Next Article in Journal
Metabolomic Profiling of Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy for Predicting Disease-Free and Overall Survival
Previous Article in Journal
The Locus Coeruleus in Chronic Pain
Previous Article in Special Issue
The Phenotypic Variability Associated with Hepatocyte Nuclear Factor 1B Genetic Defects Poses Challenges in Both Diagnosis and Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sex-Specific Differences in Kidney Function and Blood Pressure Regulation

by
Eleni Stamellou
1,2,*,
Viktor Sterzer
1,
Jessica Alam
1,
Stefanos Roumeliotis
3,
Vassilios Liakopoulos
3 and
Evangelia Dounousi
2
1
Division of Nephrology and Clinical Immunology, RWTH Aachen University, 52074 Aachen, Germany
2
Department of Nephrology, University Hospital of Ioannina, 45500 Ioannina, Greece
3
2nd Department of Nephrology, AHEPA University Hospital Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(16), 8637; https://doi.org/10.3390/ijms25168637
Submission received: 4 July 2024 / Revised: 29 July 2024 / Accepted: 30 July 2024 / Published: 8 August 2024
(This article belongs to the Special Issue Molecular Pathology, Diagnostics, and Therapeutics of Kidney Disease)

Abstract

:
Premenopausal women generally exhibit lower blood pressure and a lower prevalence of hypertension than men of the same age, but these differences reverse postmenopause due to estrogen withdrawal. Sexual dimorphism has been described in different components of kidney physiology and pathophysiology, including the renin–angiotensin–aldosterone system, endothelin system, and tubular transporters. This review explores the sex-specific differences in kidney function and blood pressure regulation. Understanding these differences provides insights into potential therapeutic targets for managing hypertension and kidney diseases, considering the patient’s sex and hormonal status.

1. Introduction

Women live longer, experience a slower age-related decline in glomerular filtration rate (GFR), and are less prone to various kidney diseases [1]. One factor contributing to these differences may be the higher blood pressure observed in men. Starting at puberty, men develop higher blood pressure compared to women [2,3,4,5]. This trend reverses after menopause, when women’s blood pressure increases more rapidly and may eventually equal or surpass that of age-matched men [6]. Studies in both humans and animals indicate a higher prevalence and incidence of hypertension in males than females [5]. Consequently, higher blood pressure in men is associated with faster age-dependent decline in GFR, a more rapid progression of chronic kidney disease, and an increased risk of cardiovascular disease [7,8]. Interestingly, men with hypotestosteronemia or hyperestrogenemia exhibit a reduced incidence of coronary disease [9].
Animal models of hypertension, including spontaneously hypertensive rats (SHRs), deoxycorticosterone acetate (DOCA)-salt, and chronic angiotensin (Ang II) infusion, demonstrate that females are more resistant to blood pressure increases compared to males [10]. The correlation between these sex-specific blood pressure changes and hormonal alterations suggests that sex hormones might play a role in regulating blood pressure. Moreover, considering the kidney’s crucial role in blood pressure regulation, it is plausible that differences in blood pressure between the sexes are influenced not only by systemic sex hormones but also by kidney-specific hormonal actions. This review aims to explore the potential mechanisms underlying these phenomena. For this narrative review, PubMed was searched for publication using the term ‘gender differences’ or ‘androgens’, ‘sex hormones’, ‘sexual dimorphism’ and ‘kidney disease’ or ‘hypertension’, and hormonal influences. Of the articles that were found those published in languages other than English were deleted. Emphasis was placed on original works involving both humans (Table 1) and animals (Table 2).

2. Sex and Hypertension

Sexual dimorphism in hypertension has been observed in both human and laboratory animal studies, indicating that sex hormones significantly influence blood pressure regulation and related pathophysiological outcomes. For instance, studies on Wistar rats have shown that testosterone supplementation increases blood pressure in both male and female Wistar rats, an effect that can be blocked by the androgen receptor (AR) blocker flutamide, suggesting a direct role of AR in mediating hypertension [34]. Furthermore, in SHRs and in the renal wrap model, testosterone has been found to exacerbate hypertension, while flutamide effectively reduces blood pressure, highlighting the AR pathway’s involvement in salt-induced hypertension [21,22,23,35,36]. In murine ren2 gene-expressing rats, the administration of flutamide not only lowered blood pressure but also reduced organ damage, emphasizing the potential therapeutic benefits of AR blockade [24]. Prenatal exposure to testosterone cypionate in female rats resulted in elevated blood pressure and increased AR expression in the heart, suggesting that the early hormonal environment can have lasting effects on cardiovascular health [25]. Additionally, castration studies in various rodent models demonstrated a reduction in blood pressure, which could be reversed by testosterone supplementation, further underlining the role of testosterone in hypertension [21,27,37,38,39,40]. Interestingly, the impact of ovariectomy on blood pressure presents a contrast; while it does not affect blood pressure in female rodents, it leads to salt-independent hypertension in Dahl salt-sensitive (DS) rats [41,42]. This indicates a protective role of estrogens against hypertension, shedding light on the complex interplay between sex hormones and blood pressure regulation.
Sex hormones also influence the susceptibility to kidney damage. In a study, castrated rats on an 8% NaCl diet exhibited lower blood pressure, decreased albuminuria, plasma renin activity, and angiotensin concentrations, along with increased sodium excretion and creatinine clearance compared to controls. Testosterone supplementation reversed these effects [43]. Castration of young rats prevented the development of renal injury and hypertension. In 18-month-old male SHRs castrated at 8 months, blood pressure significantly reduced, glomerular injury was nearly absent, and GFR improved compared to their intact counterparts [30]. Male mice are more susceptible to ischemia/reperfusion (I/R) injury, with castration providing a protective effect [31]. Conversely, female sex hormones are believed to play a protective role in preventing renal injury [44,45,46,47]. Testosterone exacerbates glomerular injury in female Sprague Dawley rats, although the injury was still much milder than in the untreated male group [48].
While testosterone has some detrimental effects, it also has beneficial functions, particularly in males. Testosterone supplements in obese men improve insulin resistance, reduce body fat, and improve lipid profiles, without increasing blood pressure [49]. Low androgen levels in men are associated with various chronic diseases such as CKD, cancer, atherosclerosis, cardiovascular disease, sarcopenia, and osteoporosis [50,51,52]. However, it remains unclear whether the reduction in androgens is the cause or the result of these diseases [53]. Testosterone supplementation in male obese Zucker rats reduces metabolic syndrome symptoms but increases blood pressure [54]. In women, increased androgen levels lead to obesity and cardiovascular disease, and testosterone supplementation in a rat model of polycystic kidney disease increases cholesterol, leptin, and insulin levels while reducing glucose tolerance [13,55].

3. Kidney and Blood Pressure

Kidney dysfunction plays a critical role in the development and maintenance of hypertension, as shown by transplant studies in both humans and animals. Transplanting kidneys from prehypertensive individuals into normotensive recipients often leads to hypertension in the latter, indicating that blood pressure “follows” the kidney [56,57]. Similarly, kidneys from donors with a family history of hypertension significantly increase the recipient’s risk of hypertension compared to kidneys from donors without such a history [58]. Conversely, transplanting a kidney from a normotensive donor can normalize blood pressure in hypertensive patients [15]. Animal studies support this relationship. For instance, transplanting a kidney from SHRs to normotensive Wistar rats induces hypertension in the recipient. Transplanting a kidney from a normotensive to a hypertensive rat decreases the recipient’s blood pressure [59].
Sex hormones profoundly influence kidney function, affecting the organ’s structure, metabolic processes, and its ability to secrete and reabsorb substances. These hormones modulate the pharmacokinetic and toxicological responses to various compounds, largely due to hormonal regulation of specific kidney enzymes, ion channels, and membrane transport proteins [60,61,62,63]. Androgens are produced within the kidney, and ARs are found in different kidney tubule segments. Alterations in tubule transporter activity, driven by androgens, can result in sodium retention, which may lead to an increase in blood pressure [43,64,65,66,67,68]. This effect aligns with evidence showing that salt-induced hypertension is gender-dependent and the pressure–natriuresis response—the process by which the kidneys excrete sodium in response to blood pressure changes—is distinct between sexes. Specifically, females can excrete equivalent amounts of sodium at lower blood pressure compared to males, which is a difference observed in both humans and animal models [21,22,41,69,70,71]. Postmenopausal women become more susceptible to salt-sensitive hypertension and exhibit a more rapid progression of kidney diseases than premenopausal women, underscoring the critical role of the androgen–estrogen balance in blood pressure regulation [11,72,73]. Furthermore, androgens mediate hypertension through the sympathetic nervous system. Denervation prevents androgen-mediated blood pressure increase in hyperandrogenemic female rats (sympathetic nervous system) [74].
A transplantation study of Ang II-infused mice found that mean arterial pressure (MAP) increased significantly in female mice receiving a male kidney compared to female mice with a female kidney transplant. Male recipients of a female kidney had reduced MAP compared to the control group. Extrarenal regulation mechanisms also seem to influence the kidney, as angiotensin 1 receptor (AT1R) mRNA levels in the female-to-male transplantation group were increased by 50% compared to female-to-female mice, whereas AT1R levels in the male-to-female group were decreased compared to male-to-male mice. Additionally, endothelin-1 (ET-1) mRNA expression in female-to-male kidneys increased five-fold compared to the female-to-female group [75].

