Abstract
Hypertension affects one-third of adults in the Western world and is the most common independent risk factor for cardiovascular disease, and the leading cause of premature death globally. Despite available therapeutic options, approximately half of the hypertensive population taking medication does not achieve adequate blood pressure (BP) control leaving them at increased risk of chronic kidney disease, renal failure, stroke, congestive heart failure, myocardial infarction, aneurysm and peripheral artery disease. New therapeutic options need to be identified for the treatment of hypertension in order to increase the percentage of individuals with controlled BP. There is a growing basic science literature regarding the role of T-cells in the pathogenesis of hypertension and BP control; however, the majority of this literature has been performed exclusively in males despite the fact that both men and women develop hypertension. This is especially problematic since hypertension is well recognized as having distinct sex differences in the prevalence, absolute BP values and molecular mechanisms contributing to the pathophysiology of the disease. The purpose of this article is to review the available literature regarding sex differences in T-cells in hypertension followed by highlighting the potential pathways that may result in sex-specific effects on T-cell activation and differentiation.
Keywords: angiotensin, blood pressure regulation, inflammation, sex differences, Th17 cell, T regulatory cell
INTRODUCTION
Hypertension affects one-third of adults in the Western world and is the most common condition seen by primary care physicians [1]. Uncontrolled hypertension leads to chronic kidney disease, renal failure, stroke, congestive heart failure, myocardial infarction, aneurysm and peripheral artery disease, and is associated with significant morbidity and mortality [2]. Despite the prevalence, negative impact on overall health and economic burden of hypertension, the pathogenesis of essential hypertension remains poorly understood. As a result, only ~36 million of the ~70 million American adults that have hypertension have adequate control of their hypertension [1]. New therapeutic options need to be identified for the treatment of hypertension in order to increase the percentage of individuals with controlled blood pressure (BP) and decrease premature death rates associated with cardiovascular disease.
Hypertension is now considered a state of low-grade inflammation. Elevations in BP are associated with the up-regulation of pro-inflammatory cytokines and the infiltration of immune cells into target organs both clinically and in experimental models of hypertension. Although early studies in the 1960s broadly implicated the immune system in hypertension, they did not directly identify the immune component responsible (for reviews see [3–7]). Significant advances in immunology research and technology have allowed for the identification and targeting of specific cells and components of the immune system allowing for the direct study of immune cells in BP control. As a result, more recent studies have significantly expanded our understanding of the role of the immune system in BP regulation and suggest a direct contribution of T-cells to the progression and development of hypertension.
SEX DIFFERENCES IN HYPERTENSION
Hypertension is well recognized as having distinct sex differences in the prevalence, absolute BP values and molecular mechanisms contributing to the pathophysiology of the disease (for reviews see [8,9]). Beginning in adolescence, males have higher BP than females and these trends persist into the sixth decade of life, after which point more women are hypertensive than men [1,10]. There are also sex differences in the relationship between elevations in BP and the degree of end-organ damage and severity of cardiovascular risk; comparable decreases in BP in men and women may not offer the same degree of cardiovascular benefit to both sexes. Decreasing BP to 125/80 in hypertensive women is required to offer comparable reductions in cardiovascular events as decreasing BP in hypertensive men to 135/85 mmHg [11]. This finding is consistent with the report that hypertensive women exhibit a steeper relationship between systolic BP and risk for cardiovascular events than men, and a 1 S.D. decrease in BP in women results in a greater cardiovascular risk reduction in women compared with men [12]. These studies become particularly important following the recent release of data from the SPRINT trial which compared the impact of traditional compared with intensive BP control in hypertensive patients on cardiovascular morbidity and mortality. More intensive BP control (target systolic BP < 120 mmHg) offered significantly greater overall cardiovascular benefit compared with achieving target BPs of 140 mmHg [13]. Approximately 35% of the participants in the SPRINT trial were women, yet the data were not presented separately by sex, therefore it remains unknown whether there were sex differences in the benefits of more aggressive BP control. Despite all of these findings, the most recent guidelines for BP control released in 2014 recommend the same approach for treating men and women with hypertension [14]. However, only ~55% of hypertensive patients achieve BP controlled to recommended levels [1] and women have been reported to have lower rates of BP control compared with men despite being more likely to take their prescribed medication [15]. These numbers highlight the need for more studies to identify the molecular mechanisms driving increases in BP in hypertension, especially in women.
T-cells have been suggested to link multiple organs in the control of BP, and based on the data discussed below we propose that T-cells may hold the key to explaining the basis of sex differences in hypertension. Unfortunately, the vast majority of the studies implicating T-cells in hypertension have been conducted exclusively using male experimental animals despite the fact that half of the hypertensive population is female. Therefore, understanding the role of T-cells in hypertensive females is an important, although understudied, area of research. Better understanding of the role of T-cells and the immune system in the development and maintenance of hypertension may also provide novel therapies to improve BP control rates in both sexes. This review will begin with a discussion of what is currently known regarding sex differences in T-cells in hypertension followed by highlighting the potential pathways and molecular mechanisms which may contribute to sex differences in the T-cell profile and BP responses to T-cells.
T-CELLS IN EXPERIMENTAL HYPERTENSION
Over the last decade there have been an increasing number of studies defining a critical role for T-cells in hypertension. Although the suppression of lymphocytes (both B- and T-cells) using the immunosuppressant, mycophenolate mofetil (MMF) significantly reduces BP in numerous experimental models of hypertension in male rats [16–19], it was not until the adoptive transfer studies by Guzik et al. [20] that T-cells were directly implicated in hypertension. Adoptive transfer of Pan CD3+ T-cells, but not B-cells, restores hypertensive response to angiotensin (Ang) II and deoxycoticosterone acetate (DOCA) salt in male recombination-activating gene (Rag)−/− mice. These findings have been confirmed by additional studies in male severe combined immune deficiency (SCID) mice [21] and male Dahl salt-sensitive rats [22,23] and directly demonstrate that Pan CD3+ T-cells are required to develop and maintain a hypertensive response in male experimental animals.
Pan CD3+ T-cells can be separated into three different subpopulations: CD1d+ natural killer T (NKT) cells, CD8+ cytotoxic T-cells, and CD4+ T helper (Th) cells, all of which have been implicated in BP control. Relatively little is known regarding the role of NKT cells in essential hypertension; however, Vinh et al. [24] showed that NKT cells do not play a role in Ang II-induced hypertension in male mice. NKT cells will not be further discussed in this review. There is growing interest in CD8+ T-cells in hypertension [25,26]. Indeed, recent studies have shown that male CD8−/− mice, but not CD4−/− mice, exhibit a blunted hypertensive response to Ang II [27]. Moreover, adoptive transfer of CD8+ T-cells resulted in greater Ang II-induced increases in BP in male Rag−/− mice than adoptive transfer of CD4+ T-cells [27]. The authors concluded that renal CD8+ T-cells contribute to experimental hypertension by contributing to sodium and volume retention and vascular rarefaction. In a separate study, CD8−/− mice (the sex of the mice was not specified) exhibited comparable increases in BP following 7 days of Ang II infusion compared with wild-type mice; however, the CD8−/− mice exhibited attenuated cardiac inflammation following Ang II infusion [26]. It should be noted that this second study employed a much higher dose of Ang II (1500 ng/kg per min) than the first study (490 ng/kg per min), raising the possibility that the role of CD8+ T-cells in Ang II hypertension compared with Ang II-induced end-organ damage depends on the dose of Ang II; the higher dose may have resulted in a ceiling effect on BP masking a genotype effect. More studies need to be done to further examine the role of CD8+ T-cells in hypertension, as well as to examine CD8+ T-cell subtypes in hypertension and BP control.
The majority of experimental studies examining T-cell subtypes in hypertension have focused on CD4+ T-cells. CD4+ T-cells are crucial for the functioning of an intact immune system and play diverse roles in immune surveillance and protection against foreign pathogens. Naïve CD4+ T-cells differentiate into one of four main subtypes: Th1, Th2, Th17 or regulatory T-cells (Tregs); however, the most is known regarding the role of Th17 cells and Tregs in hypertension and related end-organ damage. For in-depth reviews on T-cell differentiation and T-cell subtypes see [28–32].
