and cardiovascular mortality in postmenopausal diabetic women. Diabetes Care 2011; 34: 1771–1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Benn M, Voss SS, Holmegard HN, et al. Extreme concentrations of endogenous sex hormones, ischemic heart disease, and death in women. Arterioscler Thromb Vasc Biol 2015; 35: 471–477. [DOI] [PubMed] [Google Scholar]
  • 104. Wallace IR, McKinley MC, Bell PM, et al. Sex hormone binding globulin and insulin resistance. Clin Endocrinol 2013; 78: 321–329. [DOI] [PubMed] [Google Scholar]
  • 105. Laurent MR, Hammond GL, Blokland M, et al. Sex hormone-binding globulin regulation of androgen bioactivity in vivo: validation of the free hormone hypothesis. Sci Rep 2016; 6: 35539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Worboys S, Kotsopoulos D, Teede H, et al. Evidence that parenteral testosterone therapy may improve endothelium-dependent and -independent vasodilation in postmenopausal women already receiving estrogen. J Clin Endocrinol Metab 2001; 86: 158–161. [DOI] [PubMed] [Google Scholar]
  • 107. Hak AE, Westendorp ICD, Pols HAP, et al. High-dose testosterone is associated with atherosclerosis in postmenopausal women. Maturitas 2007; 56: 153–160. [DOI] [PubMed] [Google Scholar]
  • 108. Penotti M, Sironi L, Cannata L, et al. Effects of androgen supplementation of hormone replacement therapy on the vascular reactivity of cerebral arteries. Fertil Steril 2001; 76: 235–240. [DOI] [PubMed] [Google Scholar]
  • 109. Alzahrani T, Nguyen T, Ryan A, et al. Cardiovascular disease risk factors and myocardial infarction in the transgender population. Circ Cardiovasc Qual Outcomes 2019; 12: e005597. [DOI] [PubMed] [Google Scholar]
  • 110. Caceres BA, Jackman KB, Edmondson D, et al. Assessing gender identity differences in cardiovascular disease in US adults: an analysis of data from the 2014-2017 BRFSS. J Behav Med 2020; 43: 329–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Nota NM, Wiepjes CM, de Blok CJM, et al. Occurrence of acute cardiovascular events in transgender individuals receiving hormone therapy. Circulation 2019; 139: 1461–1462. [DOI] [PubMed] [Google Scholar]
  • 112. Poteat TC, Divsalar S, Streed CGJ, et al. Cardiovascular disease in a population-based sample of transgender and cisgender adults. Am J Prev Med 2021; 61: 804–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Getahun D, Nash R, Flanders WD, et al. Cross-sex hormones and acute cardiovascular events in transgender persons: a cohort study. Ann Intern Med 2018; 169: 205–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Elamin MB, Garcia MZ, Murad MH, et al. Effect of sex steroid use on cardiovascular risk in transsexual individuals: a systematic review and meta-analyses. Clin Endocrinol 2010; 72: 1–10. [DOI] [PubMed] [Google Scholar]
  • 115. Maraka S, Singh Ospina N, Rodriguez-Gutierrez R, et al. Sex steroids and cardiovascular outcomes in transgender individuals: a systematic review and meta-analysis. J Clin Endocrinol Metab 2017; 102: 3914–3923. [DOI] [PubMed] [Google Scholar]
  • 116. Velho I, Fighera TM, Ziegelmann PK, et al. Effects of testosterone therapy on BMI, blood pressure, and laboratory profile of transgender men: a systematic review. Andrology 2017; 5: 881–888. [DOI] [PubMed] [Google Scholar]
  • 117. Capellino S, Straub RH, Cutolo M. Aromatase and regulation of the estrogen-to-androgen ratio in synovial tissue inflammation: common pathway in both sexes. Ann N Y Acad Sci 2014; 1317: 24–31. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Endocrinology and Metabolism are provided here courtesy of SAGE Publications

RESOURCES