4. Endothelins

Endothelins (ETs), a group of endogenous peptides, mediate their actions through G protein-coupled receptors—endothelin A (ETA), endothelin B (ETB), and endothelin C (ETC). These peptides include endothelin 1 (ET-1), which is the more potent, endothelin 2 (ET-2), and endothelin 3 (ET-3). These receptors are distributed across various cell types within the cardiovascular system and kidneys, with ETA predominantly present on smooth muscle cells and ETB receptors located on endothelial and renal epithelial cells [76]. ET-2 and ET-2 are less studied and there are no data regarding a differential role in blood pressure regulation between sexes.
ET-1 exerts a wide range of biological effects in the kidney, including vasoconstriction of most renal vessels, mesangial cell contraction, enhancement of glomerular cell proliferation, induction of oxidative stress, expression of pro-inflammatory factors, and reduction of medullary blood flow through activation of ETA receptors. Conversely, activating renal ETB receptors induces vasodilation and inhibits Na+ and water reabsorption, resulting in subsequent natriuresis and diuresis [32,77,78]. The natriuretic effects of endothelin are primarily mediated via the ETB receptor by inhibiting Na+ reabsorption in the thick ascending limb and the collecting duct, or through vasodilation of vessels in the medulla during high salt intake, which enhances natriuresis and diuresis [32,79]. ETB receptors also influence vascular reactive oxygen species production and kidney nitric oxide synthase (NOS) activity, and they are involved in endothelin clearance, leading to its internalization and degradation. Collectively, ET-1 appears to have paradoxical effects on blood pressure regulation by promoting hypertension through vasoconstriction and relative hypotension through increased water and salt excretion [32,79,80]. Thus, the ET–ETB receptor pathway is generally seen as vasodilative and antihypertensive, in contrast to the prohypertensive ET–ETA pathway [81,82,83].
Sexual dimorphism in the ET-1 pathway is evident at various biological levels, including differences in plasma or tissue concentrations of ET-1, its production rates, receptor expression, physiological effects, and responses to receptor antagonists [58]. Evidence shows that plasma ET-1 levels are notably higher in men than in age-matched women. Moreover, in human aortic endothelial cells, ET-1 mRNA increases after stimulation with testosterone [84]. In contrast, estrogen appears to reduce ET-1 levels and prevent endothelin-mediated blood pressure elevation [85,86,87,88,89]. These differences seem to be hormonally regulated, as male-to-female transsexuals show lower ET levels after estrogen supplementation, while testosterone treatment in female-to-male transsexuals leads to an increase [12,78,90]. However, this hypothesis is challenged by findings that plasma ET-1 levels in males with hypogonadism are elevated, with a tendency to decrease after testosterone administration [91]. Furthermore, the contribution of ETB receptors to resting vascular tone differs between genders, with ETB receptors mediating tonic vasoconstriction in men, whereas in women, they mediate tonic vasodilation [92,93].
Deleting ET-1 or ETB receptor expression in collecting duct cells leads to salt retention and hypertension in mice [81]. The higher blood pressure elevation in male rats in the DOCA-salt model is presumably ETB receptor-mediated, as an ETB receptor knockout abolishes the blood pressure differences between the sexes [94]. Angiotensin II, whether stimulated exogenously or by a low-salt diet, inhibits ETB receptor function in male rats but to a much lesser degree in females [95]. Furthermore, acute infusion of an ETB agonist directly into the renal medulla enhances sodium and water excretion in both male and female rats. However, infusion of ET-1 only produces natriuresis in female rats, suggesting an altered functional distribution of ET receptors within the kidney [77]. This hypothesis is also supported by the different distribution of ET-1 receptors in the inner medullary collecting ducts of male and female rats, with females exhibiting fewer ETA receptors and an additional ETA-dependent natriuresis pathway potentially involving other cell types, such as the vasa recta, the thick ascending limb, or renomedullary interstitial cells. This excess of ETA receptors in males may inhibit ETB-mediated natriuresis [26]. This is supported by experiments in mice in which knocking out ET-1 and the ETB receptor led to salt retention and hypertension, whereas the knockout of the ETA receptor had no effect [96,97,98].
Female rats have alternative natriuretic mechanisms. Studies show effective salt excretion in ETB-deficient females, suggesting a prominent ET-independent salt excretion mechanism in females [99]. ETB-deficient female rats demonstrate higher medullary blood flow and enhanced diuresis and natriuresis following ET-1 infusion into the medulla, responses that are absent in male rats and diminished after ovariectomy [100,101]. The natriuretic response in male and ovariectomized female rats is diminished after ETA and ETB receptor blockade, highlighting a potential role for estrogens in this context [102]. Male ETB receptor-deficient rats on a normal salt diet also exhibit higher blood pressure and lower medullary NO activity than females. A high-salt diet further reduces NO plasma concentration and increases blood pressure, effects that are prevented by orchidectomy and reversed by testosterone supplementation [103]. Androgens in combination with a high-salt diet increase renal vascular resistance in male rats, whereas estradiol prevents salt-sensitive hypertension by enhancing NO production [104,105]. Conversely, female ETB receptor-deficient rats develop more severe hypertension on a high-salt diet than males, and gonadectomy increases the responsiveness of the afferent arteriole to ETB-induced vasoconstriction in females, but not males, indicating that female sex hormones may inhibit ETB-mediated vasoconstriction in the renal microcirculation [104,106].
ET-1 also contributes to kidney damage by increasing glomerular permeability for albumin and inducing glomerular oxidative stress, further exacerbated by high salt intake; these mechanisms are primarily ETA-mediated [107,108,109]. Nitric oxide (NO) production and pressure–natriuresis are decreased in mice with an ET-1 knockout in the collecting duct, suggesting that these processes are mediated by androgens, as ET-1 transgenic mice exhibit less renal injury post-castration [110,111]. Male DOCA-salt rats exhibit more severe kidney damage than females, but blocking the ETA receptor attenuates this damage in both sexes [90]. Studies shows higher renal ET-1 expression correlates with more severe kidney damage in male hypertensive rats. Estrogens seem to protect the kidney by lowering endothelin levels, as indicated by increased renal ET-1 expression and exacerbated renal injury after ovariectomy [112]. Estradiol suppresses kidney ET-1 production, which helps preventing acute kidney injury and other damage [113,114]. Male rats are more susceptible to renal ischemia/reperfusion injury, as male uninephrectomized rats had an 8% survival rate after 50 min of ischemia/reperfusion compared to 75% in females [31]. Orchidectomy reduces kidney damage and improves survival, with higher ET-1 mRNA expression in males than females supporting this hypothesis [31,111].
Overall, these findings illustrate complex interactions between sex hormones, endothelin receptors, and their effects on renal physiology and blood pressure regulation, highlighting distinct differences in the pathophysiological responses between males and females. ET-1 expression, as well as renal damage markers, are higher in male than female hypertensive rats [90,115].

5. Tubule Transporters

Ion transporters across different segments of the kidney tubule exhibit sexual dimorphism, being partially modulated by androgens [66,67,116,117]. Sexual dimorphism in Na+ handling and androgen-dependent salt sensitivity have been reported [21,40,41,68,118,119]. Research by Veiras et al. demonstrated a sex-specific expression pattern of tubular ion transporters that results in more efficient sodium excretion in females [120]. Additionally, studies have noted an increase in Na+ excretion in the absence of testosterone [43]. The physiological consequences of increased Na+ reabsorption in the renal tubule are significant, not only in terms of blood pressure regulation but also in metabolic terms. Enhanced Na+ reabsorption increases oxygen consumption and promotes the production of free radicals within the kidney [28,121,122,123]. This increased oxidative stress can contribute to renal damage and exacerbate conditions such as hypertension, highlighting the complex interactions between ion transport, hormonal regulation, and kidney health.

5.1. Epithelial Sodium Channel (ENaC)

The epithelial sodium channel (ENaC) is one of the key salt transporters in the distal renal tubule. ENaC plays a pivotal role in sodium and fluid reabsorption in the renal tubule, influencing blood pressure regulation. Testosterone has been shown to increase the expression of the alpha subunit of ENaC (αENaC) through the AR in both rat models and the human renal cell line HKC-8 [29]. Treatment with testosterone propionate in orchidectomized Sprague Dawley rats upregulates the expression of all three units of ENaC in the kidney, an effect that is reversed by the administration of both flutamide and finasteride, suggesting the involvement of a 5-hydroxytestosterone-dependent pathway [124]. Other studies have reported an increase in renal ENaC expression in ovariectomized female Wistar rats and SHRs following dihydroxytestosterone administration [125]. In contrast, orchiectomy increases all three ENaC subunits’ expression in kidneys of male Wistar rats and SHRs [126].
Aldosterone, whose synthesis is also influenced by testosterone, further enhances ENaC expression in the kidney [127,128,129]. This relationship underscores a potential mechanism whereby testosterone increases blood pressure by promoting sodium retention through the upregulation of ENaC via the activation of the renin angiotensin system (RAS) pathway. Moreover, there are indications that ENaC regulation is also influenced by estradiol [130,131,132,133], suggesting a complex interplay between sex hormones and renal sodium handling that impacts blood pressure regulation.

5.2. Sodium–Hydrogen Exchanger 3 (NHE3)

NHE3, located in the proximal tubule brush border membrane, plays a crucial role in sodium reabsorption and acid–base balance. Expression and activity of NHE3 was found to increase after an androgen-mediated upregulation of renal Ang II expression. This hormonal interaction leads to enhanced Na+ and fluid reabsorption, subsequently raising blood pressure [28,134]. The activity of the Na/H exchanger in the brush border membrane vesicles (BBMVs) is also stimulated by androgens, showing higher expression levels in males compared to females. Castration has been shown to decrease this activity, whereas testosterone supplementation reverses this effect, demonstrating the androgen dependency of this transporter system [135]. In male mice, castration leads to a decrease in Na/H+ exchanger expression, which is regulated by Ang II, further underscoring the interplay between androgens and renal hormonal pathways in regulating these transporters [135].

5.3. Aquaporins

Supplementation with testosterone of ovariectomized Wistar rats increased the expression of several aquaporins (AQ): AQ1, AQ2, AQ4, and AQ7, whereas AQ3 was downregulated. The effects of testosterone were context-dependent; under normotensive conditions, AQ3 expression decreased, but under hypertensive conditions, it increased. Expression of AQ6 has also been shown to be increased by testosterone treatment under hypertensive conditions [136,137]. Further research supports these findings, showing that testosterone also increased blood pressure and similarly affected aquaporin expression in male Sprague Dawley rats, increasing levels of AQ1, AQ2, AQ4, AQ6, and AQ7, while simultaneously decreasing AQ3 [138]. On the other hand, estradiol, a key estrogen hormone, appears to inhibit AQ2 expression in the collecting duct, underscoring a potential contrasting role of estrogens in this regulatory pathway [139]. This interaction highlights the complex hormonal regulation of aquaporins, which plays a crucial role in the body’s fluid balance and blood pressure management.