Th17 cells and hypertension
Th17 cells are pro-inflammatory T-cells that secrete the cytokines interleukin (IL)-17A, IL-17F, IL-21 and IL-22, and have been implicated in the pathogenesis of a number of autoimmune diseases (for reviews on Th17 cells see [33–35]). A role for Th17 cells in hypertension has been indirectly surmised based on studies manipulating IL-17 levels, a pro-inflammatory cytokine produced by Th17 cells. DOCA-salt increases BP, renal and cardiac IL-17 and Th17 cells in male Sprague–Dawley rats and treatment with an IL-17-neutralizing antibody attenuates increases in BP [36]. Moreover, Ang II-induced hypertension is not sustained on male IL-17−/− mice, supporting a pro-hypertensive role of Th17 cells [37]. However, DOCA-salt plus Ang II infusion results in comparable BP increases in male control (C57BL/6J mice), IL-17−/− and IL-23p19−/− mice (IL-23 is required for Th17 cell expansion) [38], calling into question the role of Th17 cells in mediating increases in BP. This may be related to the fact that, although Th17 cells are a primary source of IL-17, they are not the only immune cells to release IL-17. IL-17 is also produced by γ δT cells, NKT cells, neutrophils and eosinophils. Indeed, a recent study found that γ δT cells were responsible for Ang II-induced increases in cardiac IL-17A, and deletion of γ δT cell protected against Ang II-induced cardiac injury [38]. Additional studies using adoptive transfer of Th17 cells are needed to conclusively determine the impact of these cells on BP regulation.
Tregs and hypertension
Tregs suppress the function of other T-cells as well as macrophages and dendritic cells (DCs) to limit an immune response. As a result, Tregs are critical for maintaining immune homoeostasis, preventing autoimmune diseases and limiting chronic inflammatory diseases (for reviews on Tregs see [39,40]). In addition to indirect evidence linking Tregs to BP control [41], adoptive transfer studies have directly linked Tregs with improved cardiovascular outcomes and decreases in BP in male experimental animals. The first study to examine the direct impact of Tregs on BP and end-organ damage used male NMRI mice infused with Ang II [42]. Mice received 2×105 Tregs via intravenous injection on day 1 of Ang II infusion. Although Ang II-induced increases in BP were not altered by adoptive transfer of Tregs over the course of the 2 week study, Tregs attenuated Ang II-induced cardiac hypertrophy, fibrosis and electric remodelling. Consistent with this finding, intravenous injection of 1×106 Tregs 1 h after transaortic constriction attenuates cardiac fibrosis with no difference in BP between male mice that received Tregs or vehicle 14 days later; BP was only measured at the end of the study so it is unknown whether Tregs altered the time course by which BP increased [43]. In contrast, intraperitoneal injection of 2 × 106 Tregs three times a week for 2 weeks attenuates Ang II hypertension in male C57Bl/6J mice and improves endothelium-dependent dilation compared with control mice [44,45]. Similarly, tail vein injection of 3×105 Tregs two to three times over a 2 week period attenuates Ang II and aldosterone induced-hypertension and vascular endothelial dysfunction in male C57Bl6 mice [46,47]. Treg supplementation also attenuates increases in BP in male athymic nude mice predisposed to develop pulmonary hypertension [48]. Therefore, there is consistent reporting of a cardiovascular protective role for Tregs, although adoptive transfer of more Tregs at more regular intervals may be required to observe a BP effect. Regardless, these data support the notion that Tregs would be an attractive therapeutic target to improve BP control and attenuate hypertension-induced end-organ damage.
Th17 cells and Tregs: is it all about balance?
The above discussion regarding the proposed roles of Th17 cells compared with Tregs on BP control suggests that the balance between these two T-cell subtypes is a critical determinant of the impact of T-cell activation on cardiovascular health. However, recent evidence suggests that it may not be this simple as additional populations of these T-cell subtypes are identified and characterized. High salt induces Tregs to produce the pro-inflammatory cytokine interferon (IFN)-γ both in vivo and in vitro [49] and there is a population of Tregs isolated from plasma that secrete IL-17 and IFN-γ in patients with multiple sclerosis [50], diabetes [51] and non-small-cell lung cancer [52]. Additional studies suggest that, in humans, IL-17+ Tregs are generated in the periphery and contribute to antimicrobial defence, while also controlling autoimmunity and inflammation [53], however, how these cells affect cardiovascular health or BP control has not been examined. Moreover, the origin of these pro-inflammatory Tregs is in question. Th17 cells possess a high degree of plasticity [54], including the conversion of Th17 into Tregs [55,56]. Therefore, not only are there potentially pro-inflammatory Tregs, but also there are anti-inflammatory Th17 cells. Much more work needs to be done to define how these different subpopulations of Tregs and Th17 cells affect cardiovascular health in both sexes before a full understanding of their role in BP control can be achieved.
T-CELLS IN HUMAN HYPERTENSION
There is strong support for a role for T-cells in human hypertension. Immunosenescent CD8+ T-cells are increased in human hypertension [57] and treatment of patients with psoriasis and rheumatoid arthritis with the lymphocyte suppressant MMF exhibit a significant decrease in BP that is maintained during treatment [58]. Moreover, BP returns to pre-treatment levels following the cessation of treatment, further supporting a direct effect of lymphocytes on BP. Treatment of hypertensive patients with carotid atherosclerosis with traditional BP-lowering drugs also decreases circulating Th17 cells and increases Tregs, providing additional indirect support for an impact of BP on T-cells in humans [59]. In addition, acute psychological stress increases CD4+ T-cells and decrease Tregs in healthy men [60,61], and, although not assessed in the same study, acute mental stress also increases BP [62]. These findings raise the possibility that chronic stress contributes to T-cell-mediated hypertension in humans. In addition to changes in T-cell number, there is also evidence for alterations in T-cell function in human hypertension. T-cells from hypertensive patients have been reported to have either suppressed proliferative responses [63] or greater T-cell activity in vitro [57]. T-cells have also been implicated in pulmonary arterial hypertension, patients with pulmonary arterial hypertension have an increase in circulating Th17 cells [64]. Taken together, these data provide a strong rationale for additional studies on the role of T-cells in human hypertension.
SEX AND T-CELLS IN HYPERTENSION
Experimental hypertension
All of the experimental studies above were performed exclusively in males, calling into question the role of T-cells in BP control in females. Although none of these initial studies establishing a role for T-cells in experimental hypertension included females, the immune system has been linked to increases in BP in females (for example lupus, pre-eclampsia [65–67]). However, no studies had directly compared either normotensive or hypertensive males or females to elucidate the impact of sex on T-cell differentiation, activation or infiltration which could contribute to sex differences in hypertension.
To this end, studies by our laboratory examined the impact of MMF on BP in male and female spontaneously hypertensive rats (SHR). Similar to what is observed in male SHR, BP in female SHR decreased with MMF, supporting a pro-hypertensive role for lymphocytes in females [19]. We further examined the renal T-cell profile in male and female SHR; male SHR have more CD4+ cells and Th17 cells and females have more CD8+ cells and Tregs. Based on the proposed roles for Th17 cells compared with Tregs on BP, our findings are consistent with males having a higher BP compared with females. Additional studies confirmed that, consistent with studies in males, increases in BP are associated with increases in renal T-cells in females. Female SHR have significantly more T-cells in their kidneys than normotensive Wistar–Kyoto rats and increasing BP in female SHR increases renal T-cell counts [68].
To more directly link increases in BP to the T-cell profile in SHR, our group further treated male and female SHR with the BP-lowering agents hydrochlorothiazide and reserpine from either 6–12 weeks of age to prevent age-related increases in BP, or 11–13 weeks of age to reverse established hypertension. Neither treatment significantly altered renal CD3+, CD4+ and CD8+ T-cells or Th17 cells compared with same-sex vehicle controls. However, both attenuating age-related increases in BP and reversing established hypertension decreased renal Tregs only in female SHR abolishing the sex difference in Tregs [68]. The lack of a change in Th17 cells in either sex despite changes in BP calls into question the role of Th17 cells in BP control; however, our findings suggest a central role for Tregs in BP control especially in females. This conclusion is further supported by studies examining the impact of chronic Ang II infusion on renal T-cells in male and female Sprague–Dawley rats. Ang II infusion results in greater increases in CD4+ T-cells and Th17 cells in males, whereas Tregs increase only in females [69]. Consistent with our findings, female C57Bl/6 mice display an increase in Tregs in adipose tissue in response to a high-fat diet not observed in males, and females are protected from high-fat-diet-induced metabolic changes relative to males [70]. Based on the lower BP in multiple female experimental models of hypertension compared with males, including SHR and Ang II hypertension [8], we propose that the ability of females to increase Tregs in response to an increase in BP is a key compensatory mechanism to limit overall increases in BP. Likewise, the inability of males to increase BP may contribute to their higher overall BP and greater degree of end-organ damage seen in young males compared with age-matched females. More studies are needed to directly assess the role of Tregs on BP in females, and to determine the mechanisms by which Tregs are increased in females.
It is interesting to note that greater Tregs in female SHR as compared with male SHR are often mirrored by greater renal CD8+ T-cells as well [19,68]. Although there are both effector and regulatory CD8+ T-cells [71], to date there are no published studies that have further examined the CD8+ T-cell subtypes in hypertension. Regulatory CD8+ T-cells were the first identified subset of T-cells capable of inducing immune suppression [72]; however, difficulties in characterizing these cells have limited their study relative to Tregs. Based on recent studies described above indicating a key role for CD8+ T-cells in hypertension in males, we postulate that females may selectively up-regulate regulatory CD8+ T-cells as well as Tregs. Therefore, greater understanding of this novel T-cell subtype may provide new insight into role of T-cells in not only BP control, but overall cardiovascular health.
Three recent studies have greatly expanded our understanding of the impact of sex on T-cells and BP control using male and female Rag−/− mice. As has previously been demonstrated, adoptive transfer of CD3+ T-cells from wild-type male mice to male Rag−/− mice restores the hypertensive response to Ang II [20,73,74]. However, adoptive transfer of CD3+ T-cells from males did not increase BP responses to Ang II in female Rag−/− mice, and adoptive transfer of T-cells from female mice into male Rag−/− mice abrogated Ang II-induced increase in BP [73,74]. Additional studies further examined the role of CD4+ and CD8+ T-cells from males compared with females in mediating Ang II hypertension. Consistent with what was observed with total CD3+ T-cells, both CD4+ and CD8+ T-cells from male donors restored Ang II hypertension in male Rag−/− mice, although T-cells from females prevented Ang II-induced increases in BP in male Rag−/− mice [9]. Furthermore, since male Rag−/− mice receiving T-cells from a female donor exhibited a lower BP than if the donor T-cells were from a male, these studies clearly illustrate that both the sex of the T-cell and sex of the animal are key determinants of the responsiveness to T-cell-mediated increases in BP.
It is important to note that these sex differences in BP responses were not due to a greater number of T-cells in males following adoptive transfer of male T-cells. Indeed, male Rag−/− mice tended to have more CD3+, CD4+ and CD8+ T-cells both in the circulation and in perivascular fat following adoptive transfer of ‘female’ T-cells compared with ‘male’ T-cells, suggesting that the function of the T-cells is distinct in the recipient rat depending on the sex of the donor [74]. Consistent with this notion, male Rag−/− mice receiving CD3+ T-cells from male donors had more T-cells producing pro-inflammatory cytokines (tumour necrosis factor (TNF) and IL-17) following 2 weeks of Ang II infusion than if the T-cells were from a female, whereas plasma IL-10 levels were significantly higher if the donor T-cells were of female origin [74]. In addition, male Rag−/− mice exhibit greater increases in TNF, monocyte chemoattractant protein (MCP), and IL-2 than female Rag−/− mice following adoptive transfer of CD3+ T-cells from a male donor and Ang II infusion [73]. These data suggest that not only are females able to limit the pro-hypertensive effects of T-cells from males in response to Ang II hypertension, but also T-cells from females have a more anti-inflammatory, anti-hypertensive immune cell phenotype than T-cells from males, which is consistent with data from our group. It should be noted that Tregs were not greater in female Rag−/− mice following adoptive transfer of male T-cells or in male Rag−/− mice following adoptive transfer of female T-cells. This finding potentially highlights the importance of the entire immune profile in dictating the physiological outcome of alterations in the immune system within each sex. The key papers reporting a sex difference in T-cells in hypertension are summarized in Table 1.
Table 1.
Sex difference in T-cell involvement with BP control | Reference |
---|---|
T-cell population’s origin of sex determines the effect on BP in Rag1−/− mice | Sandberg et al. [9] |
Lymphocytes contribute to hypertension in SHR; the profile of circulating T-cells is sex-specific | Tipton et al. [19] |
BP contributes to a sex-specific renal T-cell profile in SHR; elevations in BP increase renal Tregs in females | Tipton et al. [68] |
Ang II infusion increases T-cells in SD rats; Treg levels increase only in females | Zimmerman et al. [69] |
Tregs are increased in C57 high-fat-diet-fed mice exclusively in females which protect them from metabolic changes | Pettersson et al. [70] |
Male CD3+ T-cells cause greater increases in BP and T-cell infiltration in male Rag1−/− mice than female | Pollow et al. [73] |
Sex of the T-cell itself is distinct in its function BP control in Rag1−/− mice | Ji et al. [74] |
Human hypertension
The majority of the studies discussed above supporting a role for T-cells in human hypertension have included both men and women, although the data have not been analysed separately based on sex, preventing sex-based conclusions from being drawn. However, sex differences have been reported in T-cells in human hypertension. Healthy women have higher levels of circulating CD4+ T-cells than men [75] and isolated CD4+ T-cells from women produce more IFN-γ and proliferate more than CD4+ T-cells from men, whereas CD4+ T-cells from men have greater IL-17 production [76]. Unfortunately, CD4+ T-cell subtypes were not assessed in these studies, therefore it is unknown whether differences in T-cell function are the result of greater Tregs in women or greater Th17 cells in men.
A sex-specific role for T-cells in human hypertension is also suggested by studies of autoimmune diseases, particularly systemic lupus erythaematosus (SLE). Women are more likely than men to develop SLE (9:1) and young women with SLE are approximately ten times more likely to develop hypertension than healthy young women [77,78]. Patients with SLE have an increase in circulating Th17 cells [79] and decreases in Tregs [80]. Moreover, pharmacological blockade of mammalian target of rapamycin (mTOR) using N-acetylcysteine (NAC) in patients with SLE (men and women) reported a decrease in total circulating CD4+ and CD8+ T-cells and increases in circulating Tregs and these alterations were accompanied by an improvement in patient health (BP was not reported) [81]. More women develop SLE than men, although disease severity is often worse in men; yet it is unknown whether men with SLE have greater T-cell activation. Due to the greater prevalence of SLE in women, the number of women represented in the studies above far outweighs the men and none of the data were presented separately for sex, leaving open the question of whether or not women with SLE maintain more Tregs than their male counterparts and whether this could affect the development and progression of hypertension. However, a study of 1979 lupus patients (157 men and 1822 women) reported that men are more likely than women to have disability, hypertension, thrombosis, and renal, haematological and serological manifestations of lupus [82]. Men were also more likely to exhibit end-organ damage including renal, cardiovascular, peripheral vascular disease and myocardial infarction – all of which are related to hypertension and linked to T-cell infiltration. Therefore, it is tempting to speculate that, consistent with experimental data in essential hypertension, females with SLE have enhanced Treg activation which attenuates the pro-inflammatory effector T-cells relative to males. Dysregulation in the balance of Tregs and Th17 cells has also been implicated in BP control in women with pre-eclampsia. Women with pre-eclampsia have increased total circulating CD4+ T-cells and Th17 cells and decreases in Tregs compared with women with healthy pregnancies [83–85]. Regardless, more clinical studies are needed to more fully define the T-cell phenotype and function in both healthy and hypertensive men and women to determine the likelihood of successfully targeting immune cells in a sex-dependent manner to increase BP control rates.