6. Renin–Angiotensin–Aldosterone System

Sex hormones play a significant role in regulating the renin–angiotensin system (RAS). Androgens increase the expression of several components of the RAS [53]. They stimulate the synthesis of angiotensinogen, with studies showing that castrated male rats have a 60% lower Ang II mRNA levels compared to controls [140,141]. Additionally, testosterone upregulates angiotensinogen levels in the kidneys of male SHRs and Sprague Dawley and DS rats [140,142,143]. Furthermore, there is a linear correlation between testosterone activity and plasma renin activity, with males showing higher plasma renin activity compared to females [144,145]. Testosterone treatment of ovariectomized rats results in increased plasma renin activity, while castration decreases it [140]. Acute testosterone treatment also increases plasma aldosterone levels [146].
Studies in SHRs have shown that testosterone promotes hypertension development via an androgen receptor-mediated mechanism that stimulates the systemic RAS. The final effector in the RAS, angiotensin II, promotes Na+ and water retention in the proximal tubule, contributing to higher blood pressure in males [147]. Ang II causes a greater increase in blood pressure in normotensive males than females (rats and mice) [148,149,150]. However, there is also a report that Ang II infusion had a similar effect on blood pressure in men and women on a controlled Na+ and protein diet [151,152]. The contribution of the RAS to gender-specific blood pressure differences could not be confirmed in aged heterozygous Ren-2 transgenic rats [42]. A possible explanation is the loss of the protective effect of estrogens in aged females as estrogen levels decline after menopause.
Besides the systemic RAS, the intrarenal RAS is a key component of blood pressure regulation, and sex steroids can modulate the expression of intrarenal RAS components [151,152,153,154]. The intrarenal RAS is more activated in males than in females and is downregulated after castration in DS rats and SHRs [128,143,155]. Androgens increase kidney angiotensinogen (AGT) levels significantly but have only a slight effect on liver AGT levels [140]. Angiotensinogen mRNA decreases in castrated male rats in the kidney and to a much lesser extent in the liver. Testosterone-treated castrates have the same angiotensinogen levels as the controls [140].
In contrast to androgens, estrogens induce the expression of non-classic RAS components that have a blood-pressure-lowering effect (Figure 1) [156,157]. This aligns with the observation that female normotensive rats have a higher Ace2 and Masr gene expression than males [158,159]. Treatment with estrogen also decreases aldosterone production by upregulating the AT2R and downregulating the AT1R expression [160], thereby shifting the RAS towards blood-pressure-lowering pathways. Gupte et al. showed that estrogen protects female rats from hypertension by reducing the formation of Ang II in favor of Ang (1–7) in a high-fat diet (HFD) model [161]. Studies by another group revealed that estradiol downregulates the AT1R and ACE in the kidney, adrenal vasculature, and brain in female normotensive rats, thereby shifting the RAS towards non-canonical pathways [162]. It is also known that the AT2R is upregulated in female kidney of mice and rats, especially at high Ang II levels, and that females have a lower renal AT1R/AT2R ratio than males [33,163]. Ovariectomy, on the other hand, reduces the AT2R and upregulates the AT1R expression in SHRs [42]. This effect is reversed by estrogen administration. Castration of Sprague Dawley rats increases AT2R mRNA expression, which could be reversed by testosterone administration [164]. AT2R also reduces the sensitivity of the tubulo-glomerular feedback mechanism (regulator of GFR) only in females [165].
It also seems that there are some sex-specific differences regarding the response to treatment with the specific components of the RAS pathway. For example, treatment with angiotensin receptor blockers in women with congestive heart failure provides better survival rates than treatment with ACE inhibitors [16,166]. Interestingly, angiotensin receptor blockers reduce proteinuria in females more efficiently than in men [155]. Women also have a more rapid response to the AT1R antagonist irbesartan [151]. ACE inhibitors also reduce proteinuria in females more effectively than in males; however, blood pressure reduction by these agents seems to be similar in both genders [17,167,168].
Metabolites of arachidonic acid play a crucial role in kidney function and blood pressure regulation [169,170]. It has been shown that androgens can alter the metabolism of arachidonic acid in the renal cortex [169]. Androgen mediates production of 20-hydroxyeicosatetraenoic acid (20-HETE), an eicosanoid metabolite of arachidonic acid, which has varying effects depending on its site of expression. When 20-HETE is expressed in the renal vasculature, it promotes vasoconstriction and hypertension. Conversely, if expressed in the kidney tubule, it prevents sodium reabsorption, leading to higher natriuresis and lower blood pressure [171]. Testosterone increases the expression of the 20-HETE synthase Cyp4a12a in kidney of mice on a 129S6 background.

7. Sex-Specific Genetic Susceptibilities in Hypertension: Insights from GWAS

Even though several studies have reported both common and rare variants with a significant genome-wide association to hypertension [172,173,174,175,176], only a recent one identified genetic loci that have differential susceptibility to hypertension and blood pressure based on sex [18]. They demonstrate sex-specific genetic susceptibilities to hypertension. One notable finding is the rs11066015 variant of the ACAD10 gene, which showed exome-wide significance in males but not in sex-combined analyses. This variant, common in Asian populations, influences hypertension potentially through mitochondrial oxidative stress pathways. Furthermore, the HECTD4 gene variants, rs11066280 and rs2074356, displayed male-specific genetic significance, possibly linked to their role in the ubiquitin system and androgen receptor promotion. Similarly, the rs671 variant of the ALDH2 gene was associated with hypertension predominantly in men, likely due to its function in reducing oxidative stress. These sex-specific genetic effects suggest that hormonal differences may modulate these genetic pathways, contributing to the differential risk of hypertension between males and females.

8. Sex-Specific Nutraceutical Approaches

Evidence suggests that women and men may respond differently to dietary interventions due to sex hormones [177,178]. However, few controlled studies have assessed sex differences in cardiovascular response to specific diets [179]. For example, adherence to a Mediterranean diet (MedDiet) has been shown to significantly lower cardiovascular disease (CVD) risk more in women than in men [179]. Regarding hypertension, epidemiological data indicate that whole grains and legumes reduce hypertension risk in women, but not in men, even after controlling for confounders [19]. In the Korean National Health and Nutrition Examination Survey, women who frequently consumed fried foods (≥2 times/week) had a 2.4-fold higher risk of hypertension compared to those who rarely consumed fried foods, a significant association not seen in men [20]. Additionally, reductions in blood pressure with diets low in sodium, and those supplemented with potassium or dark chocolate, were greater in women than in men [14]. A healthy diet was more effective in alleviating hypertension risk factors in women than in men [180].
Overall, the existing literature underscores the importance of investigating sex-related differences in dietary responses. Such research could lead to individualized dietary recommendations, improving health outcomes for both men and women.