DOES SEX AFFECT T-CELL ACTIVATION IN HYPERTENSION?
With increasing support for the notion that T-cells play a critical role in BP control in hypertension, numerous studies have been initiated to understand how T-cells are activated in hypertension and identify the antigen(s) responsible. Current evidence suggests that T-cell activation occurs in response to increases in BP resulting in co-ordinated activation of innate and adaptive immune cells following the release and presentation of an endogenous antigen [6,7,86]. Although no studies to date have directly compared T-cell activation in hypertensive males and females, numerous steps along this pathway have been suggested to be differentially regulated in males compared with females. As a result, hypertensive stimuli may have sex-specific effects on T-cell activation and differentiation in hypertension.
Antigens: cell death and danger molecules
Vascular necrosis is a hallmark of malignant hypertension [87–89] and recent studies in male Dahl salt-sensitive rats indicate that renal injury in response to a high-salt diet, which is in part mediated by increases in T-cell infiltration, is dependent on increases in renal perfusion pressure and injury [90,91]. These studies suggest that elevated hydrostatic pressure in the kidney and vasculature in response to persistent hypertensive stimuli induces cellular necrosis. Necrosis is an uncontrolled pathological process characterized by loss of plasma membrane integrity resulting in the release of cellular material that is typically sequestered inside the cell. Matzinger [92] proposed the Danger Model of immunity in 1994 which suggested that the immune system is more concerned with damage than simply differentiating between ‘self’ and ‘non-self’. The Danger Model proposes that an immune response can be induced by either exogenous pathogen-associated molecular patterns (PAMPs) or endogenous damage-associated molecular patterns (DAMPs). As a result, cellular necrosis following injury results in the release of DAMPs into the extracellular space which are recognized by cells of the innate immune system leading to activation of the adaptive immune response. As a result, DAMPs may serve as antigens to induce T-cell activation in hypertension.
There is evidence in the literature to suggest that there are sex differences in the susceptibility of cells to undergo cell death. Isolated primary cortical neuron cultures from male rats more readily undergo autophagy and die, whereas neurons from females mobilize fatty acids, accumulate triacylglycerols, form lipid droplets and survive longer [93]. Similarly, cardiomyocytes from female mice exhibit greater survival in response to H2O2 (100 μmol/l) compared with cardiomyocytes from males [94]. These studies suggest an innate cellular difference in the cell death/cell survival machinery between the sexes, where cells from females are better able to tolerate cellular stress to resist death. In support of this notion, key proteins involved in cellular death are differentially expressed in males and females in a sex-hormone-dependent manner [95,96]. Testosterone increases Fas, Fas ligand (FasL) and Fas-associated death domain (FADD) expression to promote apoptosis in vascular endothelial cells and renal tubular cells [97,98]. In contrast, Akt, an anti-apoptotic protein that promotes cell survival, expression is greater in kidneys from females than males [99]. Moreover, testosterone inhibits Akt expression whereas oestrogen increases Akt [99–101], and oestrogen-mediated increases in Akt are associated with decreased apoptosis following coronary artery ligation in vivo [102]. These data suggest that sex steroids are critical in determining which cell death pathway is activated in damaged tissue resulting in sex differences seen in the extent of injury and hypertension. This could explain the basis for sex differences observed in T-cell profiles in experimental animals in key organs associated with the regulation of BP. Additional studies are needed to determine the impact of sex on cell death in vivo in hypertension; greater cellular necrosis in males in response to hypertensive stimuli may explain greater effector T-cell activation compared with females. Greater necrotic cell death in males would result in greater release of DAMPs, which would result in more pro-inflammatory stimuli.
DAMPs are ideal candidates to act as antigens for T-cell activation in hypertension. Although there is very little known regarding sex differences in DAMPs, DAMPs are associated with numerous diseases and levels have been shown to be increased in hypertensive humans and experimental animals [103,104]. DAMPs can either be ‘normal’ proteins that are not typically found in the extracellular space in healthy tissues, or proteins that have been damaged, misfolded or otherwise modified by the oxidative extracellular environment. DAMPs include, but are not limited to, asymmetric dimethylarginine, biglycan, C-phosphate-G DNA/mitochondrial DNA, C-reactive protein, DNA, RNA, fibrinogen, high mobility group box-1, heat-shock proteins, hyaluronan, uric acid, serum amyloid A protein, ATP, uric acid and IL-1α [104,105]. Recent studies have shown that proteins oxidatively modified by highly reactive γ -ketoaldehydes (isoketals) are formed in experimental hypertension and accumulate in DCs leading to T-cell-induced hypertension [25]. Although these studies only included male experimental animals, it is well established that females have lower levels of oxidative stress relative to males [8,106], therefore we would speculate that females would exhibit less T-cell activation compared with males. Isoketal adducts have also been measured in hypertensive men (two) and women (ten) [25]. Isoketals are greater in circulating monocytes and CD83+ cells (marker of human DCs) from humans with hypertension compared with healthy subjects. Moreover, the percentage of isoketal-positive cells was positively correlated with systolic BP, providing a potential link to human hypertension.
DAMPs activate T-cells via pattern recognition receptor (PRR) activation on cells of the innate immune system, including macrophages, leucocytes and DCs [107]. PRRs can be divided into four families including Toll-like receptors (TLRs), and TLRs have been linked to the development and progression of numerous cardiovascular disorders, including hypertension [103,104,108]. Although there are no studies to date that have directly compared the impact of TLR inhibition on BP in males and females, there is evidence to support sex differences in TLR expression and function. We have previously reported that female SHR have greater TLR3, TLR5 and TLR6 mRNA expression in the renal cortex and greater TLR4 and TLR6 expression in mesenteric arteries compared with males [109]. Consistent with our data, women in the Framingham Heart Study have higher platelet TLR mRNA expression than men, although TLR expression in men is more commonly associated with circulating cytokines, whereas in women, TLR expression was associated with greater soluble P-selectin levels [110]. In addition, hypertensive women with an D299G polymorphism of TLR4 have lower left ventricular (LV) mass and reduced LV hypertrophy compared with women with the wild-type genotype, although this polymorphism had no effect on LV structure in men, further supporting sex-specific effects of TLR on cardiovascular function [111].
T-cell receptor activation and antigen-presenting cells
Women typically exhibit a more robust immune response post-infection than men [112], raising the possibility that there are sex differences in the ability of T-cells to be activated following presentation of an antigen. This could reflect a sex difference in either the antigen-presenting cell (APC) or the T-cell. T-cell activation requires both T-cell receptor ligation (signal 1) and co-stimulation (signal 2). Signal 1 is the binding of antigen in the major histocompatibility complex (MHC) of APCs to the T-cell antigen receptor. Signal 2 is provided by interaction of the T-cell co-receptor CD28 with the APC B7 ligands, CD80 or CD86. Although there are no studies to date that have directly examined whether sex differencs in APCs or T-cell activation contribute to sex differences in BP, there is evidence to suggest this as a promising line of investigation since blocking B7-dependent co-stimulation of T-cells attenuates Ang II and DOCA-induced hypertension in male mice [24]. In adddition, a functional T-cell receptor is required for male Dahl salt-sensitive rats to fully develop hypertension in response to a high-salt diet [23]. Although the same studies have not been performed in female experimental animals, B7 blockade more effectively treats B16 melanoma in C57/BL6 female mice compared with males [113] which is consistent with our finding that female SHR have greater CD80 and CD86 mRNA expression in the renal cortex than male SHR [109]. These data could explain why females typically mount a more robust immune response than males; however, this would not necessarily explain sex differences in the T-cell subtype activation.
Alternatively, a sex difference in APCs could contribute to sex differences in both T-cell activation and differentiation. The most efficient professional APCs are DCs and DCs from hypertensive male mice have increased surface expression of the B7 ligands CD80 and CD86 [24]. The use of adoptive transfer studies have further established that donor DCs from Ang II-infused mice promote T-cell proliferation and activation resulting in a hypertensive response to low-dose Ang II in recipient mice [25,114]. Although no studies to date have examined DC phenotype and function in hypertensive males compared with females, female sex hormones promote the differentiation of functional DCs [115,116]. Oestradiol acts via oestrogen receptor α to either induce DC differentiation, leading to the development of DCs with increased functional capacity, or decrease the number of differentiated DCs [117]. Therefore, female sex hormones have the capacity to affect the DC phenotype and function and it is not unreasonable to speculate that this may result in sex differences in the differentiation and activation of distinct T-cell subtypes in females compared with males.
SUMMARY
There is an ever-expanding literature base supporting a causal role of T-cells in experimental hypertension. The challenge now is to fully understand the molecular mechanisms by which the immune system regulates BP in both sexes and how the different components of the immune system interact so that specific mechanisms can be targeted therapeutically without compromising natural immune defences. Although men and women both become hypertensive, many of the molecular pathways by which they become hypertensive differ and there is mounting evidence to suggest that the same will be the case for T-cells. Sex differences in BP control and cardiovascular disease have been known for over 60 years, and recent experimental studies in Rag−/− mice highlight the critical role of lymphocytes in mediating sex differences in basal BP control [73,74]. Therefore, based on the potential therapeutic application of Tregs, better understanding of how females are able to maintain higher numbers of Tregs will greatly aid in our ability to expand these T-cells in vivo to improve cardiovascular outcomes for all hypertensive patients.
FUNDING
This work was supported by the American Heart Association pre-doctoral fellowship (to G.R.C.) and [grant number 14GRNT20480199 (to J.C.S.)].
Abbreviations:
- Ang
angiotensin
- APC
antigen-presenting cell
- BP
blood pressure
- DAMP
damage-associated molecular pattern
- DC
dendritic cell
- DOCA
deoxycoticosterone acetate
- IFN
interferon
- IL
interleukin
- LV
left ventricular
- MMF
mycophenolate mofetil
- NAC
N-acetylcysteine
- NKT
natural killer T-cells
- PRR
pattern recognition receptor
- Rag
recombination-activating gene
- SHR
spontaneously hypertensive rats
- SLE
systemic lupus erythaematosus
- Th
T helper
- TLR
Toll-like receptor
- TNF
tumour necrosis factor
- Treg
regulatory T-cell
REFERENCES
- 1.Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ et al. (2015) Heart disease and stroke statistics-2015 update: a report from the American Heart Association. Circulation 131, e29–322 [DOI] [PubMed] [Google Scholar]
- 2.Cohen JD (2009) Hypertension epidemiology and economic burden: refining risk assessment to lower costs. Manag. Care 18, 51–58 [PubMed] [Google Scholar]
- 3.Harrison DG, Guzik TJ, Lob HE, Madhur MS, Marvar PJ, Thabet SR, Vinh A and Weyand CM (2011) Inflammation, immunity, and hypertension. Hypertension 57, 132–140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Harrison DG, Vinh A, Lob H and Madhur MS (2010) Role of the adaptive immune system in hypertension. Curr. Opin. Pharmacol 10, 203–207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Trott DW and Harrison DG (2014) The immune system in hypertension. Adv. Physiol. Educ 38, 20–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.McMaster WG, Kirabo A, Madhur MS and Harrison DG (2015) Inflammation, immunity, and hypertensive end-organ damage. Circ. Res 116, 1022–1033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang J and Crowley SD (2015) Role of T lymphocytes in hypertension. Curr. Opin. Pharmacol 21, 14–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zimmerman MA and Sullivan JC (2013) Hypertension: what’s sex got to do with it? Physiology 28, 234–244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sandberg K, Ji H and Hay M (2015) Sex-specific immune modulation of primary hypertension. Cell. Immunol 294, 95–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cheng S, Xanthakis V, Sullivan LM and Vasan RS (2012) Blood pressure tracking over the adult life course: patterns and correlates in the Framingham Heart Study. Hypertension 60, 1393–1399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hermida RC, Ayala DE, Mojon A, Fontao MJ, Chayan L and Fernandez JR (2013) Differences between men and women in ambulatory blood pressure thresholds for diagnosis of hypertension based on cardiovascular outcomes. Chronobiol. Int 30, 221–232 [DOI] [PubMed] [Google Scholar]
- 12.Boggia J, Thijs L, Hansen TW, Li Y, Kikuya M, Bjorklund K, Richart T, Ohkubo T, Jeppesen J, Torp-Pedersen C et al. (2011) Ambulatory blood pressure monitoring in 9357 subjects from 11 populations highlights missed opportunities for cardiovascular prevention in women. Hypertension 57, 397–405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wright JT Jr, Williamson JD, Whelton PK, Snyder JK, Sink KM, Rocco MV, Reboussin DM, Rahman M, Oparil S, Lewis CE et al. (2015) A randomized trial of intensive versus standard blood-pressure control. N. Engl. J. Med 373, 2103–2116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.James PA, Oparil S, Carter BL, Cushman WC, Dennison-Himmelfarb C, Handler J, Lackland DT, LeFevre ML, MacKenzie TD, Ogedegbe O et al. (2014) 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the eighth joint national committee (JNC 8). JAMA 311, 507–520 [DOI] [PubMed] [Google Scholar]
- 15.Gu Q, Burt VL, Paulose-Ram R and Dillon CF (2008) Gender differences in hypertension treatment, drug utilization patterns, and blood pressure control among us adults with hypertension: data from the National Health and Nutrition Examination survey. Am. J. Hypertens 21, 789–798 [DOI] [PubMed] [Google Scholar]
- 16.Rodriguez-Iturbe B, Quiroz Y, Nava M, Bonet L, Chavez M, Herrera-Acosta J, Johnson RJ and Pons HA (2002) Reduction of renal immune cell infiltration results in blood pressure control in genetically hypertensive rats. Am. J. Physiol. Renal. Physiol 282, F191–F201 [DOI] [PubMed] [Google Scholar]
- 17.De Miguel C, Das S, Lund H and Mattson DL (2010) T lymphocytes mediate hypertension and kidney damage in dahl salt-sensitive rats. Am. J. Physiol. Regul. Integr Comp. Physiol 298, R1136–R1142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mattson DL, James L, Berdan EA and Meister CJ (2006) Immune suppression attenuates hypertension and renal disease in the Dahl salt-sensitive rat. Hypertension 48, 149–156 [DOI] [PubMed] [Google Scholar]