9. Conclusions

This review highlights the significant impact of androgens on kidney function and blood pressure regulation, emphasizing the sex-specific differences mediated by these hormones (Figure 2). Understanding these mechanisms provides insights into potential therapeutic targets for managing hypertension and kidney diseases, considering the patient’s sex and hormonal status. Future research should focus on elucidating the precise molecular interactions and long-term impacts of sex hormones on renal and cardiovascular health, paving the way for more personalized treatment strategies.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Berg, U.B. Differences in decline in GFR with age between males and females. Reference data on clearances of inulin and PAH in potential kidney donors. Nephrol. Dial. Transpl. 2006, 21, 2577–2582. [Google Scholar] [CrossRef] [PubMed]
  2. Bachmann, J.; Feldmer, M.; Ganten, U.; Stock, G.; Ganten, D. Sexual dimorphism of blood pressure: Possible role of the renin-angiotensin system. J. Steroid Biochem. Mol. Biol. 1991, 40, 511–515. [Google Scholar] [CrossRef] [PubMed]
  3. Harshfield, G.A.; Alpert, B.S.; Pulliam, D.A.; Somes, G.W.; Wilson, D.K. Ambulatory blood pressure recordings in children and adolescents. Pediatrics 1994, 94, 180–184. [Google Scholar] [CrossRef] [PubMed]
  4. Burt, V.L.; Whelton, P.; Roccella, E.J.; Brown, C.; Cutler, J.A.; Higgins, M.; Horan, M.J.; Labarthe, D. Prevalence of hypertension in the US adult population. Results from the Third National Health and Nutrition Examination Survey, 1988–1991. Hypertension 1995, 25, 305–313. [Google Scholar] [CrossRef] [PubMed]
  5. Stamler, R.; Stamler, J.; Riedlinger, W.F.; Algera, G.; Roberts, R.H. Weight and blood pressure. Findings in hypertension screening of 1 million Americans. JAMA 1978, 240, 1607–1610. [Google Scholar] [CrossRef] [PubMed]
  6. Narkiewicz, K.; Phillips, B.G.; Kato, M.; Hering, D.; Bieniaszewski, L.; Somers, V.K. Gender-selective interaction between aging, blood pressure, and sympathetic nerve activity. Hypertension 2005, 45, 522–525. [Google Scholar] [CrossRef] [PubMed]
  7. Balafa, O.; Fernandez-Fernandez, B.; Ortiz, A.; Dounousi, E.; Ekart, R.; Ferro, C.J.; Mark, P.B.; Valdivielso, J.M.; Del Vecchio, L.; Mallamaci, F. Sex disparities in mortality and cardiovascular outcomes in chronic kidney disease. Clin. Kidney J. 2024, 17, sfae044. [Google Scholar] [CrossRef]
  8. Chesnaye, N.C.; Carrero, J.J.; Hecking, M.; Jager, K.J. Differences in the epidemiology, management and outcomes of kidney disease in men and women. Nat. Rev. Nephrol. 2024, 20, 7–20. [Google Scholar] [CrossRef] [PubMed]
  9. Phillips, G.B.; Pinkernell, B.H.; Jing, T.Y. The association of hypotestosteronemia with coronary artery disease in men. Arterioscler. Thromb. 1994, 14, 701–706. [Google Scholar] [CrossRef]
  10. Yanes, L.L.; Sartori-Valinotti, J.C.; Reckelhoff, J.F. Sex steroids and renal disease: Lessons from animal studies. Hypertension 2008, 51, 976–981. [Google Scholar] [CrossRef]
  11. Messerli, F.H.; Garavaglia, G.E.; Schmieder, R.E.; Sundgaard-Riise, K.; Nunez, B.D.; Amodeo, C. Disparate cardiovascular findings in men and women with essential hypertension. Ann. Intern. Med. 1987, 107, 158–161. [Google Scholar] [CrossRef] [PubMed]
  12. van Kesteren, P.J.M.; Kooistra, T.; Lansink, M.; van Kamp, G.J.; Asscheman, H.; Gooren, L.J.G.; Emeis, J.J.; Vischer, U.M.; Stehouwer, C.D.A. The effects of sex steroids on plasma levels of marker proteins of endothelial cell functioning. Thromb. Haemost. 1998, 79, 1029–1033. [Google Scholar] [CrossRef]
  13. Christakou, C.D.; Diamanti-Kandarakis, E. Role of androgen excess on metabolic aberrations and cardiovascular risk in women with polycystic ovary syndrome. Womens Health 2008, 4, 583–594. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, W.; Srinivasan, S.R.; Li, S.; Boerwinkle, E.; Berenson, G.S. Gender-specific influence of NO synthase gene on blood pressure since childhood: The Bogalusa Heart Study. Hypertension 2004, 44, 668–673. [Google Scholar] [CrossRef] [PubMed]
  15. Curtis, J.J.; Luke, R.G.; Dustan, H.P.; Kashgarian, M.; Whelchel, J.D.; Jones, P.; Diethelm, A.G. Remission of essential hypertension after renal transplantation. N. Engl. J. Med. 1983, 309, 1009–1015. [Google Scholar] [CrossRef] [PubMed]
  16. Zapater, P.; Novalbos, J.; Gallego-Sandín, S.; Hernández, F.T.; Abad-Santos, F. Gender differences in angiotensin-converting enzyme (ACE) activity and inhibition by enalaprilat in healthy volunteers. J. Cardiovasc. Pharmacol. 2004, 43, 737–744. [Google Scholar] [CrossRef]
  17. Ruggenenti, P.; Perna, A.; Zoccali, C.; Gherardi, G.; Benini, R.; Testa, A.; Remuzzi, G. Chronic proteinuric nephropathies. II. Outcomes and response to treatment in a prospective cohort of 352 patients: Differences between women and men in relation to the ACE gene polymorphism. Gruppo Italiano di Studi Epidemologici in Nefrologia (Gisen). J. Am. Soc. Nephrol. 2000, 11, 88–96. [Google Scholar] [CrossRef] [PubMed]
  18. Cho, S.B.; Jang, J.A. Genome-Wide Association Study of a Korean Population Identifies Genetic Susceptibility to Hypertension Based on Sex-Specific Differences. Genes 2021, 12, 1804. [Google Scholar] [CrossRef] [PubMed]
  19. Song, S.; Kim, J.; Kim, J. Gender Differences in the Association between Dietary Pattern and the Incidence of Hypertension in Middle-Aged and Older Adults. Nutrients 2018, 10, 252. [Google Scholar] [CrossRef]
  20. Kang, Y.; Kim, J. Association between fried food consumption and hypertension in Korean adults. Br. J. Nutr. 2016, 115, 87–94. [Google Scholar] [CrossRef]
  21. Reckelhoff, J.F.; Zhang, H.; Granger, J.P. Testosterone exacerbates hypertension and reduces pressure-natriuresis in male spontaneously hypertensive rats. Hypertension 1998, 31, 435–439. [Google Scholar] [CrossRef] [PubMed]
  22. Reckelhoff, J.F. Gender differences in the regulation of blood pressure. Hypertension 2001, 37, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  23. Caplea, A.; Seachrist, D.; Dunphy, G.; Ely, D. Sodium-induced rise in blood pressure is suppressed by androgen receptor blockade. Am. J. Physiol. Heart Circ. Physiol. 2001, 280, H1793–H1801. [Google Scholar] [CrossRef] [PubMed]
  24. Baltatu, O.; Cayla, C.A.A.; Iliescu, R.; Andreev, D.; Jordan, C.; Bader, M. Abolition of hypertension-induced end-organ damage by androgen receptor blockade in transgenic rats harboring the mouse ren-2 gene. J. Am. Soc. Nephrol. 2002, 13, 2681–2687. [Google Scholar] [CrossRef] [PubMed]
  25. Sherman, S.B.; Sarsour, N.; Salehi, M.; Schroering, A.; Mell, B.; Joe, B.; Hill, J.W. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes 2018, 9, 400–421. [Google Scholar] [CrossRef] [PubMed]
  26. Jin, C.; Speed, J.S.; Hyndman, K.A.; O’Connor, P.M.; Pollock, D.M.; Douma, L.G.; Solocinski, K.; Holzworth, M.R.; Crislip, G.R.; Masten, S.H.; et al. Sex differences in ET-1 receptor expression and Ca2+ signaling in the IMCD. Am. J. Physiol. Renal. Physiol. 2013, 305, F1099-104. [Google Scholar] [CrossRef]
  27. Iams, S.G.; McMurthy, J.P.; Wexler, B.C. Aldosterone, deoxycorticosterone, corticosterone, and prolactin changes during the lifespan of chronically and spontaneously hypertensive rats. Endocrinology 1979, 104, 1357–1363. [Google Scholar] [CrossRef] [PubMed]
  28. Kienitz, T.; Allolio, B.; Strasburger, C.; Quinkler, M. Sex-specific regulation of ENaC and androgen receptor in female rat kidney. Horm. Metab. Res. 2009, 41, 356–362. [Google Scholar] [CrossRef]
  29. Loh, S.Y.; Giribabu, N.; Salleh, N. Changes in plasma aldosterone and electrolytes levels, kidney epithelial sodium channel (ENaC) and blood pressure in normotensive WKY and hypertensive SHR rats following gonadectomy and chronic testosterone treatment. Steroids 2017, 128, 128–135. [Google Scholar] [CrossRef]
  30. Fortepiani, L.A.; Yanes, L.; Zhang, H.; Racusen, L.C.; Reckelhoff, J.F. Role of androgens in mediating renal injury in aging SHR. Hypertension 2003, 42, 952–955. [Google Scholar] [CrossRef]
  31. Müller, V.; Losonczy, G.; Heemann, U.; Vannay, Á.; Fekete, A.; Reusz, G.; Tulassay, T.; Szabó, A.J. Sexual dimorphism in renal ischemia-reperfusion injury in rats: Possible role of endothelin. Kidney Int. 2002, 62, 1364–1371. [Google Scholar] [CrossRef] [PubMed]
  32. Vassileva, I.; Mountain, C.; Pollock, D.M. Functional role of ETB receptors in the renal medulla. Hypertension 2003, 41, 1359–1363. [Google Scholar] [CrossRef] [PubMed]
  33. Silva-Antonialli, M.M.; Tostes, R.C.; Fernandes, L.; Fior-Chadi, D.R.; Akamine, E.H.; Carvalho, M.H.C.; Fortes, Z.B.; Nigro, D. A lower ratio of AT1/AT2 receptors of angiotensin II is found in female than in male spontaneously hypertensive rats. Cardiovasc. Res. 2004, 62, 587–593. [Google Scholar] [CrossRef] [PubMed]
  34. Hofmann, P.J.; Michaelis, M.; Gotz, F.; Bartel, C.; Kienitz, T.; Quinkler, M. Flutamide increases aldosterone levels in gonadectomized male but not female Wistar rats. Am. J. Hypertens. 2012, 25, 697–703. [Google Scholar] [CrossRef] [PubMed]
  35. Ji, H.; Menini, S.; Mok, K.; Zheng, W.; Pesce, C.; Kim, J.; Mulroney, S.; Sandberg, K.; Zimmerman, M.A.; Hutson, D.D.; et al. Gonadal steroid regulation of renal injury in renal wrap hypertension. Am. J. Physiol. Ren. Physiol. 2005, 288, F513-20. [Google Scholar] [CrossRef] [PubMed]
  36. Khalid, M.; Ilhami, N.; Giudicelli, Y.; Dausse, J.-P. Testosterone dependence of salt-induced hypertension in Sabra rats and role of renal alpha(2)-adrenoceptor subtypes. J. Pharmacol. Exp. Ther. 2002, 300, 43–49. [Google Scholar] [CrossRef] [PubMed]
  37. Masubuchi, Y.; Kumai, T.; Uematsu, A.; Komoriyama, K.; Hirai, M. Gonadectomy-induced reduction of blood pressure in adult spontaneously hypertensive rats. Acta Endocrinol. 1982, 101, 154–160. [Google Scholar] [CrossRef]
  38. Iams, S.G.; Wexler, B.C. Retardation in the development of spontaneous hypertension in SH rats by gonadectomy. J. Lab. Clin. Med. 1977, 90, 997–1003. [Google Scholar] [PubMed]
  39. Crofton, J.T.; Ota, M.; Share, L. Role of vasopressin, the renin-angiotensin system and sex in Dahl salt-sensitive hypertension. J. Hypertens 1993, 11, 1031–1038. [Google Scholar] [CrossRef]
  40. Chen, Y.F.; Meng, Q.C. Sexual dimorphism of blood pressure in spontaneously hypertensive rats is androgen dependent. Life Sci. 1991, 48, 85–96. [Google Scholar] [CrossRef]
  41. Hinojosa-Laborde, C.; Lange, D.L.; Haywood, J.R. Role of female sex hormones in the development and reversal of dahl hypertension. Hypertension 2000, 35, 484–489. [Google Scholar] [CrossRef]
  42. Vaněčková, I.; Husková, Z.; Vaňourková, Z.; Červenka, L. Castration has antihypertensive and organoprotective effects in male but not in female heterozygous Ren-2 rats. Kidney Blood Press Res. 2011, 34, 46–52. [Google Scholar] [CrossRef]
  43. Hu, J.; Tan, S.; Zhong, Y. Effects of testosterone on renal function in salt-loaded rats. Am. J. Med. Sci. 2011, 342, 38–43. [Google Scholar] [CrossRef]
  44. Dubey, R.K.; Jackson, E.K. Estrogen-induced cardiorenal protection: Potential cellular, biochemical, and molecular mechanisms. Am. J. Physiol. Renal. Physiol. 2001, 280, F365–F388. [Google Scholar] [CrossRef] [PubMed]
  45. Tostes, R.; Nigro, D.; Fortes, Z.; Carvalho, M. Effects of estrogen on the vascular system. Braz. J. Med. Biol. Res. 2003, 36, 1143–1158. [Google Scholar] [CrossRef] [PubMed]
  46. Khalil, R.A. Sex hormones as potential modulators of vascular function in hypertension. Hypertension 2005, 46, 249–254. [Google Scholar] [CrossRef] [PubMed]
  47. Mankhey, R.W.; Bhatti, F.; Maric, C. 17beta-Estradiol replacement improves renal function and pathology associated with diabetic nephropathy. Am. J. Physiol. Renal. Physiol. 2005, 288, F399–F405. [Google Scholar] [CrossRef]
  48. Sakemi, T.; Ohtsuka, N.; Tomiyoshi, Y.; Morito, F. The ovaries attenuate the aggravating effect of testosterone on glomerular injury in Adriamycin-induced nephropathy of female rats. Kidney Blood Press Res. 1997, 20, 44–50. [Google Scholar] [CrossRef]
  49. Corona, G.; Giagulli, V.A.; Maseroli, E.; Vignozzi, L.; Aversa, A.; Zitzmann, M.; Saad, F.; Mannucci, E.; Maggi, M. Testosterone supplementation and body composition: Results from a meta-analysis of observational studies. J. Endocrinol. Invest. 2016, 39, 967–981. [Google Scholar] [CrossRef]
  50. Buvat, J.; Maggi, M.; Guay, A.; Torres, L.O. Testosterone deficiency in men: Systematic review and standard operating procedures for diagnosis and treatment. J. Sex. Med. 2013, 10, 245–284. [Google Scholar] [CrossRef]
  51. Liu, P.Y.; Death, A.K.; Handelsman, D.J. Androgens and cardiovascular disease. Endocr. Rev. 2003, 24, 313–340. [Google Scholar] [CrossRef] [PubMed]
  52. Biswas, M.; Hampton, D.; Turkes, A.; Newcombe, R.G.; Rees, D.A. Reduced total testosterone concentrations in young healthy South Asian men are partly explained by increased insulin resistance but not by altered adiposity. Clin. Endocrinol. 2010, 73, 457–462. [Google Scholar] [CrossRef] [PubMed]
  53. Reckelhoff, J.F. Androgens and Blood Pressure Control: Sex Differences and Mechanisms. Mayo Clin. Proc. 2019, 94, 536–543. [Google Scholar] [PubMed]
  54. Davis, D.D.; Lopez Ruiz, A.; Yanes, L.L.; Iliescu, R.; Yuan, K.; Moulana, M.; Racusen, L.C.; Reckelhoff, J.F. Testosterone supplementation in male obese Zucker rats reduces body weight and improves insulin sensitivity but increases blood pressure. Hypertension 2012, 59, 726–731. [Google Scholar] [CrossRef] [PubMed]
  55. Yanes, L.L.; Romero, D.G.; Moulana, M.; Lima, R.; Davis, D.D.; Zhang, H.; Lockhart, R.; Racusen, L.C.; Reckelhoff, J.F. Cardiovascular-renal and metabolic characterization of a rat model of polycystic ovary syndrome. Gend. Med. 2011, 8, 103–115. [Google Scholar] [CrossRef] [PubMed]
  56. Reckelhoff, J.F.; Granger, J.P. Role of androgens in mediating hypertension and renal injury. Clin. Exp. Pharmacol. Physiol. 1999, 26, 127–131. [Google Scholar] [CrossRef] [PubMed]
  57. Guyton, A.C.; Coleman, T.G.; Cowley, A.W.; Scheel, K.W.; Manning, R.D.; Norman, R.A. Arterial pressure regulation. Overriding dominance of the kidneys in long-term regulation and in hypertension. Am. J. Med. 1972, 52, 584–594. [Google Scholar] [CrossRef] [PubMed]
  58. Wadei, H.M.; Textor, S.C. The role of the kidney in regulating arterial blood pressure. Nat. Rev. Nephrol. 2012, 8, 602–609. [Google Scholar] [CrossRef]
  59. Rettig, R.; Folberth, C.G.; Stauss, H.; Kopf, D.; Waldherr, R.; Baldauf, G.; Unger, T. Hypertension in rats induced by renal grafts from renovascular hypertensive donors. Hypertension 1990, 15, 429–435. [Google Scholar] [CrossRef]
  60. Hediger, M.A.; Romero, M.F.; Peng, J.B.; Rolfs, A.; Takanaga, H.; Bruford, E.A. The ABCs of solute carriers: Physiological, pathological and therapeutic implications of human membrane transport proteinsIntroduction. Pflugers Arch. 2004, 447, 465–468. [Google Scholar] [CrossRef]
  61. Morris, M.E.; Lee, H.J.; Predko, L.M. Gender differences in the membrane transport of endogenous and exogenous compounds. Pharmacol. Rev. 2003, 55, 229–240. [Google Scholar] [CrossRef]
  62. Rizwan, A.N.; Burckhardt, G. Organic anion transporters of the SLC22 family: Biopharmaceutical, physiological, and pathological roles. Pharm. Res. 2007, 24, 450–470. [Google Scholar] [CrossRef] [PubMed]
  63. Wright, S.H. Role of organic cation transporters in the renal handling of therapeutic agents and xenobiotics. Toxicol. Appl. Pharmacol. 2005, 204, 309–319. [Google Scholar] [CrossRef] [PubMed]
  64. Quinkler, M.; Bumke-Vogt, C.; Meyer, B.; Bähr, V.; Oelkers, W.; Diederich, S. The human kidney is a progesterone-metabolizing and androgen-producing organ. J. Clin. Endocrinol. Metab. 2003, 88, 2803–2809. [Google Scholar] [CrossRef] [PubMed]
  65. Grimont, A.; Bloch-Faure, M.; El Abida, B.; Crambert, G. Mapping of sex hormone receptors and their modulators along the nephron of male and female mice. FEBS Lett. 2009, 583, 1644–1648. [Google Scholar] [CrossRef] [PubMed]
  66. Takeda, H.; Mizuno, T.; Lasnitzki, I. Autoradiographic studies of androgen-binding sites in the rat urogenital sinus and postnatal prostate. J. Endocrinol. 1985, 104, 87–92. [Google Scholar] [CrossRef] [PubMed]
  67. Boulkroun, S.; Le Moellic, C.; Blot-Chabaud, M.; Farman, N.; Courtois-Coutry, N. Expression of androgen receptor and androgen regulation of NDRG2 in the rat renal collecting duct. Pflugers Arch. 2005, 451, 388–394. [Google Scholar] [CrossRef] [PubMed]
  68. Toot, J.; Jenkins, C.; Dunphy, G.; Boehme, S.; Hart, M.; Milsted, A.; Turner, M.; Ely, D. Testosterone influences renal electrolyte excretion in SHR/y and WKY males. BMC Physiol. 2008, 8, 5. [Google Scholar] [CrossRef] [PubMed]
  69. Khraibi, A.A.; Liang, M.; Berndt, T.J. Role of gender on renal interstitial hydrostatic pressure and sodium excretion in rats. Am. J. Hypertens. 2001, 14, 893–896. [Google Scholar] [CrossRef]
  70. Hilliard, L.M.; Nematbakhsh, M.; Kett, M.M.; Teichman, E.; Sampson, A.K.; Widdop, R.E.; Evans, R.G.; Denton, K.M. Gender differences in pressure-natriuresis and renal autoregulation: Role of the Angiotensin type 2 receptor. Hypertension 2011, 57, 275–282. [Google Scholar] [CrossRef]
  71. Hall, J.E.; Mizelle, H.L.; Hildebrandt, D.A.; Brands, M.W. Abnormal pressure natriuresis. A cause or a consequence of hypertension? Hypertension 1990, 15, 547–559. [Google Scholar] [CrossRef] [PubMed]
  72. Weinberger, M.H.; Fineberg, N.S. Sodium and volume sensitivity of blood pressure. Age and pressure change over time. Hypertension 1991, 18, 67–71. [Google Scholar] [CrossRef] [PubMed]
  73. Pechere-Bertschi, A.; Burnier, M. Gonadal steroids, salt-sensitivity and renal function. Curr. Opin. Nephrol. Hypertens. 2007, 16, 16–21. [Google Scholar] [CrossRef] [PubMed]
  74. Maranon, R.; Lima, R.; Spradley, F.T.; Carmo, J.M.D.; Zhang, H.; Smith, A.D.; Bui, E.; Thomas, R.L.; Moulana, M.; Hall, J.E.; et al. Roles for the sympathetic nervous system, renal nerves, and CNS melanocortin-4 receptor in the elevated blood pressure in hyperandrogenemic female rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 308, R708–R713. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, L.; Wang, X.; Qu, H.Y.; Jiang, S.; Zhang, J.; Fu, L.; Buggs, J.; Pang, B.; Wei, J.; Liu, R. Role of Kidneys in Sex Differences in Angiotensin II-Induced Hypertension. Hypertension 2017, 70, 1219–1227. [Google Scholar] [CrossRef] [PubMed]
  76. Inoue, A.; Yanagisawa, M.; Kimura, S.; Kasuya, Y.; Miyauchi, T.; Goto, K.; Masaki, T. The human endothelin family: Three structurally and pharmacologically distinct isopeptides predicted by three separate genes. Proc. Natl. Acad. Sci. USA 1989, 86, 2863–2867. [Google Scholar] [CrossRef] [PubMed]
  77. Nakano, D.; Pollock, D.M. Contribution of endothelin A receptors in endothelin 1-dependent natriuresis in female rats. Hypertension 2009, 53, 324–330. [Google Scholar] [CrossRef]
  78. Tostes, R.C.; Fortes, Z.B.; Callera, G.E.; Montezano, A.C.; Touyz, R.M.; Webb, R.C.; Carvalho, M.H.C. Endothelin, sex and hypertension. Clin. Sci. 2008, 114, 85–97. [Google Scholar] [CrossRef] [PubMed]
  79. Kohan, D.E. The renal medullary endothelin system in control of sodium and water excretion and systemic blood pressure. Curr. Opin. Nephrol. Hypertens. 2006, 15, 34–40. [Google Scholar] [CrossRef]
  80. Granger, J.P.; Abram, S.; Stec, D.; Chandler, D.; Speed, J.; LaMarca, B. Endothelin, the kidney, and hypertension. Curr. Hypertens. Rep. 2006, 8, 298–303. [Google Scholar] [CrossRef]
  81. Ge, Y.; Bagnall, A.; Stricklett, P.K.; Webb, D.; Kotelevtsev, Y.; Kohan, D.E. Combined knockout of collecting duct endothelin A and B receptors causes hypertension and sodium retention. Am. J. Physiol. Renal. Physiol. 2008, 295, F1635–F1640. [Google Scholar] [CrossRef] [PubMed]