- 19.Tipton AJ, Baban B and Sullivan JC (2012) Female spontaneously hypertensive rats have greater renal anti-inflammatory T lymphocyte infiltration than males. Am. J. Physiol. Regul. Integr. Comp. Physiol 303, R359–R367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C and Harrison DG (2007) Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J. Exp. Med 204, 2449–2460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Crowley SD, Song YS, Lin EE, Griffiths R, Kim HS and Ruiz P (2010) Lymphocyte responses exacerbate angiotensin II-dependent hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol 298, R1089–R1097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mattson DL, Lund H, Guo C, Rudemiller N, Geurts AM and Jacob H (2013) Genetic mutation of recombination activating gene 1 in Dahl salt-sensitive rats attenuates hypertension and renal damage. Am. J. Physiol. Regul. Integr. Comp. Physiol 304, R407–R414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rudemiller N, Lund H, Jacob HJ, Geurts AM and Mattson DL (2014) Cd247 modulates blood pressure by altering T-lymphocyte infiltration in the kidney. Hypertension 63, 559–564 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Vinh A, Chen W, Blinder Y, Weiss D, Taylor WR, Goronzy JJ, Weyand CM, Harrison DG and Guzik TJ (2010) Inhibition and genetic ablation of the B7/CD28 T-cell costimulation axis prevents experimental hypertension. Circulation 122, 2529–2537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kirabo A, Fontana V, de Faria AP, Loperena R, Galindo CL, Wu J, Bikineyeva AT, Dikalov S, Xiao L, Chen W et al. (2014) DC isoketal-modified proteins activate T cells and promote hypertension. J. Clin. Invest 124, 4642–4656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ma F, Feng J, Zhang C, Li Y, Qi G, Li H, Wu Y, Fu Y, Zhao Y, Chen H et al. (2014) The requirement of CD8 T cells to initiate and augment acute cardiac inflammatory+ response to high blood pressure. J. Immunol 192, 3365–3373 [DOI] [PubMed] [Google Scholar]
- 27.Trott DW, Thabet SR, Kirabo A, Saleh MA, Itani H, Norlander AE, Wu J, Goldstein A, Arendshorst WJ, Madhur MS et al. (2014) Oligoclonal CD8+ T cells play a critical role in the development of hypertension. Hypertension 64, 1108–1115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Dong C (2008) Th17 cells in development: an updated view of their molecular identity and genetic programming. Nat. Rev. Immunol 8, 337–348 [DOI] [PubMed] [Google Scholar]
- 29.Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM and Weaver CT (2005) Interleukin 17-producing CD4+ effector Tt cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat. Immunol 6, 1123–1132 [DOI] [PubMed] [Google Scholar]
- 30.Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ and Littman DR (2006) The orphan nuclear receptor rorgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121–1133 [DOI] [PubMed] [Google Scholar]
- 31.Raphael I, Nalawade S, Eagar TN and Forsthuber TG (2015) T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 74, 5–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhang Y, Gu W and Sun B (2014) Th1/Th2 cell differentiation and molecular signals. Adv. Exp. Med. Biol 841, 15–44 [DOI] [PubMed] [Google Scholar]
- 33.Korn T, Oukka M, Kuchroo V and Bettelli E (2007) Th17 cells: effector T cells with inflammatory properties. Semin. Immunol 19, 362–371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Maddur MS, Miossec P, Kaveri SV and Bayry J (2012) Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am. J. Pathol 181, 8–18 [DOI] [PubMed] [Google Scholar]
- 35.Kimura A and Kishimoto T (2011) Th17 cells in inflammation. Int. Immunopharmacol 11, 319–322 [DOI] [PubMed] [Google Scholar]
- 36.Amador CA, Barrientos V, Pena J, Herrada AA, Gonzalez M, Valdes S, Carrasco L, Alzamora R, Figueroa F, Kalergis AM and Michea L (2014) Spironolactone decreases doca-salt-induced organ damage by blocking the activation of T helper 17 and the downregulation of regulatory T lymphocytes. Hypertension 63, 797–803 [DOI] [PubMed] [Google Scholar]
- 37.Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y, Guzik TJ and Harrison DG (2010) Interleukin 17 promotes angiotensin II-induced hypertension and vascular dysfunction. Hypertension 55, 500–507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Krebs CF, Lange S, Niemann G, Rosendahl A, Lehners A, Meyer-Schwesinger C, Stahl RA, Benndorf RA, Velden J, Paust HJ et al. (2014) Deficiency of the interleukin 17/23 axis accelerates renal injury in mice with deoxycorticosterone acetate+angiotensin II-induced hypertension. Hypertension 63, 565–571 [DOI] [PubMed] [Google Scholar]
- 39.Vignali DA, Collison LW and Workman CJ (2008) How regulatory T cells work. Nat. Rev. Immunol 8, 523–532 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Campbell DJ (2015) Control of regulatory T cell migration, function, and homeostasis. J. Immunol 195, 2507–2513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Viel EC, Lemarie CA, Benkirane K, Paradis P and Schiffrin EL (2010) Immune regulation and vascular inflammation in genetic hypertension. Am. J. Physiol. Heart Circ. Physiol 298, H938–H944 [DOI] [PubMed] [Google Scholar]
- 42.Kvakan H, Kleinewietfeld M, Qadri F, Park JK, Fischer R, Schwarz I, Rahn HP, Plehm R, Wellner M, Elitok S et al. (2009) Regulatory T cells ameliorate angiotensin II-induced cardiac damage. Circulation 119, 2904–2912 [DOI] [PubMed] [Google Scholar]
- 43.Kanellakis P, Dinh TN, Agrotis A and Bobik A (2011) CD4(+)CD25(+)Foxp3(+) regulatory T cells suppress cardiac fibrosis in the hypertensive heart. J. Hypertens 29, 1820–1828 [DOI] [PubMed] [Google Scholar]
- 44.Kassan M, Galan M, Partyka M, Trebak M and Matrougui K (2011) Interleukin-10 released by CD4(+)CD25(+) natural regulatory T cells improves microvascular endothelial function through inhibition of NADPH oxidase activity in hypertensive mice. Arterioscler. Thromb. Vasc. Biol 31, 2534–2542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Matrougui K, Abd Elmageed Z, Kassan M, Choi S, Nair D, Gonzalez-Villalobos RA, Chentoufi AA, Kadowitz P, Belmadani S and Partyka M (2011) Natural regulatory T cells control coronary arteriolar endothelial dysfunction in hypertensive mice. Am. J. Pathol 178, 434–441 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Barhoumi T, Kasal DA, Li MW, Shbat L, Laurant P, Neves MF, Paradis P and Schiffrin EL (2011) T regulatory lymphocytes prevent angiotensin II-induced hypertension and vascular injury. Hypertension 57, 469–476 [DOI] [PubMed] [Google Scholar]
- 47.Kasal DA, Barhoumi T, Li MW, Yamamoto N, Zdanovich E, Rehman A, Neves MF, Laurant P, Paradis P and Schiffrin EL (2012) T regulatory lymphocytes prevent aldosterone-induced vascular injury. Hypertension 59, 324–330 [DOI] [PubMed] [Google Scholar]
- 48.Tamosiuniene R, Tian W, Dhillon G, Wang L, Sung YK, Gera L, Patterson AJ, Agrawal R, Rabinovitch M, Ambler K et al. (2011) Regulatory T cells limit vascular endothelial injury and prevent pulmonary hypertension. Circ. Res 109, 867–879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hernandez AL, Kitz A, Wu C, Lowther DE, Rodriguez DM, Vudattu N, Deng S, Herold KC, Kuchroo VK, Kleinewietfeld M and Hafler DA (2015) Sodium chloride inhibits the suppressive function of Foxp3 regulatory T cells. J. Clin. Invest 125, 4212–4222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Baecher-Allan CM, Costantino CM, Cvetanovich GL, Ashley CW, Beriou G, Dominguez-Villar M and Hafler DA (2011) CD2 costimulation reveals defective activity by human CD4+CD25(hi) regulatory cells in patients with multiple sclerosis. J. Immunol 186, 3317–3326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.McClymont SA, Putnam AL, Lee MR, Esensten JH, Liu W, Hulme MA, Hoffmuller U, Baron U, Olek S, Bluestone JA and Brusko TM (2011) Plasticity of human regulatory T cells in healthy subjects and patients with type 1 diabetes. J. Immunol 186, 3918–3926 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Phillips JD, Knab LM, Blatner NR, Haghi L, DeCamp MM, Meyerson SL, Heiferman MJ, Heiferman JR, Gounari F, Bentrem DJ and Khazaie K (2015) Preferential expansion of pro-inflammatory tregs in human non-small cell lung cancer. Cancer Immunol. Immunother 64, 1185–1191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Voo KS, Wang YH, Santori FR, Boggiano C, Arima K, Bover L, Hanabuchi S, Khalili J, Marinova E, Zheng B, Littman DR and Liu YJ (2009) Identification of IL-17-producing Foxp3+ regulatory T cells in humans. Proc. Natl. Acad. Sci. U.S.A 106, 4793–4798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Guery L and Hugues S (2015) Th17 cell plasticity and functions in cancer immunity. Biomed. Res. Int 2015, 314620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gagliani N, Vesely MC, Iseppon A, Brockmann L, Xu H, Palm NW, de Zoete MR, Licona-Limon P, Paiva RS, Ching T et al. (2015) Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 523, 221–225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Obermajer N, Popp FC, Soeder Y, Haarer J, Geissler EK, Schlitt HJ and Dahlke MH (2014) Conversion of Th17 into IL-17a(neg) regulatory T cells: a novel mechanism in prolonged allograft survival promoted by mesenchymal stem cell-supported minimized immunosuppressive therapy. J. Immunol 193, 4988–4999 [DOI] [PubMed] [Google Scholar]
- 57.Youn JC, Yu HT, Lim BJ, Koh MJ, Lee J, Chang DY, Choi YS, Lee SH, Kang SM, Jang Y et al. (2013) Immunosenescent CD8+ T cells and c-x-c chemokine receptor type 3 chemokines are increased in human hypertension. Hypertension 62, 126–133 [DOI] [PubMed] [Google Scholar]
- 58.Herrera J, Ferrebuz A, MacGregor EG and Rodriguez-Iturbe B (2006) Mycophenolate mofetil treatment improves hypertension in patients with psoriasis and rheumatoid arthritis. J. Am. Soc. Nephrol 17, S218–S225 [DOI] [PubMed] [Google Scholar]
- 59.Liu Z, Zhao Y, Wei F, Ye L, Lu F, Zhang H, Diao Y, Song H and Qi Z (2014) Treatment with telmisartan/rosuvastatin combination has a beneficial synergistic effect on ameliorating Th17/Treg functional imbalance in hypertensive patients with carotid atherosclerosis. Atherosclerosis 233, 291–299 [DOI] [PubMed] [Google Scholar]
- 60.Freier E, Weber CS, Nowottne U, Horn C, Bartels K, Meyer S, Hildebrandt Y, Luetkens T, Cao Y, Pabst C et al. (2010) Decrease of CD4(+)Foxp3(+) T regulatory cells in the peripheral blood of human subjects undergoing a mental stressor. Psychoneuroendocrinology 35, 663–673 [DOI] [PubMed] [Google Scholar]
- 61.Atanackovic D, Schnee B, Schuch G, Faltz C, Schulze J, Weber CS, Schafhausen P, Bartels K, Bokemeyer C, Brunner-Weinzierl MC and Deter HC (2006) Acute psychological stress alerts the adaptive immune response: stress-induced mobilization of effector t cells. J. Neuroimmunol 176, 141–152 [DOI] [PubMed] [Google Scholar]
- 62.Treiber FA, Jackson RW, Davis H, Pollock JS, Kapuku G, Mensah GA and Pollock DM (2000) Racial differences in endothelin-1 at rest and in response to acute stress in adolescent males. Hypertension 35, 722–725 [DOI] [PubMed] [Google Scholar]
- 63.Sonmez A, Kisa U, Uckaya G, Eyileten T, Comert B, Koc B, Kocabalkan F and Ozata M (2001) Effects of losartan treatment on T-cell activities and plasma leptin concentrations in primary hypertension. J. Renin Angiotensin. Aldosterone Syst 2, 112–116 [DOI] [PubMed] [Google Scholar]
- 64.Hautefort A, Girerd B, Montani D, Cohen-Kaminsky S, Price L, Lambrecht BN, Humbert M and Perros F (2015) T-helper 17 cell polarization in pulmonary arterial hypertension. Chest 147, 1610–1620 [DOI] [PubMed] [Google Scholar]
- 65.Perez-Sepulveda A, Torres MJ, Khoury M and Illanes SE (2014) Innate immune system and preeclampsia. Front. Immunol 5, 244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Laresgoiti-Servitje E (2013) A leading role for the immune system in the pathophysiology of preeclampsia. J. Leukoc. Biol 94, 247–257 [DOI] [PubMed] [Google Scholar]
- 67.Cornelius DC, Hogg JP, Scott J, Wallace K, Herse F, Moseley J, Wallukat G, Dechend R and LaMarca B (2013) Administration of interleukin-17 soluble receptor c suppresses Th17 cells, oxidative stress, and hypertension in response to placental ischemia during pregnancy. Hypertension 62, 1068–1073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tipton AJ, Baban B and Sullivan JC (2014) Female spontaneously hypertensive rats have a compensatory increase in renal regulatory T cells in response to elevations in blood pressure. Hypertension 64, 557–564 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zimmerman MA, Baban B, Tipton AJ, O’Connor PM and Sullivan JC (2014) Chronic ang II infusion induces sex-specific increases in renal T cells in Sprague Dawley rats. Am. J. Physiol. Renal. Physiol 308, F706–F712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Pettersson US, Walden TB, Carlsson PO, Jansson L and Phillipson M (2012) Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. PLoS One 7, e46057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wang YM and Alexander SI (2009) CD8 regulatory T cells: what’s old is now new. Immunol. Cell Biol 87, 192–193 [DOI] [PubMed] [Google Scholar]
- 72.Cantor H, Shen FW and Boyse EA (1976) Separation of helper T cells from suppressor T cells expressing different ly components. Ii. Activation by antigen: After immunization, antigen-specific suppressor and helper activities are mediated by distinct T-cell subclasses. J. Exp. Med 143, 1391–1340 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pollow DP, Uhrlaub J, Romero-Aleshire MJ, Sandberg K, Nikolich-Zugich J, Brooks HL and Hay M (2014) Sex differences in T-lymphocyte tissue infiltration and development of angiotensin II hypertension. Hypertension 64, 384–390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ji H, Zheng W, Li X, Liu J, Wu X, Zhang MA, Umans JG, Hay M, Speth RC, Dunn SE and Sandberg K (2014) Sex-specific T-cell regulation of angiotensin II-dependent hypertension. Hypertension 64, 573–582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Amadori A, Zamarchi R, De Silvestro G, Forza G, Cavatton G, Danieli GA, Clementi M and Chieco-Bianchi L (1995) Genetic control of the CD4/CD8 T-cell ratio in humans. Nat. Med 1, 1279–1283 [DOI] [PubMed] [Google Scholar]
- 76.Zhang MA, Rego D, Moshkova M, Kebir H, Chruscinski A, Nguyen H, Akkermann R, Stanczyk FZ, Prat A, Steinman L and Dunn SE (2012) Peroxisome proliferator-activated receptor (PPAR)alpha and -gamma regulate IFNgamma and IL-17a production by human T cells in a sex-specific way. Proc. Natl. Acad. Sci. U.S.A 109, 9505–9510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Tedeschi SK, Bermas B and Costenbader KH (2013) Sexual disparities in the incidence and course of SLE and RA. Clin. Immunol 149, 211–218 [DOI] [PubMed] [Google Scholar]
- 78.Ryan MJ (2009) The pathophysiology of hypertension in systemic lupus erythematosus. Am. J. Physiol. Regul. Integr. Comp. Physiol 296, R1258–R1267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yang J, Chu Y, Yang X, Gao D, Zhu L, Wan L and Li M (2009) Th17 and natural Treg cell population dynamics in systemic lupus erythematosus. Arthritis Rheum. 60, 1472–1483 [DOI] [PubMed] [Google Scholar]
- 80.Lee HY, Hong YK, Yun HJ, Kim YM, Kim JR and Yoo WH (2008) Altered frequency and migration capacity of CD4+CD25+ regulatory T cells in systemic lupus erythematosus. Rheumatology 47, 789–794 [DOI] [PubMed] [Google Scholar]
- 81.Lai ZW, Hanczko R, Bonilla E, Caza TN, Clair B, Bartos A, Miklossy G, Jimah J, Doherty E, Tily H et al. (2012) N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 64, 2937–2946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tan TC, Fang H, Magder LS and Petri MA (2012) Differences between male and female systemic lupus erythematosus in a multiethnic population. J. Rheumatol 39, 759–769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wallace K, Cornelius DC, Scott J, Heath J, Moseley J, Chatman K and LaMarca B (2014) CD4+ T cells are important mediators of oxidative stress that cause hypertension in response to placental ischemia. Hypertension 64, 1151–1158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Cao X, Wang LL and Luo X (2013) Expression of regulatory T and helper T cells in peripheral blood of patients with pregnancy-induced hypertension. Clin. Exp. Obstet. Gynecol 40, 502–504 [PubMed] [Google Scholar]