  82. Ge, Y.; Bagnall, A.; Stricklett, P.K.; Strait, K.; Webb, D.J.; Kotelevtsev, Y.; Kohan, D.E. Collecting duct-specific knockout of the endothelin B receptor causes hypertension and sodium retention. Am. J. Physiol. Renal. Physiol. 2006, 291, F1274–F1280. [Google Scholar] [CrossRef] [PubMed]
  83. Ahn, D.; Ge, Y.; Stricklett, P.K.; Gill, P.; Taylor, D.; Hughes, A.K.; Yanagisawa, M.; Miller, L.; Nelson, R.D.; Kohan, D.E. Collecting duct-specific knockout of endothelin-1 causes hypertension and sodium retention. J. Clin. Invest. 2004, 114, 504–511. [Google Scholar] [CrossRef] [PubMed]
  84. Pearson, L.J.; Yandle, T.G.; Nicholls, M.G.; Evans, J.J. Regulation of endothelin-1 release from human endothelial cells by sex steroids and angiotensin-II. Peptides 2008, 29, 1057–1061. [Google Scholar] [CrossRef] [PubMed]
  85. Esposito, C.; Dal Canton, A. Functional changes in the aging kidney. J. Nephrol. 2010, 23, S41–S45. [Google Scholar] [PubMed]
  86. Burrell, L.M.; Johnston, C.I. Angiotensin II receptor antagonists. Potential in elderly patients with cardiovascular disease. Drugs Aging 1997, 10, 421–434. [Google Scholar] [CrossRef] [PubMed]
  87. Benigni, A.; Corna, D.; Zoja, C.; Sonzogni, A.; Latini, R.; Salio, M.; Conti, S.; Rottoli, D.; Longaretti, L.; Cassis, P.; et al. Disruption of the Ang II type 1 receptor promotes longevity in mice. J. Clin. Invest. 2009, 119, 524–530. [Google Scholar] [CrossRef] [PubMed]
  88. Basso, N.; Paglia, N.; Stella, I.; de Cavanagh, E.M.; Ferder, L.; Arnaiz, M.d.R.L.; Inserra, F. Protective effect of the inhibition of the renin-angiotensin system on aging. Regul. Pept. 2005, 128, 247–252. [Google Scholar] [CrossRef] [PubMed]
  89. Jones, E.S.; Black, M.J.; Widdop, R.E. Influence of Angiotensin II Subtype 2 Receptor (AT(2)R) Antagonist, PD123319, on Cardiovascular Remodelling of Aged Spontaneously Hypertensive Rats during Chronic Angiotensin II Subtype 1 Receptor (AT(1)R) Blockade. Int. J. Hypertens. 2012, 2012, 543062. [Google Scholar] [CrossRef]
  90. Polderman, K.H.; Stehouwer, C.D.A.; van Kamp, G.J.; Dekker, G.A.; Verheugt, F.W.A.; Gooren, L.J.G. Influence of sex hormones on plasma endothelin levels. Ann. Intern. Med. 1993, 118, 429–432. [Google Scholar] [CrossRef]
  91. Kumanov, P.; Tomova, A.; Kirilov, G. Testosterone replacement therapy in male hypogonadism is not associated with increase of endothelin-1 levels. Int. J. Androl. 2007, 30, 41–47. [Google Scholar] [CrossRef]
  92. Montezano, A.; Callera, G.; Mota, A.; Fortes, Z.; Nigro, D.; Carvalho, M.; Zorn, T.; Tostes, R. Endothelin-1 contributes to the sexual differences in renal damage in DOCA-salt rats. Peptides 2005, 26, 1454–1462. [Google Scholar] [CrossRef] [PubMed]
  93. Kellogg, D.L.; Liu, Y., Jr.; Pergola, P.E. Selected contribution: Gender differences in the endothelin-B receptor contribution to basal cutaneous vascular tone in humans. J. Appl. Physiol. 2001, 91, 2407–2411. [Google Scholar] [CrossRef]
  94. Kawanishi, H.; Hasegawa, Y.; Nakano, D.; Ohkita, M.; Takaoka, M.; Ohno, Y.; Matsumura, Y. Involvement of the endothelin ET(B) receptor in gender differences in deoxycorticosterone acetate-salt-induced hypertension. Clin. Exp. Pharmacol. Physiol. 2007, 34, 280–285. [Google Scholar] [CrossRef]
  95. Kittikulsuth, W.; Pollock, J.S.; Pollock, D.M. Sex differences in renal medullary endothelin receptor function in angiotensin II hypertensive rats. Hypertension 2011, 58, 212–218. [Google Scholar] [CrossRef]
  96. Rafiq, K.; Noma, T.; Fujisawa, Y.; Ishihara, Y.; Arai, Y.; Nabi, A.N.; Suzuki, F.; Nagai, Y.; Nakano, D.; Hitomi, H.; et al. Renal sympathetic denervation suppresses de novo podocyte injury and albuminuria in rats with aortic regurgitation. Circulation 2012, 125, 1402–1413. [Google Scholar] [CrossRef] [PubMed]
  97. Chun, T.-Y.; Bankir, L.; Eckert, G.J.; Bichet, D.G.; Saha, C.; Zaidi, S.-A.; Wagner, M.A.; Pratt, J.H. Ethnic differences in renal responses to furosemide. Hypertension 2008, 52, 241–248. [Google Scholar] [CrossRef] [PubMed]
  98. Sarafidis, P.A.; Li, S.; Chen, S.-C.; Collins, A.J.; Brown, W.W.; Klag, M.J.; Bakris, G.L. Hypertension awareness, treatment, and control in chronic kidney disease. Am. J. Med. 2008, 121, 332–340. [Google Scholar] [CrossRef] [PubMed]
  99. Johnston, J.G.; Speed, J.S.; Jin, C.; Pollock, D.M.; Ryan, M.J.; Sullivan, J.C.; Douma, L.G.; Solocinski, K.; Holzworth, M.R.; Crislip, G.R.; et al. Loss of endothelin B receptor function impairs sodium excretion in a time- and sex-dependent manner. Am. J. Physiol. Renal. Physiol. 2016, 311, F991–F998. [Google Scholar] [CrossRef]
  100. Nakano, D.; Pollock, J.S.; Pollock, D.M. Renal medullary ETB receptors produce diuresis and natriuresis via NOS1. Am. J. Physiol. Renal. Physiol. 2008, 294, F1205-11. [Google Scholar] [CrossRef]
  101. Gohar, E.Y.; Speed, J.S.; Kasztan, M.; Jin, C.; Pollock, D.M.; Becker, B.K. Activation of purinergic receptors (P2) in the renal medulla promotes endothelin-dependent natriuresis in male rats. Am. J. Physiol. Renal. Physiol. 2016, 311, F260–F267. [Google Scholar] [CrossRef] [PubMed]
  102. Gohar, E.Y.; Kasztan, M.; Becker, B.K.; Speed, J.S.; Pollock, D.M. Ovariectomy uncovers purinergic receptor activation of endothelin-dependent natriuresis. Am. J. Physiol. Renal. Physiol. 2017, 313, F361–F369. [Google Scholar] [CrossRef] [PubMed]
  103. Oloyo, A.K.; Sofola, O.A.; Yakubu, M.A. Orchidectomy attenuates high-salt diet-induced increases in blood pressure, renovascular resistance, and hind limb vascular dysfunction: Role of testosterone. Clin. Exp. Pharmacol. Physiol. 2016, 43, 825–833. [Google Scholar] [CrossRef]
  104. Taylor, T.A.; Gariepy, C.E.; Pollock, D.M.; Pollock, J.S. Gender differences in ET and NOS systems in ETB receptor-deficient rats: Effect of a high salt diet. Hypertension 2003, 41, 657–662. [Google Scholar] [CrossRef] [PubMed]
  105. Brinson, K.N.; Rafikova, O.; Sullivan, J.C. Female sex hormones protect against salt-sensitive hypertension but not essential hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R149–R157. [Google Scholar] [CrossRef] [PubMed]
  106. Gohar, E.Y.; Cook, A.K.; Pollock, D.M.; Inscho, E.W. Afferent arteriole responsiveness to endothelin receptor activation: Does sex matter? Biol. Sex. Differ. 2019, 10, 1. [Google Scholar] [CrossRef]
  107. Saleh, M.A.; Sandoval, R.M.; Rhodes, G.J.; Campos-Bilderback, S.B.; Molitoris, B.A.; Pollock, D.M. Chronic endothelin-1 infusion elevates glomerular sieving coefficient and proximal tubular albumin reuptake in the rat. Life Sci. 2012, 91, 634–637. [Google Scholar] [CrossRef] [PubMed]
  108. Saleh, M.A.; Boesen, E.I.; Pollock, J.S.; Savin, V.J.; Pollock, D.M. Endothelin-1 increases glomerular permeability and inflammation independent of blood pressure in the rat. Hypertension 2010, 56, 942–949. [Google Scholar] [CrossRef]
  109. Heimlich, J.B.; Speed, J.S.; Bloom, C.J.; O’Connor, P.M.; Pollock, J.S.; Pollock, D.M. ET-1 increases reactive oxygen species following hypoxia and high-salt diet in the mouse glomerulus. Acta Physiol. 2015, 213, 722–730. [Google Scholar] [CrossRef]
  110. Schneider, M.P.; Ge, Y.; Pollock, D.M.; Pollock, J.S.; Kohan, D.E. Collecting duct-derived endothelin regulates arterial pressure and Na excretion via nitric oxide. Hypertension 2008, 51, 1605–1610. [Google Scholar] [CrossRef]
  111. Kalk, P.; Thöne-Reineke, C.; Schwarz, A.; Godes, M.; Bauer, C.; Pfab, T.; Hocher, B. Renal phenotype of ET-1 transgenic mice is modulated by androgens. Eur. J. Med. Res. 2009, 14, 55–58. [Google Scholar] [CrossRef] [PubMed]
  112. Stringer, K.D.; Komers, R.; Osman, S.A.; Oyama, T.T.; Lindsley, J.N.; Anderson, S. Gender hormones and the progression of experimental polycystic kidney disease. Kidney Int. 2005, 68, 1729–1739. [Google Scholar] [CrossRef] [PubMed]
  113. Takaoka, M.; Yuba, M.; Fujii, T.; Ohkita, M.; Matsumura, Y. Oestrogen protects against ischaemic acute renal failure in rats by suppressing renal endothelin-1 overproduction. Clin. Sci. 2002, 103, 434S–437S. [Google Scholar] [CrossRef]
  114. De Miguel, C.; Speed, J.S.; Kasztan, M.; Gohar, E.Y.; Pollock, D.M. Endothelin-1 and the kidney: New perspectives and recent findings. Curr. Opin. Nephrol. Hypertens. 2016, 25, 35–41. [Google Scholar] [CrossRef]
  115. Yanes, L.L.; Romero, D.G.; Cucchiarelli, V.E.; Fortepiani, L.A.; Gomez-Sanchez, C.E.; Santacruz, F.; Reckelhoff, J.F. Role of endothelin in mediating postmenopausal hypertension in a rat model. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R229–R233. [Google Scholar] [CrossRef]
  116. Kontula, K.K.; Seppanen, P.J.; VAN Duyne, P.; Bardin, C.W.; Janne, O.A. Effect of a nonsteroidal antiandrogen, flutamide, on androgen receptor dynamics and ornithine decarboxylase gene expression in mouse kidney. Endocrinology 1985, 116, 226–233. [Google Scholar] [CrossRef] [PubMed]
  117. Riazi, S.; Madala-Halagappa, V.K.; Hu, X.; Ecelbarger, C.A. Sex and body-type interactions in the regulation of renal sodium transporter levels, urinary excretion, and activity in lean and obese Zucker rats. Gend. Med. 2006, 3, 309–327. [Google Scholar] [CrossRef]
  118. Veiras, L.C.; Girardi, A.C.; Curry, J.; Pei, L.; Ralph, D.L.; Tran, A.; Castelo-Branco, R.C.; Pastor-Soler, N.; Arranz, C.T.; Yu, A.S.; et al. Sexual Dimorphic Pattern of Renal Transporters and Electrolyte Homeostasis. J. Am. Soc. Nephrol. 2017, 28, 3504–3517. [Google Scholar] [CrossRef]
  119. Wilson, D.K.; Bayer, L.; Sica, D.A. Variability in salt sensitivity classifications in black male versus female adolescents. Hypertension 1996, 28, 250–255. [Google Scholar] [CrossRef]
  120. Crofton, J.T.; Share, L. Gonadal hormones modulate deoxycorticosterone-salt hypertension in male and female rats. Hypertension 1997, 29, 494–499. [Google Scholar] [CrossRef]