- 85.Cornelius DC and Lamarca B (2014) Th17- and IL-17-mediated autoantibodies and placental oxidative stress play a role in the pathophysiology of pre-eclampsia. Minerva Ginecol. 66, 243–249 [PMC free article] [PubMed] [Google Scholar]
- 86.Marvar PJ, Thabet SR, Guzik TJ, Lob HE, McCann LA, Weyand C, Gordon FJ and Harrison DG (2010) Central and peripheral mechanisms of T-lymphocyte activation and vascular inflammation produced by angiotensin II-induced hypertension. Circ. Res 107, 263–270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gustafsson F (1997) Hypertensive arteriolar necrosis revisited. Blood Press. 6, 71–77 [DOI] [PubMed] [Google Scholar]
- 88.Gardner DL (1963) Arteriolar necrosis and the prenecrotic phase of experimental hypertension. Q. J. Exp. Physiol. Cogn. Med. Sci 48, 156–163 [DOI] [PubMed] [Google Scholar]
- 89.Gardner DL and Brooks PW (1963) Arteriolar necrosis in adrenal-regeneration hypertension: influence of preventive treatment with hydralazine on tissue electrolvtes. Br. J. Exp. Pathol 44, 31–37 [PMC free article] [PubMed] [Google Scholar]
- 90.Mattson DL (2014) Infiltrating immune cells in the kidney in salt-sensitive hypertension and renal injury. Am. J. Physiol. Renal. Physiol 307, F499–F508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Mori T, Polichnowski A, Glocka P, Kaldunski M, Ohsaki Y, Liang M and Cowley AW Jr (2008) High perfusion pressure accelerates renal injury in salt-sensitive hypertension. J. Am. Soc. Nephrol 19, 1472–1482 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Matzinger P (2002) The danger model: a renewed sense of self. Science 296, 301–305 [DOI] [PubMed] [Google Scholar]
- 93.Du L, Hickey RW, Bayir H, Watkins SC, Tyurin VA, Guo F, Kochanek PM, Jenkins LW, Ren J, Gibson G et al. (2009) Starving neurons show sex difference in autophagy. J. Biol. Chem 284, 2383–2396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wang F, He Q, Sun Y, Dai X and Yang XP (2010) Female adult mouse cardiomyocytes are protected against oxidative stress. Hypertension 55, 1172–1178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Metcalfe PD and Meldrum KK (2006) Sex differences and the role of sex steroids in renal injury. J. Urol 176, 15–21 [DOI] [PubMed] [Google Scholar]
- 96.Kher A, Meldrum KK, Wang M, Tsai BM, Pitcher JM and Meldrum DR (2005) Cellular and molecular mechanisms of sex differences in renal ischemia-reperfusion injury. Cardiovasc. Res 67, 594–603 [DOI] [PubMed] [Google Scholar]
- 97.Ling S, Dai A, Williams MR, Myles K, Dilley RJ, Komesaroff PA and Sudhir K (2002) Testosterone (t) enhances apoptosis-related damage in human vascular endothelial cells. Endocrinology 143, 1119–1125 [DOI] [PubMed] [Google Scholar]
- 98.Verzola D, Gandolfo MT, Salvatore F, Villaggio B, Gianiorio F, Traverso P, Deferrari G and Garibotto G (2004) Testosterone promotes apoptotic damage in human renal tubular cells. Kidney Int 65, 1252–1261 [DOI] [PubMed] [Google Scholar]
- 99.Park KM, Kim JI, Ahn Y, Bonventre AJ and Bonventre JV (2004) Testosterone is responsible for enhanced susceptibility of males to ischemic renal injury. J. Biol. Chem 279, 52282–52292 [DOI] [PubMed] [Google Scholar]
- 100.Camper-Kirby D, Welch S, Walker A, Shiraishi I, Setchell KD, Schaefer E, Kajstura J, Anversa P and Sussman MA (2001) Myocardial Akt activation and gender: Increased nuclear activity in females versus males. Circ. Res 88, 1020–1027 [DOI] [PubMed] [Google Scholar]
- 101.Patten RD, Pourati I, Aronovitz MJ, Baur J, Celestin F, Chen X, Michael A, Haq S, Nuedling S, Grohe C et al. (2004) 17beta-estradiol reduces cardiomyocyte apoptosis in vivo and in vitro via activation of phospho-inositide-3 kinase/Akt signaling. Circ. Res 95, 692–699 [DOI] [PubMed] [Google Scholar]
- 102.Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW and Liao JK (2000) Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-oh kinase. Nature 407, 538–541 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mian MO, Paradis P and Schiffrin EL (2014) Innate immunity in hypertension. Curr. Hypertens. Rep 16, 413. [DOI] [PubMed] [Google Scholar]
- 104.McCarthy CG, Goulopoulou S, Wenceslau CF, Spitler K, Matsumoto T and Webb RC (2014) Toll-like receptors and damage-associated molecular patterns: novel links between inflammation and hypertension. Am. J. Physiol. Heart Circ. Physiol 306, H184–H196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Jounai N, Kobiyama K, Takeshita F and Ishii KJ (2012) Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination. Front. Cell. Infect. Microbiol 2, 168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Lopez-Ruiz A, Sartori-Valinotti J, Yanes LL, Iliescu R and Reckelhoff JF (2008) Sex differences in control of blood pressure: role of oxidative stress in hypertension in females. Am. J. Physiol. Heart Circ. Physiol 295, H466–H474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Takeuchi O and Akira S (2010) Pattern recognition receptors and inflammation. Cell 140, 805–820 [DOI] [PubMed] [Google Scholar]
- 108.Singh MV and Abboud FM (2014) Toll-like receptors and hypertension. Am. J. Physiol. Regul. Integr. Comp. Physiol 307, R501–R504 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Tipton AJ and Sullivan JC (2014) Sex differences in T cells in hypertension. Clin. Ther 36, 1882–1900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Koupenova M, Mick E, Mikhalev E, Benjamin EJ, Tanriverdi K and Freedman JE (2015) Sex differences in platelet toll-like receptors and their association with cardiovascular risk factors. Arterioscler. Thromb. Vasc. Biol 35, 1030–1037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sales ML, Schreiber R, Ferreira-Sae MC, Fernandes MN, Piveta C, Cipolli JA, Calixto A, Matos-Souza JR, Geloneze B, Franchini KG and Nadruz W Jr (2010) The functional toll-like receptor 4 Asp299Gly polymorphism is associated with lower left ventricular mass in hypertensive women. Clin. Chim. Acta 411, 744–748 [DOI] [PubMed] [Google Scholar]
- 112.Marriott I and Huet-Hudson YM (2006) Sexual dimorphism in innate immune responses to infectious organisms. Immunol. Res 34, 177–192 [DOI] [PubMed] [Google Scholar]
- 113.Lin PY, Sun L, Thibodeaux SR, Ludwig SM, Vadlamudi RK, Hurez VJ, Bahar R, Kious MJ, Livi CB, Wall SR et al. (2010) B7-h1-dependent sex-related differences in tumor immunity and immunotherapy responses. J. Immunol 185, 2747–2753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Xiao L, Kirabo A, Wu J, Saleh MA, Zhu L, Wang F, Takahashi T, Loperena R, Foss JD, Mernaugh RL et al. (2015) Renal denervation prevents immune cell activation and renal inflammation in angiotensin II-induced hypertension. Circ. Res 117, 547–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Paharkova-Vatchkova V, Maldonado R and Kovats S (2004) Estrogen preferentially promotes the differentiation of CD11c+CD11b (intermediate) dendritic cells from bone marrow precursors. J. Immunol 172, 1426–1436 [DOI] [PubMed] [Google Scholar]
- 116.Douin-Echinard V, Laffont S, Seillet C, Delpy L, Krust A, Chambon P, Gourdy P, Arnal JF and Guery JC (2008) Estrogen receptor alpha, but not beta, is required for optimal dendritic cell differentiation and [corrected] CD40-induced cytokine production. J. Immunol 180, 3661–3669 [DOI] [PubMed] [Google Scholar]
- 117.Kovats S (2012) Estrogen receptors regulate an inflammatory pathway of dendritic cell differentiation: mechanisms and implications for immunity. Horm. Behav 62, 254–262 [DOI] [PMC free article] [PubMed] [Google Scholar]