  121. Quan, A.; Chakravarty, S.; Chen, J.-C.; Loleh, S.; Saini, N.; Harris, R.C.; Capdevila, J.; Quigley, R. Androgens augment proximal tubule transport. Am. J. Physiol. Renal. Physiol. 2004, 287, F452–F459. [Google Scholar] [CrossRef] [PubMed]
  122. Quinkler, M.; Bujalska, I.J.; Kaur, K.; Onyimba, C.U.; Buhner, S.; Allolio, B.; Hughes, S.V.; Hewison, M.; Stewart, P.M. Androgen receptor-mediated regulation of the alpha-subunit of the epithelial sodium channel in human kidney. Hypertension 2005, 46, 787–798. [Google Scholar] [CrossRef] [PubMed]
  123. Loh, S.Y.; Giribabu, N.; Salleh, N. Sub-chronic testosterone treatment increases the levels of epithelial sodium channel (ENaC)-alpha, beta and gamma in the kidney of orchidectomized adult male Sprague-Dawley rats. PeerJ 2016, 4, e2145. [Google Scholar] [CrossRef] [PubMed]
  124. Quinn, S.; Harvey, B.J.; Thomas, W. Rapid aldosterone actions on epithelial sodium channel trafficking and cell proliferation. Steroids 2014, 81, 43–48. [Google Scholar] [CrossRef] [PubMed]
  125. Reckelhoff, J.F.; Zhang, H.; Srivastava, K.; Granger, J.P. Gender differences in hypertension in spontaneously hypertensive rats: Role of androgens and androgen receptor. Hypertension 1999, 34, 920–923. [Google Scholar] [CrossRef] [PubMed]
  126. Masilamani, S.; Kim, G.H.; Mitchell, C.; Wade, J.B.; Knepper, M.A. Aldosterone-mediated regulation of ENaC alpha, beta, and gamma subunit proteins in rat kidney. J. Clin. Invest. 1999, 104, R19–R23. [Google Scholar] [CrossRef]
  127. Riazi, S.; Maric, C.; Ecelbarger, C.A. 17-beta Estradiol attenuates streptozotocin-induced diabetes and regulates the expression of renal sodium transporters. Kidney Int. 2006, 69, 471–480. [Google Scholar] [CrossRef] [PubMed]
  128. Gambling, L.; Dunford, S.; Wilson, C.A.; McArdle, H.J.; Baines, D.L. Estrogen and progesterone regulate alpha, beta, and gammaENaC subunit mRNA levels in female rat kidney. Kidney Int. 2004, 65, 1774–1781. [Google Scholar] [CrossRef] [PubMed]
  129. Chang, C.-T.; Sun, C.-Y.; Pong, C.-Y.; Chen, Y.-C.; Lin, G.-P.; Chang, T.-C.; Wu, M.-S. Interaction of estrogen and progesterone in the regulation of sodium channels in collecting tubular cells. Chang Gung Med. J. 2007, 30, 305–312. [Google Scholar] [PubMed]
  130. Arias-Loza, P.A.; Muehlfelder, M.; Elmore, S.A.; Maronpot, R.; Hu, K.; Blode, H.; Hegele-Hartung, C.; Fritzemeier, K.H.; Ertl, G.; Pelzer, T. Differential effects of 17beta-estradiol and of synthetic progestins on aldosterone-salt-induced kidney disease. Toxicol. Pathol. 2009, 37, 969–982. [Google Scholar] [CrossRef]
  131. Pearce, D.; Soundararajan, R.; Trimpert, C.; Kashlan, O.B.; Deen, P.M.; Kohan, D.E. Collecting duct principal cell transport processes and their regulation. Clin. J. Am. Soc. Nephrol. 2015, 10, 135–146. [Google Scholar] [CrossRef] [PubMed]
  132. Loh, S.Y.; Giribabu, N.; Salleh, N. Effects of gonadectomy and testosterone treatment on aquaporin expression in the kidney of normotensive and hypertensive rats. Exp. Biol. Med. 2017, 242, 1376–1386. [Google Scholar] [CrossRef] [PubMed]
  133. Loh, S.Y.; Giribabu, N.; Gholami, K.; Salleh, N. Effects of testosterone on mean arterial pressure and aquaporin (AQP)-1, 2, 3, 4, 6 and 7 expressions in the kidney of orchidectomized, adult male Sprague-Dawley rats. Arch. Biochem. Biophys. 2017, 614, 41–49. [Google Scholar] [CrossRef] [PubMed]
  134. Ellis, B.; Li, X.C.; Miguel-Qin, E.; Gu, V.; Zhuo, J.L. Evidence for a functional intracellular angiotensin system in the proximal tubule of the kidney. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2012, 302, R494–R509. [Google Scholar] [CrossRef] [PubMed]
  135. Mačković, M.; Zimolo, Z.; Burckhardt, G.; Sabolić, I. Isolation of renal brush-border membrane vesicles by a low-speed centrifugation; effect of sex hormones on Na+-H+ exchange in rat and mouse kidney. Biochim. Biophys. Acta 1986, 862, 141–152. [Google Scholar] [CrossRef] [PubMed]
  136. Carrisoza-Gaytan, R.; Ray, E.C.; Flores, D.; Marciszyn, A.L.; Wu, P.; Liu, L.; Subramanya, A.R.; Wang, W.; Sheng, S.; Nkashama, L.J.; et al. Intercalated cell BKalpha subunit is required for flow-induced K+ secretion. JCI Insight 2020, 5, e130553. [Google Scholar] [CrossRef] [PubMed]
  137. Ong, A.C.; Fine, L.G. Loss of glomerular function and tubulointerstitial fibrosis: Cause or effect? Kidney Int. 1994, 45, 345–351. [Google Scholar] [CrossRef] [PubMed]
  138. Wolf, G.; Neilson, E.G. Molecular mechanisms of tubulointerstitial hypertrophy and hyperplasia. Kidney Int. 1991, 39, 401–420. [Google Scholar] [CrossRef] [PubMed]
  139. Harris, D.C.; Chan, L.; Schrier, R.W. Remnant kidney hypermetabolism and progression of chronic renal failure. Am. J. Physiol. 1988, 254, F267-76. [Google Scholar] [CrossRef]
  140. Ellison, K.E.; Ingelfinger, J.R.; Pivor, M.; Dzau, V.J. Androgen regulation of rat renal angiotensinogen messenger RNA expression. J. Clin. Invest. 1989, 83, 1941–1945. [Google Scholar] [CrossRef]
  141. Givens, J.R.; Andersen, R.N.; Ragland, J.B.; Wiser, W.L.; Umstot, E.S. Adrenal function in hirsutism I. Diurnal change and response of plasma androstenedione, testosterone, 17-hydroxyprogesterone, cortisol, LH and FSH to dexamethasone and 1/2 unit of ACTH. J. Clin. Endocrinol. Metab. 1975, 40, 988–1000. [Google Scholar] [CrossRef]
  142. Chen, Y.F.; Naftilan, A.J.; Oparil, S. Androgen-dependent angiotensinogen and renin messenger RNA expression in hypertensive rats. Hypertension 1992, 19, 456–463. [Google Scholar] [CrossRef] [PubMed]
  143. Yanes, L.L.; Sartori-Valinotti, J.C.; Iliescu, R.; Romero, D.G.; Racusen, L.C.; Zhang, H.; Reckelhoff, J.F. Testosterone-dependent hypertension and upregulation of intrarenal angiotensinogen in Dahl salt-sensitive rats. Am. J. Physiol. Renal. Physiol. 2009, 296, F771–F779. [Google Scholar] [CrossRef]
  144. James, G.D.; Sealey, J.E.; Müller, F.; Alderman, M.; Madhavan, S.; Laragh, J.H. Renin relationship to sex, race and age in a normotensive population. J. Hypertens. Suppl. 1986, 4, S387–S389. [Google Scholar]
  145. Kienitz, T.; Quinkler, M. Testosterone and blood pressure regulation. Kidney Blood Press Res. 2008, 31, 71–79. [Google Scholar] [CrossRef] [PubMed]
  146. Kau, M.-M.; Lo, M.-J.; Wang, S.-W.; Tsai, S.-C.; Chen, J.-J.; Chiao, Y.-C.; Yeh, J.-Y.; Lin, H.; Shum, A.Y.-C.; Fang, V.S.; et al. Inhibition of aldosterone production by testosterone in male rats. Metabolism 1999, 48, 1108–1114. [Google Scholar] [CrossRef]
  147. Herak-Kramberger, C.M.; Breljak, D.; Ljubojević, M.; Matokanović, M.; Lovrić, M.; Rogić, D.; Brzica, H.; Vrhovac, I.; Karaica, D.; Micek, V.; et al. Sex-dependent expression of water channel AQP1 along the rat nephron. Am. J. Physiol. Renal. Physiol. 2015, 308, F809–F821. [Google Scholar] [CrossRef] [PubMed]
  148. Ebrahimian, T.; He, Y.; Schiffrin, E.L.; Touyz, R.M. Differential regulation of thioredoxin and NAD(P)H oxidase by angiotensin II in male and female mice. J. Hypertens. 2007, 25, 1263–1271. [Google Scholar] [CrossRef]
  149. Tatchum-Talom, R.; Eyster, K.M.; Martin, D.S. Sexual dimorphism in angiotensin II-induced hypertension and vascular alterations. Can. J. Physiol. Pharmacol. 2005, 83, 413–422. [Google Scholar] [CrossRef]
  150. Xue, B.; Pamidimukkala, J.; Hay, M. Sex differences in the development of angiotensin II-induced hypertension in conscious mice. Am. J. Physiol. Heart. Circ. Physiol. 2005, 288, H2177–H2184. [Google Scholar] [CrossRef]
  151. Sartori-Valinotti, J.C.; Iliescu, R.; Yanes, L.L.; Dorsett-Martin, W.; Reckelhoff, J.F. Sex differences in the pressor response to angiotensin II when the endogenous renin-angiotensin system is blocked. Hypertension 2008, 51, 1170–1176. [Google Scholar] [CrossRef] [PubMed]
  152. Miller, J.A.; Anacta, L.A.; Cattran, D.C. Impact of gender on the renal response to angiotensin II. Kidney Int. 1999, 55, 278–285. [Google Scholar] [CrossRef]
  153. Howard, C.G.; Mitchell, K.D. Renal functional responses to selective intrarenal renin inhibition in Cyp1a1-Ren2 transgenic rats with ANG II-dependent malignant hypertension. Am. J. Physiol. Renal. Physiol. 2012, 302, F52–F59. [Google Scholar] [CrossRef]
  154. Machnik, A.; Neuhofer, W.; Jantsch, J.; Dahlmann, A.; Tammela, T.; Machura, K.; Park, J.-K.; Beck, F.-X.; Müller, D.N.; Derer, W.; et al. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat. Med. 2009, 15, 545–552. [Google Scholar] [CrossRef] [PubMed]
  155. Rands, V.F.; Seth, D.M.; Kobori, H.; Prieto, M.C. Sexual dimorphism in urinary angiotensinogen excretion during chronic angiotensin II-salt hypertension. Gend. Med. 2012, 9, 207–218. [Google Scholar] [CrossRef]
  156. Ji, H.; Menini, S.; Zheng, W.; Pesce, C.; Wu, X.; Sandberg, K. Role of angiotensin-converting enzyme 2 and angiotensin(1-7) in 17beta-oestradiol regulation of renal pathology in renal wrap hypertension in rats. Exp. Physiol. 2008, 93, 648–657. [Google Scholar] [CrossRef]
  157. Brosnihan, K.B.; Li, P.; Ganten, D.; Ferrario, C.M. Estrogen protects transgenic hypertensive rats by shifting the vasoconstrictor-vasodilator balance of RAS. Am. J. Physiol. 1997, 273, R1908–R1915. [Google Scholar] [CrossRef]
  158. Sampson, A.K.; Moritz, K.M.; Denton, K.M. Postnatal ontogeny of angiotensin receptors and ACE2 in male and female rats. Gend. Med. 2012, 9, 21–32. [Google Scholar] [CrossRef]
  159. Sampson, A.K.; Moritz, K.M.; Jones, E.S.; Flower, R.L.; Widdop, R.E.; Denton, K.M. Enhanced angiotensin II type 2 receptor mechanisms mediate decreases in arterial pressure attributable to chronic low-dose angiotensin II in female rats. Hypertension 2008, 52, 666–671. [Google Scholar] [CrossRef] [PubMed]
  160. Baiardi, G.; Macova, M.; Armando, I.; Ando, H.; Tyurmin, D.; Saavedra, J.M. Estrogen upregulates renal angiotensin II AT1 and AT2 receptors in the rat. Regul. Pept. 2005, 124, 7–17. [Google Scholar] [CrossRef]
  161. Dean, S.A.; Tan, J.; O’Brien, E.R.; Leenen, F.H. 17beta-estradiol downregulates tissue angiotensin-converting enzyme and ANG II type 1 receptor in female rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R759–R766. [Google Scholar] [CrossRef] [PubMed]
  162. Hilliard, L.M.; Sampson, A.K.; Brown, R.D.; Denton, K.M. The “his and hers” of the renin-angiotensin system. Curr. Hypertens. Rep. 2013, 15, 71–79. [Google Scholar] [CrossRef]
  163. Abadir, P.M.; Carey, R.M.; Siragy, H.M. Angiotensin AT2 receptors directly stimulate renal nitric oxide in bradykinin B2-receptor-null mice. Hypertension 2003, 42, 600–604. [Google Scholar] [CrossRef] [PubMed]
  164. Brown, R.D.; Hilliard, L.M.; Head, G.A.; Jones, E.S.; Widdop, R.E.; Denton, K.M. Sex differences in the pressor and tubuloglomerular feedback response to angiotensin II. Hypertension 2012, 59, 129–135. [Google Scholar] [CrossRef]
  165. Os, I.; Franco, V.; Kjeldsen, S.E.; Manhem, K.; Devereux, R.B.; Gerdts, E.; Hille, D.A.; Lyle, P.A.; Okin, P.M.; Dahlof, B.; et al. Effects of losartan in women with hypertension and left ventricular hypertrophy: Results from the Losartan Intervention for Endpoint Reduction in Hypertension Study. Hypertension 2008, 51, 1103–1108. [Google Scholar] [CrossRef] [PubMed]
  166. Hudson, M.; Rahme, E.; Behlouli, H.; Sheppard, R.; Pilote, L. Sex differences in the effectiveness of angiotensin receptor blockers and angiotensin converting enzyme inhibitors in patients with congestive heart failure--a population study. Eur. J. Heart Fail. 2007, 9, 602–609. [Google Scholar] [CrossRef] [PubMed]
  167. Reckelhoff, J.F.; Zhang, H.; Srivastava, K. Gender differences in development of hypertension in spontaneously hypertensive rats: Role of the renin-angiotensin system. Hypertension 2000, 35, 480–483. [Google Scholar] [CrossRef]
  168. Nakagawa, K.; Marji, J.S.; Schwartzman, M.L.; Waterman, M.R.; Capdevila, J.H. Androgen-mediated induction of the kidney arachidonate hydroxylases is associated with the development of hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2003, 284, R1055–R1062. [Google Scholar] [CrossRef] [PubMed]
  169. Maier, K.G.; Roman, R.J. Cytochrome P450 metabolites of arachidonic acid in the control of renal function. Curr. Opin. Nephrol. Hypertens. 2001, 10, 81–87. [Google Scholar] [CrossRef]
  170. Wu, C.C.; Schwartzman, M.L. The role of 20-HETE in androgen-mediated hypertension. Prostaglandins Other Lipid Mediat. 2011, 96, 45–53. [Google Scholar] [CrossRef]
  171. Cheema, M.U.; Irsik, D.L.; Wang, Y.; Miller-Little, W.; Hyndman, K.A.; Marks, E.S.; Frøkiær, J.; Boesen, E.I.; Norregaard, R. Estradiol regulates AQP2 expression in the collecting duct: A novel inhibitory role for estrogen receptor alpha. Am. J. Physiol. Renal. Physiol. 2015, 309, F305–F317. [Google Scholar] [CrossRef] [PubMed]
  172. O’Donald, P. “Haldane’s dilemma” and the rate of natural selection. Nature 1969, 221, 815–817. [Google Scholar] [CrossRef] [PubMed]
  173. Tran, N.T.; Aslibekyan, S.; Tiwari, H.K.; Zhi, D.; Sung, Y.J.; Hunt, S.C.; Rao, D.C.; Broeckel, U.; Judd, S.E.; Muntner, P.; et al. PCSK9 variation and association with blood pressure in African Americans: Preliminary findings from the HyperGEN and REGARDS studies. Front. Genet. 2015, 6, 136. [Google Scholar] [CrossRef] [PubMed]
  174. Ohlsson, T.; Lindgren, A.; Engström, G.; Jern, C.; Melander, O. A stop-codon of the phosphodiesterase 11A gene is associated with elevated blood pressure and measures of obesity. J. Hypertens. 2016, 34, 445–451. [Google Scholar] [CrossRef] [PubMed]
  175. Kim, Y.; Han, B.G.; KoGES Group. Cohort Profile: The Korean Genome and Epidemiology Study (KoGES) Consortium. Int. J. Epidemiol. 2017, 46, e20. [Google Scholar] [CrossRef] [PubMed]
  176. Yasukochi, Y.; Sakuma, J.; Takeuchi, I.; Kato, K.; Oguri, M.; Fujimaki, T.; Horibe, H.; Yamada, Y. Longitudinal exome-wide association study to identify genetic susceptibility loci for hypertension in a Japanese population. Exp. Mol. Med. 2017, 49, e409. [Google Scholar] [CrossRef] [PubMed]
  177. Knopp, R.H.; Paramsothy, P.; Retzlaff, B.M.; Fish, B.; Walden, C.; Dowdy, A.; Tsunehara, C.; Aikawa, K.; Cheung, M.C. Gender differences in lipoprotein metabolism and dietary response: Basis in hormonal differences and implications for cardiovascular disease. Curr. Atheroscler. Rep. 2005, 7, 472–479. [Google Scholar] [CrossRef]
  178. Lapointe, A.; Balk, E.M.; Lichtenstein, A.H. Gender differences in plasma lipid response to dietary fat. Nutr. Rev. 2006, 64, 234–249. [Google Scholar] [CrossRef] [PubMed]
  179. Chrysohoou, C.; Panagiotakos, D.B.; Pitsavos, C.; Kokkinos, P.; Marinakis, N.; Stefanadis, C.; Toutouzas, P.K. Gender differences on the risk evaluation of acute coronary syndromes: The CARDIO2000 study. Prev. Cardiol. 2003, 6, 71–77. [Google Scholar] [CrossRef]
  180. Abramson, B.L.; Melvin, R.G. Cardiovascular risk in women: Focus on hypertension. Can. J. Cardiol. 2014, 30, 553–559. [Google Scholar] [CrossRef]
Figure 1. Sex-specific pathophysiological differences in kidney function and blood pressure regulation.
Figure 1. Sex-specific pathophysiological differences in kidney function and blood pressure regulation.
Ijms 25 08637 g001
Figure 2. Overview of suggested sex differences in the renin–angiotensin–aldosterone system. Males typically have a more active ACE/Angiotensin II/AT1R pathway, increasing the risk of hypertension, while females tend to have a more active AT2R/MasR pathway, offering protective effects against hypertension.
Figure 2. Overview of suggested sex differences in the renin–angiotensin–aldosterone system. Males typically have a more active ACE/Angiotensin II/AT1R pathway, increasing the risk of hypertension, while females tend to have a more active AT2R/MasR pathway, offering protective effects against hypertension.
Ijms 25 08637 g002
Table 1. Important human studies on sexual dimorphism in kidney disease and hypertension.
Table 1. Important human studies on sexual dimorphism in kidney disease and hypertension.
StudyPopulationKey Findings
Messerli, F.H. et al. [11]Male and female patients with hypertensionFound that men have higher blood pressure compared to women starting at puberty, with this trend reversing after menopause.
van Kesteren, P.J.M [12]M—>F and F→M transexualsMale-to-female transsexuals exhibit lower ET levels after estrogen supplementation, whereas testosterone treatment in female-to-male transsexuals leads to an increase in ET levels.
Phillips, G.B. et al. [9]Men with coronary artery diseaseFound an association of hypotestosteronemia with coronary artery disease in men.
Christakou, C.D. et al. [13]Women with polycystic ovary syndromeDiscussed the role of androgen excess on metabolic aberrations and cardiovascular risk in women with PCOS.
Chen, W. et al. [14]Men and women without hypertension at baselineSuggested that the endothelial NO synthase gene may contribute to the predisposition of females for hypertension
Curtis, J.J. et al. [15]Hypertensive patients undergoing renal transplantationFound remission of essential hypertension after renal transplantation.
Zapater P. et al. [16]Men and women with hypertensionFound that women had lower blood pressure and angiotensin converting enzyme (ACE) activities than men
Ruggenenti P. et al. [17]Men and women with hypertensionFound that men exhibit a lower response to ACE inhibitor treatment
Cho, S.B. et al. [18]GWASIndicated a sex-specific genetic susceptibility to hypertension
Song, S. et al. [19]Men and women without hypertension at baselineFound that a diet rich in grains and legumes is inversely associated with the risk of hypertension in Korean women
Kang, Y. et al. [20]Men and women without hypertension at baselineFound that frequent fried food consumption is associated with hypertension in Korean women
Table 2. Sex-specific findings in various animal models of hypertension.
Table 2. Sex-specific findings in various animal models of hypertension.
Animal ModelSex-Specific FindingsReferences
Spontaneously Hypertensive Rats (SHRs)Testosterone mediates hypertension via AR; flutamide decreases blood pressure[21,22,23,24,25]
Deoxycorticosterone Acetate (DOCA)-Salt RatsHigher blood pressure elevation in males; Endothelin B (ETB) receptor knockout abolishes blood pressure differences[26]
Ren2 Gene-Expressing RatsFlutamide lowers blood pressure and reduces organ damage[27]
Orchidectomized Sprague Dawley RatsTestosterone increases ENaC expression; castration decreases blood pressure[28,29]
Ovariectomized RatsSalt-independent hypertension in DS rats post-ovariectomy[30,31]
Angiotensin II (Ang II)-Infused MiceBlood pressure increase in females with male kidney; Angiotensing 1 receptor (AT1R) and endothelin 1 (ET-1) mRNA differences[32]
High-Fat Diet (HFD)-Induced HypertensionHFD induces hypertension in males; losartan prevents this; and females resistant due to Ang (1–7)[33]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stamellou, E.; Sterzer, V.; Alam, J.; Roumeliotis, S.; Liakopoulos, V.; Dounousi, E. Sex-Specific Differences in Kidney Function and Blood Pressure Regulation. Int. J. Mol. Sci. 2024, 25, 8637. https://doi.org/10.3390/ijms25168637

AMA Style

Stamellou E, Sterzer V, Alam J, Roumeliotis S, Liakopoulos V, Dounousi E. Sex-Specific Differences in Kidney Function and Blood Pressure Regulation. International Journal of Molecular Sciences. 2024; 25(16):8637. https://doi.org/10.3390/ijms25168637

Chicago/Turabian Style

Stamellou, Eleni, Viktor Sterzer, Jessica Alam, Stefanos Roumeliotis, Vassilios Liakopoulos, and Evangelia Dounousi. 2024. "Sex-Specific Differences in Kidney Function and Blood Pressure Regulation" International Journal of Molecular Sciences 25, no. 16: 8637. https://doi.org/10.3390/ijms25168637

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop