Sei sulla pagina 1di 9

Pflugers Arch - Eur J Physiol (2015) 467:543–550

DOI 10.1007/s00424-014-1659-z

INVITED REVIEW

Sodium chloride, SGK1, and Th17 activation


Katrina J. Binger & Ralf A. Linker & Dominik N. Muller &
Markus Kleinewietfeld

Received: 16 October 2014 / Revised: 19 November 2014 / Accepted: 20 November 2014 / Published online: 4 December 2014
# Springer-Verlag Berlin Heidelberg 2014

Abstract The incidence of autoimmune diseases in Western (T1D) in Western societies are increasing rapidly, but the
civilizations is increasing rapidly, suggesting an influence of underlying cause for this has not been elucidated [4, 74].
environmental factors, such as diet. The pathogenesis of sev- MS and psoriasis are both relapsing and remitting autoim-
eral of these autoimmune diseases is characterized by aberrant mune diseases, accompanied by increased hypertension and
activation of T helper 17 (Th17) cells. Recent reports have cardiovascular risk. Genome-wide association studies have
shown that the differentiation of Th17 cells is sensitive to demonstrated that genetic factors predispose to the develop-
changes in local microenvironments, in particular salt (NaCl) ment of autoimmune diseases [48, 52, 64]. Environmental
concentrations, in a molecular mechanism centered around the factors may also be important triggers for these diseases, as
serum- and glucocorticoid-inducible kinase 1 (SGK1). In this there is a relatively low concordance rate between monozy-
review, we summarize the recently disclosed mechanisms by gotic twins for most of these diseases [8]. The “Western diet,”
which salt has been shown to affect SGK1 and, subsequently, high in saturated fatty acids and salt, has long been postulated
Th17 activation. as one potential environmental cause for the increasing inci-
dence of autoimmune diseases in developed countries where
Keywords Salt . SGK1 . T cells . Autoimmunity processed and fast food consumption is more common.

Introduction Differentiation of Th17 cells

Incidence and prevalence of human autoimmune diseases Helper T lymphocytes play an essential role in combatting
such as multiple sclerosis (MS), psoriasis, and type 1 diabetes infection. During active inflammation, naïve T cells differen-
tiate into various T helper cell types (e.g. Th1, Th2, Th17),
K. J. Binger : D. N. Muller
depending on the local cytokine milieu. Th1 cells typically
Experimental and Clinical Research Center, an institutional produce interferon (IFN)-γ and tumor necrosis factor-α, while
cooperation between the Charité Medical Faculty and the Th2 cells produce interleukin (IL)-4 and 13, and Th17 cells
Max-Delbrueck Center for Molecular Medicine, Berlin 13125, release IL-17, IL-21, and IL-22; cytokines which are pivotal in
Germany
the defense against bacteria and fungi. Besides Th17 cells, IL-
R. A. Linker 17 is also produced by γδ T cells, natural killer cells, and
Friedrich-Alexander-University of Erlangen-Nuremberg, natural killer T cells [60]. The differentiation of inflammatory
Erlangen 91054, Germany Th17 cells, required for host defense and autoimmunity, can
occur with a number of different cues. These include stimula-
D. N. Muller (*)
Max-Delbrueck Center for Molecular Medicine, Berlin 13125, tion with TGF-β3 in combination with IL-23 [39], IL1β, and
Germany IL-6 or IL-23 [27], where, in both cases, IL-17A single-
e-mail: dominik.mueller@mdc-berlin.de positive cells are converted into IL-17A/interferon (IFN)-γ
double-positive cells. Additionally, IL-1β has been shown to
M. Kleinewietfeld
Translational Immunology, Medical Faculty Carl Gustav Carus, TU induce the differentiation of inflammatory Th17 cells by
Dresden, 01307 Dresden, Germany repressing IL-10 production (a cytokine with potent anti-
544 Pflugers Arch - Eur J Physiol (2015) 467:543–550

inflammatory and regulatory properties) in human Th17 cells Role of Th17 cells in atherosclerosis and hypertension
[78]. Although it was initially thought that Th17 cells are
exclusively inflammatory, there has been recent accumulating As mentioned above, the differentiation of inflammatory
evidence of a ‘regulatory’ IL-17 producing Th17 subset. In Th17 cells greatly depends on the cytokine milieu and local
this setting, it was demonstrated that with combined TGF-β1 microenvironment [reviewed by 61], which may be particu-
and IL-6 signaling, or by dedicated mechanisms in the small larly relevant to cardiovascular disease. Atherosclerosis, a
intestine in vivo, subsets of Th17 cells are generated which chronic inflammatory arterial disease dependent on both in-
produce high levels of IL-10 [23, 44]. Taken together, the nate and adaptive immunity, is most likely the best-studied
inflammatory potential of Th17 cells seems to be determined cardiovascular disease with regard to the immune system.
by the cytokine milieu and local environment by which these However, research on the contribution of Th17 cells in pro-
cells face. moting the pathogenesis of atherosclerosis is conflicting [61].
IL-17 has been shown to directly promote the production of
pro-atherogenic cytokines from macrophages [15]. However,
Autoimmune diseases and Th17 cells studies to block IL-17 have not been promising [reviewed by
[61]. It may be that some of this confusion is due to the
Th17 cells are prominent in peripheral tissues, in particular the influence of confounding factors, such as effects from the
gut, where the production of IL-17 is important for the re- method used to block IL-17 and dietary composition or feed-
cruitment of other immune cells and the production of other ing duration directly regulating the immune response. It has
factors to promote pathogen clearance [reviewed by 34]. also been shown that reducing Th17 can promote the number
However, Th17 cells are also linked to the development of of Th1 cells. These are important considerations as atheroscle-
several autoimmune diseases, and, in this context, their aber- rotic plaque size correlates with the Th1 cytokine IFN-γ, and
rant activation can be considered pathogenic. The develop- thus, enhancement of Th1 responses is associated with in-
ment of autoimmune diseases such as multiple sclerosis, pso- creased lesion size. Conversely, a reduction in IFN-γ produc-
riasis, type I diabetes, primary Sjögren’s syndrome, asthma, ing cells correlates with a reduction in plaque size [12, 67].
and rheumatoid arthritis are all shown to involve Th17-driven The role of IL-17 in hypertension is similarly controversial.
inflammation [reviewed by 1, 40, 57]. Madhur et al. showed that angiotensin II infusion increases IL-
What causes Th17 cells to become pathogenic and drive 17 production from T cells and IL-17 protein in the aortic
these diseases is unclear. Generally, the formation of Th17 media layer. However, the initial hypertensive response to
cells is dependent on TGFβ signaling in combination with IL- angiotensin II infusion was similar with IL-17 knockout and
6 or other pro-inflammatory cytokines. TGFβ is also essential C57BL/6 J wild-type control mice, while 4 weeks after infu-
for the formation of regulatory T cells (Treg), in this case with sion, hypertension was not sustained in IL-17 knockout mice.
co-stimulation with IL-2 or IL-10 [41]. Tregs are generally Vessels from IL-17 knockout mice displayed a preserved
considered to prevent autoimmunity, and thus, it is striking vascular function, decreased superoxide production, and re-
that with the same primary stimulus (TGFβ), Th17 and Treg duced T cell infiltration in response to angiotensin II [42].
cells adopt such different fates. Understanding the conditions While a reduction in blood pressure only during late phase
by which Th17 cells are enhanced and if these can be con- points towards Th17 cells having an indirect effect in the
verted or dedifferentiated into Tregs are the focus of much pathogenesis of hypertension, a recent study of Amador
recent work. Retinoic acid (RA; a vitamin A metabolite) was et al. in rats demonstrated an immediate decrease in blood
one of the first molecules described to affect Th17-Treg dif- pressure after the injection of neutralizing anti-IL-17 antibod-
ferentiation. Here, RA was shown to blunt IL-6 stimulation ies [2]. In contrast, Marko et al. did not observe any lowering
and to promote Treg formation [46]. Along this same line, of blood pressure upon anti-IL-23 or anti-IL-17 antibody
cellular metabolism is emerging as a key feature defining the treatment [43]. This latter study is in agreement with a report
difference between Th17 and Treg cells [56], which when by Krebs et al., which showed that genetic disruption of the
modulated, can promote the formation of one or the other cell IL-23/IL-17 axis does not alter hypertension after angiotensin
type. Th17 cells are characterized by glycolytic metabolism, treatment in the presence of excess mineralocorticoids [35].
controlled by HIF1α, which when deleted results in the for- One alternative explanation for this inconsistency in de-
mation of Treg [56]. In contrast, Treg cells depend on lipid scribing the role of IL-17 in atherosclerosis and hypertension
oxidation and mitochondrial oxidative phosphorylation. The could be due to a lack of characterization of the gut
modulation of Th17 cellular metabolism from glycolysis to microbiome in these studies. Commensal intestinal bacteria
oxidative phosphorylation with chemicals such as Rapamycin influence multiple metabolic and physiological functions in
[18, 56], or biomolecules such as Soraphen A [6], also pro- the host, and thus have the potential to profoundly affect the
motes differentiation into Treg. These studies further highlight host immune system [63]. Ivanov et al. observed that inbred
the sensitivity of Th17 cells to their microenvironment. C57BL/6 mice obtained from Taconic Biosciences Inc.
Pflugers Arch - Eur J Physiol (2015) 467:543–550 545

developed a higher Th17-responsive phenotype compared to apparently dispensable for basic cellular and organ function.
mice obtained from Jackson Laboratories, indicating that the In contrast, various experimental and clinical studies have
Taconic environment/diet increased the levels of gut- demonstrated that excessive SGK1 expression and activity
associated Th17 cells [31, 32]. This has, as yet, not been increases the pathogenesis of several disorders including hy-
deduced in the setting of atherosclerosis or hypertension. pertension, obesity, diabetes, thrombosis, stroke, fibrotic dis-
eases, infertility, and tumor growth. A SGK1 gene variant in
exon 6 (I6CC) and 8 (E8CC/CT) with a prevalence of 3–5 %
Regulation of SGK1 expression and activity in Caucasians and about 10 % in Africans is associated with
and SGK1-regulated functions hypertension [11, 69], QT interval alterations [11], stroke [17],
obesity, and type 2 diabetes [53]. It was also shown that
High dietary salt (sodium chloride; NaCl) is an environmental increased SGK1 activity promotes pulmonary vascular re-
risk factor newly postulated to be linked to autoimmune dis- modeling [5] and macrophage activation in the development
eases [16, 25, 28, 45, 58]. A molecule important for sensing and of pulmonary arterial hypertension [76]. In addition, SGK1
responding to changes in extracellular Na+ is the serum- and may play a decisive role in promoting fetal programming of
glucocorticoid-inducible kinase (SGK1), which was cloned as a hypertension induced by prenatal protein restriction. Follow-
gene regulated by the hydration state of a cell [71, reviewed by ing prenatal protein restriction, the weight gain in offspring of
26]. SGK1 is a downstream target of tonicity-responsive en- wild-type mothers was slower and associated with higher
hancer binding protein (TonEBP; also known as Nuclear factor blood pressure after birth. In contrast, prenatal protein restric-
of activated T cells 5 (NFAT5)) [14]. Under physiological tion of SGK1 knockout mothers had no effect on postnatal
conditions, SGK1 is expressed at low levels but is significantly weight gain and blood pressure [51]. SGK1 knockout mice
increased under glucocorticoid or mineralocorticoid excess, infused with angiotensin II [77] or mineralocorticoid excess
hyperglycemia, ischemia, inflammation and hypertonicity, or with high salt [66] did not show an altered progression of
cell shrinkage [38]. Upon stimulation with insulin and growth hypertension although cardiac hypertrophy, renal damage, and
factors such as TGF-β, SGK1 is phosphorylated within mi- cardiac and renal fibrosis were markedly reduced. In addition,
nutes via signaling pathways involving phosphatidylinositide- SGK1 can promote cardiac remodeling and development of
3-kinase, 3-phosphoinositide-dependent kinase, and the mam- heart failure following an excessive pressure overload [68].
malian target of Rapamycin [50]. TGFβ stimulation also Taken together, the role of SGK1 in the development of
upregulates SGK1 gene expression [37, 72]. In contrast, aldo- hypertension and cardiovascular disease remains ambiguous,
sterone upregulates SGK1 solely on the genomic level via the suggesting the existence of unknown mechanisms.
mineralocorticoid receptor [62]. Altogether, non-genomic or
genomic activation of SGK1 leads to the increased activity of
a variety of ion channels including the epithelial Na+ channel Role of sodium, SGK1 in Th17 polarization
ENaC [70], the voltage gated Na+ channel SCN5A [7], the and autoimmunity
transient receptor potential channels TRPV4-6 [7, 38], the Ca2+
release activated Ca2+ channel Orai1/STIM1 [24], the K+ chan- Two recent studies have shed new light on the role of sodium,
nel ROMK [30], the voltage gated K+ channels KCNE1/ SGK1, and the differentiation of naïve T cells to Th17 cells
KCNQ1 [22] and cystic fibrosis transmembrane regulator [33, 75]. Wu et al. performed genome-wide messenger RNA
[70], several ion carriers (including Na+/Cl− NCC, Na+/K+/ (mRNA) profiling to identify molecular mechanisms by
2Cl− NKCC2 cotransporter, Na+/H+ exchangers NHE1 and which naïve T cells develop into Th17 cells. Surprisingly,
NHE3, Na+/glucose symporter SGLT1) [38], as well as Na+/ SGK1 was one of the top-ranked genes whose transcriptional
K+-ATPase [54]. Furthermore, SGK1 affects the activity of regulation was clearly associated with IL-23 signaling and
several molecules; it increases the phosphorylation of ubiquitin highly induced with Th17 differentiation. Interestingly,
ligase Nedd4-2 [19] and glycogen synthase kinase-3 [47], SGK1 was not equally expressed in other T cell subsets
negatively regulates transcription factors such as forkhead tran- (Th0, Th1, or Th2). TGF-β-induced regulatory T cells
scription factor 3a [9, 20] and beta-catenin [20], and enhances (iTreg) had weak SGK1 expression [29, 75]. IL-23 was shown
the activity of the transcription factor nuclear factor kappa B to specifically induce and maintain the expression of SGK1 in
[59]. Th17 cells [75]. Utilizing a curated database of protein-protein
interaction analysis, the authors further identified the tran-
scription factor forkhead box O (FOXO) 1 as one of the
SGK1, hypertension, and cardiovascular organ damage highest-ranking nodes downstream of SGK1. FOXO1 is
known to direct or fine-tune multiple biological functions that
The phenotype of SGK1 knockout mice is mild, and these are crucial for metabolism and differentiation of cells, includ-
mice show normal blood pressure [66]. Thus, SGK1 is ing cell cycle, apoptosis, oxidative stress response, enzyme
546 Pflugers Arch - Eur J Physiol (2015) 467:543–550

IL-23r

p-p38
Th17
Th17 polarizing NFAT5 polarizing
Naive conditions 1.
phospo-FOXO1
conditions Th17
SGK1
T cell NaCl-induced
hypertonicity 2.
phospo-FOXO1
4.
cells
3. RORγt

Fig. 1 The induction of Th17 cells is augmented by salt via SGK1-dependent mechanisms

activities, or DNA damage repair [10, 13, 65]. Interestingly, di hypertonicity leads to cell shrinkage accompanied by the
Pietro et al. showed that SGK1 promotes adipocyte differen- influx of extracellular sodium via ENaC, NHE1, and
tiation by regulating FOXO1 phosphorylation and thus re- NKCC2 [36]. This adaptation to hypertonicity is controlled
vealed a potentially important function for this kinase in the in mammalian cells by multiple signaling pathways that
control of body fat mass and function [21]. Upon FOXO1 include p38 MAP kinase [55]. A direct outcome of p38
phosphorylation, FOXO is deactivated and translocates from MAP kinase signaling upon hypertonicity is NFAT5 acti-
the nucleus to the cytoplasm. In SGK1-deficient T cells, IL-23 vation [3]. This has been shown to be important in vivo,
stimulation leads to a compromised nuclear phosphorylation where macrophages utilize this signaling axis to regulate
of FOXO1 leading to an increase of its own mRNA and whole-body sodium storage [73]. Downstream of this,
protein expression [75]. Regarding Th17 cell differentiation, SGK1 is a direct target of NFAT5 [14]. Thus, under
Wu et al. suggested that SGK1-deficient cells have enriched Th17-polarizing conditions, blockade, or genetic knock-
FOXO1 binding to the IL-23 receptor (IL-23r) promoter, down of either p38 MAP kinase, NFAT5 or SGK1
providing an explanation for the increased suppression of prevented Na+-mediated Th17 induction, thus identifying
IL-23r transcription. Vice versa, IL-23-induced SGK1 phos- the molecular mechanism by which excess salt enhances
phorylation led to reduced FOXO1 activity and increased IL- Th17 differentiation [33]. Importantly, a high salt diet was
23r expression and induction of retinoic-acid-receptor-related shown to accelerate the pathogenesis of experimental au-
orphan receptor γt (RORγt), a master regulator of Th17 toimmune encephalomyelitis (EAE), a mouse model of
development (Fig. 1). In the same paper, Wu et al. demon- multiple sclerosis, demonstrating an increased induction
strated that SGK1 deletion in CD4+ or IL-17-specific T cells of Th17 cells in vivo [33, 75]. Heightened EAE induction
resulted in a milder disease course in an experimental model was dependent on SGK1 expression, since the specific
of multiple sclerosis due to the differentiation of less patho- deletion of SGK1 in CD4+ expressing cells prevented the
genic Th17 cells [75]. high-salt-induced effect [75].
SGK1 has also been shown to be important in the
differentiation of human T cells. Here, a Th17-promoting
cytokine cocktail (IL-1β, IL-6, IL-21, IL-23, and Conclusion
TGF-β1), together with increased NaCl-concentrations,
substantially accelerated the induction of Th17 cells [33]. Altogether, these recent studies demonstrated that SGK1
Th17 cells produced in this manner displayed an inflam- could play an important role in promoting the differentiation
matory phenotype, as indicated by the increased induction of Th17 cells in vitro and in vivo [49]. Under elevated salt
of other pro-inflammatory cytokines like CSF2 (also called concentrations, Na+-mediated hypertonicity accelerates a
GM-CSF). Surprisingly, the effect by which salt promoted SGK1 dependent signaling cascade, leading to an increased
Th17 differentiation was dependent on Na+, as non-ionic induction of pro-inflammatory Th17 cells. Excess dietary salt
hypertonic osmolytes (such mannitol or urea) were not able intake might therefore represent an environmental risk factor
to promote the same degree of Th17 cell induction. Th1 for the development (or exacerbation) of autoimmune diseases
and Th2 polarization was not affected by Na+-induced via the induction of pathogenic Th17 cells. Interestingly, a
hypertonicity, suggesting a specific effect of salt on Th17 recent observational study by Farez et al. reported that patients
differentiation. As described above, extracellular with relapsing-remitting multiple sclerosis who consume high
Pflugers Arch - Eur J Physiol (2015) 467:543–550 547

levels of sodium have an increased risk of disease exacerba- 9. Brunet A, Park J, Tran H, Hu LS, Hemmings BA, Greenberg ME
(2001) Protein kinase SGK mediates survival signals by phosphory-
tion and clinical and radiological disease activity [25]. As
lating the forkhead transcription factor FKHRL1 (FOXO3a). Mol
Th17 cells also contribute to the development of hypertension Cell Biol 21(3):952–965. doi:10.1128/MCB. 21.3.952-965.2001
and cardiovascular diseases, we thus speculate that the mech- 10. Burgering BM, Medema RH (2003) Decisions on life and death:
anisms related to heightened CNS autoimmunity may also be FOXO forkhead transcription factors are in command when PKB/
Akt is off duty. J Leukoc Biol 73(6):689–701
applicable to these diseases.
11. Busjahn A, Aydin A, Uhlmann R, Krasko C, Bahring S, Szelestei T,
Upon stimulation with Th17 polarizing conditions, p38 Feng Y, Dahm S, Sharma AM, Luft FC, Lang F (2002) Serum- and
MAP kinase (p38) is phosphorylated, followed by an induc- glucocorticoid-regulated kinase (SGK1) gene and blood pressure.
tion of NFAT5 and SGK1 (1). SGK1 itself then induces the Hypertension 40(3):256–260
12. Butcher MJ, Gjurich BN, Phillips T, Galkina EV (2012) The IL-17A/
phosphorylation of FOXO1. Normally, FOXO1 acts as a
IL-17RA axis plays a proatherogenic role via the regulation of aortic
repressor of the IL-23 receptor (IL-23r) by directly binding myeloid cell recruitment. Circ Res 110(5):675–687. doi:10.1161/
to the IL-23r promoter and inhibiting RORγt-mediated IL-23r CIRCRESAHA.111.261784
transactivation. Phosphorylation of FOXO1 leads to its deac- 13. Chang YW, Zhao YF, Cao YL, Gu XF, Li ZQ, Wang SQ, Miao JH,
Zhan HS (2013) Liver X receptor alpha inhibits osteosarcoma cell
tivation, resulting in its export from the nucleus to the cyto-
proliferation through up-regulation of FoxO1. Cell Physiol Biochem
plasm (2), thereby promoting unopposed RORγt (3) mediated 32(1):180–186. doi:10.1159/000350134
IL-23r transcription (4). In summary, Th17 polarizing condi- 14. Chen S, Grigsby CL, Law CS, Ni X, Nekrep N, Olsen K, Humphreys
tions in combination with Na+-mediated hypertonicity accel- MH, Gardner DG (2009) Tonicity-dependent induction of Sgk1
expression has a potential role in dehydration-induced natriuresis in
erates this SGK1-dependent signaling cascade, leading to an
rodents. J Clin Invest 119(6):1647–1658. doi:10.1172/JCI35314
increased induction of pro-inflammatory Th17 cells. 15. Chen J, Liao MY, Gao XL, Zhong Q, Tang TT, Yu X, Liao YH,
Cheng X (2013) IL-17A induces pro-inflammatory cytokines pro-
duction in macrophages via MAPKinases, NF-kappaB and AP-1.
Cell Physiol Biochem 32(5):1265–1274. doi:10.1159/000354525
16. D’Elia L, Galletti F, Strazzullo P (2014) Dietary salt intake and risk of
References gastric cancer. Cancer Treat Res 159:83–95. doi:10.1007/978-3-642-
38007-5_6
17. Dahlberg J, Smith G, Norrving B, Nilsson P, Hedblad B, Engstrom G,
1. Alunno A, Carubbi F, Bartoloni E, Bistoni O, Caterbi S, Cipriani P, Lovkvist H, Carlson J, Lindgren A, Melander O (2011) Genetic
Giacomelli R, Gerli R (2014) Unmasking the pathogenic role of IL- variants in serum and glucocortocoid regulated kinase 1, a regulator
17 axis in primary Sjogren’s syndrome: a new era for therapeutic of the epithelial sodium channel, are associated with ischaemic
targeting? Autoimmun Rev. doi:10.1016/j.autrev.2014.08.022 stroke. J Hypertens 29(5):884–889. doi:10.1097/HJH.
2. Amador CA, Barrientos V, Pena J, Herrada AA, Gonzalez M, Valdes 0b013e3283455117
S, Carrasco L, Alzamora R, Figueroa F, Kalergis AM, Michea L 18. Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Bordman Z,
(2014) Spironolactone decreases DOCA-salt-induced organ damage Fu J, Kim Y, Yen HR, Luo W, Zeller K, Shimoda L, Topalian SL,
by blocking the activation of T helper 17 and the downregulation of Semenza GL, Dang CV, Pardoll DM, Pan F (2011) Control of T
regulatory T lymphocytes. Hypertension 63(4):797–803. doi:10. (H)17/T (reg) balance by hypoxia-inducible factor 1. Cell 146(5):
1161/HYPERTENSIONAHA.113.02883 772–784. doi:10.1016/j.cell.2011.07.033
3. Aramburu J, Lopez-Rodriguez C (2009) Brx shines a light on the 19. Debonneville C, Flores SY, Kamynina E, Plant PJ, Tauxe C,
route from hyperosmolarity to NFAT5. Sci Signal 2 (65):pe20. doi: Thomas MA, Munster C, Chraibi A, Pratt JH, Horisberger JD,
10.1126/scisignal.265pe20 Pearce D, Loffing J, Staub O (2001) Phosphorylation of
4. Bach JF (2002) The effect of infections on susceptibility to autoim- Nedd4-2 by Sgk1 regulates epithelial Na(+) channel cell sur-
mune and allergic diseases. N Engl J Med 347(12):911–920. doi:10. face expression. Embo J 20(24):7052–7059. doi:10.1093/
1056/NEJMra020100 emboj/20.24.7052
5. BelAiba RS, Djordjevic T, Bonello S, Artunc F, Lang F, Hess J, 20. Dehner M, Hadjihannas M, Weiske J, Huber O, Behrens J (2008)
Gorlach A (2006) The serum- and glucocorticoid-inducible kinase Wnt signaling inhibits forkhead box O3a-induced transcription and
Sgk-1 is involved in pulmonary vascular remodeling: role in redox- apoptosis through up-regulation of serum- and glucocorticoid-
sensitive regulation of tissue factor by thrombin. Circ Res 98(6):828– inducible kinase 1. J Biol Chem 283(28):19201–19210. doi:10.
836. doi:10.1161/01.RES.0000210539.54861.27 1074/jbc.M710366200
6. Berod L, Friedrich C, Nandan A, Freitag J, Hagemann S, Harmrolfs 21. Di Pietro N, Panel V, Hayes S, Bagattin A, Meruvu S, Pandolfi A,
K, Sandouk A, Hesse C, Castro CN, Bahre H, Tschirner SK, Gorinski Hugendubler L, Fejes-Toth G, Naray-Fejes-Toth A, Mueller E (2010)
N, Gohmert M, Mayer CT, Huehn J, Ponimaskin E, Abraham WR, Serum- and glucocorticoid-inducible kinase 1 (SGK1) regulates ad-
Muller R, Lochner M, Sparwasser T (2014) De novo fatty acid ipocyte differentiation via forkhead box O1. Mol Endocrinol 24(2):
synthesis controls the fate between regulatory T and T helper 17 370–380. doi:10.1210/me.2009-0265
cells. Nat Med 20(11):1327–1333. doi:10.1038/nm.3704 22. Embark HM, Bohmer C, Vallon V, Luft F, Lang F (2003) Regulation
7. Boehmer C, Wilhelm V, Palmada M, Wallisch S, Henke G, of KCNE1-dependent K (+) current by the serum and glucocorticoid-
Brinkmeier H, Cohen P, Pieske B, Lang F (2003) Serum and gluco- inducible kinase (SGK) isoforms. Pflugers Arch 445(5):601–606.
corticoid inducible kinases in the regulation of the cardiac sodium doi:10.1007/s00424-002-0982-y
channel SCN5A. Cardiovasc Res 57(4):1079–1084 23. Esplugues E, Huber S, Gagliani N, Hauser AE, Town T, Wan YY,
8. Bogdanos DP, Smyk DS, Rigopoulou EI, Mytilinaiou MG, O’Connor W Jr, Rongvaux A, Van Rooijen N, Haberman AM,
Heneghan MA, Selmi C, Gershwin ME (2012) Twin studies in Iwakura Y, Kuchroo VK, Kolls JK, Bluestone JA, Herold KC,
autoimmune disease: genetics, gender and environment. J Flavell RA (2011) Control of TH17 cells occurs in the small intestine.
Autoimmun 38(2–3):J156–169. doi:10.1016/j.jaut.2011.11.003 Nature 475(7357):514–518. doi:10.1038/nature10228
548 Pflugers Arch - Eur J Physiol (2015) 467:543–550

24. Eylenstein A, Gehring EM, Heise N, Shumilina E, Schmidt S, Szteyn 39. Lee Y, Awasthi A, Yosef N, Quintana FJ, Xiao S, Peters A, Wu C,
K, Munzer P, Nurbaeva MK, Eichenmuller M, Tyan L, Regel I, Foller Kleinewietfeld M, Kunder S, Hafler DA, Sobel RA, Regev A,
M, Kuhl D, Soboloff J, Penner R, Lang F (2011) Stimulation of Kuchroo VK (2012) Induction and molecular signature of pathogenic
Ca2+-channel Orai1/STIM1 by serum- and glucocorticoid-inducible TH17 cells. Nat Immunol 13(10):991–999. doi:10.1038/ni.2416
kinase 1 (SGK1). Faseb J 25(6):2012–2021. doi:10.1096/fj.10- 40. Luchtman DW, Ellwardt E, Larochelle C, Zipp F (2014) IL-17 and
178210 related cytokines involved in the pathology and immunotherapy of
25. Farez MF, Fiol MP, Gaitan MI, Quintana FJ, Correale J (2014) multiple sclerosis: current and future developments. Cytokine
Sodium intake is associated with increased disease activity in multi- Growth Factor Rev 25(4):403–413. doi:10.1016/j.cytogfr.2014.07.
ple sclerosis. J Neurol Neurosurg Psychiatry. doi:10.1136/jnnp-2014- 013
307928 41. Luo CT, Li MO (2013) Transcriptional control of regulatory T cell
26. Firestone GL, Giampaolo JR, O’Keeffe BA (2003) Stimulus- development and function. Trends Immunol 34(11):531–539. doi:10.
dependent regulation of serum and glucocorticoid inducible protein 1016/j.it.2013.08.003
kinase (SGK) transcription, subcellular localization and enzymatic 42. Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y, Guzik TJ,
activity. Cell Physiol Biochem 13(1):1–12 Harrison DG (2010) Interleukin 17 promotes angiotensin II-induced
27. Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, hypertension and vascular dysfunction. Hypertension 55(2):500–
Konkel JE, Ramos HL, Wei L, Davidson TS, Bouladoux N, 507. doi:10.1161/HYPERTENSIONAHA.109.145094
Grainger JR, Chen Q, Kanno Y, Watford WT, Sun HW, Eberl 43. Marko L, Kvakan H, Park JK, Qadri F, Spallek B, Binger KJ,
G, Shevach EM, Belkaid Y, Cua DJ, Chen W, O’Shea JJ Bowman EP, Kleinewietfeld M, Fokuhl V, Dechend R, Muller DN
(2010) Generation of pathogenic T(H)17 cells in the absence (2012) Interferon-gamma signaling inhibition ameliorates angioten-
of TGF-beta signalling. Nature 467(7318):967–971. doi:10. sin II-induced cardiac damage. Hypertension 60(6):1430–1436. doi:
1038/nature09447 10.1161/HYPERTENSIONAHA.112.199265
28. He FJ, Li J, Macgregor GA (2013) Effect of longer-term modest salt 44. McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W,
reduction on blood pressure. Cochrane Database Syst Rev 4: McClanahan T, Cua DJ (2007) TGF-beta and IL-6 drive the produc-
CD004937. doi:10.1002/14651858.CD004937.pub2 tion of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated
29. Heikamp EB, Patel CH, Collins S, Waickman A, Oh MH, Sun IH, pathology. Nat Immunol 8(12):1390–1397. doi:10.1038/ni1539
Illei P, Sharma A, Naray-Fejes-Toth A, Fejes-Toth G, Misra-Sen J, 45. Mozaffarian D, Fahimi S, Singh GM, Micha R, Khatibzadeh S,
Horton MR, Powell JD (2014) The AGC kinase SGK1 regulates TH1 Engell RE, Lim S, Danaei G, Ezzati M, Powles J (2014) Global
and TH2 differentiation downstream of the mTORC2 complex. Nat sodium consumption and death from cardiovascular causes. N Engl J
Immunol 15(5):457–464. doi:10.1038/ni.2867 Med 371(7):624–634. doi:10.1056/NEJMoa1304127
30. Huang DY, Wulff P, Volkl H, Loffing J, Richter K, Kuhl D, Lang F, 46. Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M,
Vallon V (2004) Impaired regulation of renal K+ elimination in the Cheroutre H (2007) Reciprocal TH17 and regulatory T cell differen-
sgk1-knockout mouse. J Am Soc Nephrol 15(4):885–891 tiation mediated by retinoic acid. Science 317(5835):256–260. doi:
31. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei 10.1126/science.1145697
D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, 47. Murray JT, Campbell DG, Morrice N, Auld GC, Shpiro N, Marquez
Takeda K, Umesaki Y, Honda K, Littman DR (2009) Induction of R, Peggie M, Bain J, Bloomberg GB, Grahammer F, Lang F, Wulff P,
intestinal Th17 cells by segmented filamentous bacteria. Cell 139(3): Kuhl D, Cohen P (2004) Exploitation of KESTREL to identify
485–498. doi:10.1016/j.cell.2009.09.033 NDRG family members as physiological substrates for SGK1 and
32. Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, Sartor GSK3. Biochem J 384(Pt 3):477–488. doi:10.1042/BJ20041057
RB, Finlay BB, Littman DR (2008) Specific microbiota direct the 48. Nestle FO, Kaplan DH, Barker J (2009) Psoriasis. N Engl J Med
differentiation of IL-17-producing T-helper cells in the mucosa of the 361(5):496–509. doi:10.1056/NEJMra0804595
small intestine. Cell Host Microbe 4(4):337–349. doi:10.1016/j. 49. Norton M, Screaton RA (2014) SGK1: master and commander of the
chom.2008.09.009 fate of helper T cells. Nat Immunol 15(5):411–413. doi:10.1038/ni.
33. Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker 2875
RA, Muller DN, Hafler DA (2013) Sodium chloride drives autoim- 50. Park J, Leong ML, Buse P, Maiyar AC, Firestone GL, Hemmings BA
mune disease by the induction of pathogenic T17 cells. Nature. doi: (1999) Serum and glucocorticoid-inducible kinase (SGK) is a target
10.1038/nature11868 of the PI 3-kinase-stimulated signaling pathway. Embo J 18(11):
34. Korn T, Bettelli E, Oukka M, Kuchroo VK (2009) IL-17 and Th17 3024–3033. doi:10.1093/emboj/18.11.3024
Cells. Annu Rev Immunol 27:485–517. doi:10.1146/annurev. 51. Rexhepaj R, Boini KM, Huang DY, Amann K, Artunc F, Wang K,
immunol.021908.132710 Brosens JJ, Kuhl D, Lang F (2008) Role of maternal glucocorticoid
35. Krebs CF, Lange S, Niemann G, Rosendahl A, Lehners A, Meyer- inducible kinase SGK1 in fetal programming of blood pressure in
Schwesinger C, Stahl RA, Benndorf RA, Velden J, Paust HJ, Panzer response to prenatal diet. Am J Physiol Regul Integr Comp Physiol
U, Ehmke H, Wenzel UO (2014) Deficiency of the interleukin 17/23 294(6):R2008–2013. doi:10.1152/ajpregu.00737.2007
axis accelerates renal injury in mice with deoxycorticosterone ace- 52. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA,
tate+angiotensin ii-induced hypertension. Hypertension 63(3):565– Moutsianas L, Dilthey A, Su Z, Freeman C, Hunt SE, Edkins S, Gray
571. doi:10.1161/HYPERTENSIONAHA.113.02620 E, Booth DR, Potter SC, Goris A, Band G, Oturai AB, Strange A,
36. Lang F (2011) Effect of cell hydration on metabolism. Nestle Nutr Inst Saarela J, Bellenguez C, Fontaine B, Gillman M, Hemmer B,
Workshop Ser 69:115–126. doi:10.1159/000329290, discussion 126–130 Gwilliam R, Zipp F, Jayakumar A, Martin R, Leslie S, Hawkins S,
37. Lang F, Klingel K, Wagner CA, Stegen C, Warntges S, Friedrich B, Giannoulatou E, D’Alfonso S, Blackburn H, Martinelli Boneschi F,
Lanzendorfer M, Melzig J, Moschen I, Steuer S, Waldegger S, Sauter Liddle J, Harbo HF, Perez ML, Spurkland A, Waller MJ, Mycko MP,
M, Paulmichl M, Gerke V, Risler T, Gamba G, Capasso G, Kandolf Ricketts M, Comabella M, Hammond N, Kockum I, McCann OT,
R, Hebert SC, Massry SG, Broer S (2000) Deranged transcriptional Ban M, Whittaker P, Kemppinen A, Weston P, Hawkins C, Widaa S,
regulation of cell-volume-sensitive kinase hSGK in diabetic nephrop- Zajicek J, Dronov S, Robertson N, Bumpstead SJ, Barcellos LF,
athy. Proc Natl Acad Sci U S A 97(14):8157–8162 Ravindrarajah R, Abraham R, Alfredsson L, Ardlie K, Aubin C,
38. Lang F, Voelkl J (2013) Therapeutic potential of serum and gluco- Baker A, Baker K, Baranzini SE, Bergamaschi L, Bergamaschi R,
corticoid inducible kinase inhibition. Expert Opin Investig Drugs Bernstein A, Berthele A, Boggild M, Bradfield JP, Brassat D,
22(6):701–714. doi:10.1517/13543784.2013.778971 Broadley SA, Buck D, Butzkueven H, Capra R, Carroll WM,
Pflugers Arch - Eur J Physiol (2015) 467:543–550 549

Cavalla P, Celius EG, Cepok S, Chiavacci R, Clerget-Darpoux F, 62. Terada Y, Kuwana H, Kobayashi T, Okado T, Suzuki N, Yoshimoto
Clysters K, Comi G, Cossburn M, Cournu-Rebeix I, Cox MB, Cozen T, Hirata Y, Sasaki S (2008) Aldosterone-stimulated SGK1 activity
W, Cree BA, Cross AH, Cusi D, Daly MJ, Davis E, de Bakker PI, mediates profibrotic signaling in the mesangium. J Am Soc Nephrol
Debouverie M, D’Hooghe MB, Dixon K, Dobosi R, Dubois B, 19(2):298–309. doi:10.1681/ASN.2007050531
Ellinghaus D, Elovaara I, Esposito F, Fontenille C, Foote S, Franke 63. Tremaroli V, Backhed F (2012) Functional interactions between the
A, Galimberti D, Ghezzi A, Glessner J, Gomez R, Gout O, Graham gut microbiota and host metabolism. Nature 489(7415):242–249.
C, Grant SF, Guerini FR, Hakonarson H, Hall P, Hamsten A, Hartung doi:10.1038/nature11552
HP, Heard RN, Heath S, Hobart J, Hoshi M, Infante-Duarte C, 64. Tsoi LC, Spain SL, Knight J, Ellinghaus E, Stuart PE, Capon F, Ding
Ingram G, Ingram W, Islam T, Jagodic M, Kabesch M, Kermode J, Li Y, Tejasvi T, Gudjonsson JE, Kang HM, Allen MH, McManus
AG, Kilpatrick TJ, Kim C, Klopp N, Koivisto K, Larsson M, Lathrop R, Novelli G, Samuelsson L, Schalkwijk J, Stahle M, Burden AD,
M, Lechner-Scott JS, Leone MA, Leppa V, Liljedahl U, Bomfim IL, Smith CH, Cork MJ, Estivill X, Bowcock AM, Krueger GG, Weger
Lincoln RR, Link J, Liu J, Lorentzen AR, Lupoli S, Macciardi F, W, Worthington J, Tazi-Ahnini R, Nestle FO, Hayday A, Hoffmann
Mack T, Marriott M, Martinelli V, Mason D, McCauley JL, Mentch P, Winkelmann J, Wijmenga C, Langford C, Edkins S, Andrews R,
F, Mero IL, Mihalova T, Montalban X, Mottershead J, Myhr KM, Blackburn H, Strange A, Band G, Pearson RD, Vukcevic D, Spencer
Naldi P, Ollier W, Page A, Palotie A, Pelletier J, Piccio L, Pickersgill CC, Deloukas P, Mrowietz U, Schreiber S, Weidinger S, Koks S,
T, Piehl F, Pobywajlo S, Quach HL, Ramsay PP, Reunanen M, Kingo K, Esko T, Metspalu A, Lim HW, Voorhees JJ, Weichenthal
Reynolds R, Rioux JD, Rodegher M, Roesner S, Rubio JP, Ruckert M, Wichmann HE, Chandran V, Rosen CF, Rahman P, Gladman DD,
IM, Salvetti M, Salvi E, Santaniello A, Schaefer CA, Schreiber S, Griffiths CE, Reis A, Kere J, Nair RP, Franke A, Barker JN, Abecasis
Schulze C, Scott RJ, Sellebjerg F, Selmaj KW, Sexton D, Shen L, GR, Elder JT, Trembath RC (2012) Identification of 15 new psoriasis
Simms-Acuna B, Skidmore S, Sleiman PM, Smestad C, Sorensen susceptibility loci highlights the role of innate immunity. Nat Genet
PS, Sondergaard HB, Stankovich J, Strange RC, Sulonen AM, 44(12):1341–1348. doi:10.1038/ng.2467
Sundqvist E, Syvanen AC, Taddeo F, Taylor B, Blackwell JM, 65. Tzivion G, Dobson M, Ramakrishnan G (2011) FoxO transcrip-
Tienari P, Bramon E, Tourbah A, Brown MA, Tronczynska E, tion factors; Regulation by AKT and 14-3-3 proteins. Biochim
Casas JP, Tubridy N, Corvin A, Vickery J, Jankowski J, Villoslada Biophys Acta 1813(11):1938–1945. doi:10.1016/j.bbamcr.2011.
P, Markus HS, Wang K, Mathew CG, Wason J, Palmer CN, 06.002
Wichmann HE, Plomin R, Willoughby E, Rautanen A, 66. Vallon V, Wyatt AW, Klingel K, Huang DY, Hussain A, Berchtold S,
Winkelmann J, Wittig M, Trembath RC, Yaouanq J, Viswanathan Friedrich B, Grahammer F, Belaiba RS, Gorlach A, Wulff P, Daut J,
AC, Zhang H, Wood NW, Zuvich R, Deloukas P, Langford C, Dalton ND, Ross J Jr, Flogel U, Schrader J, Osswald H, Kandolf R,
Duncanson A, Oksenberg JR, Pericak-Vance MA, Haines JL, Kuhl D, Lang F (2006) SGK1-dependent cardiac CTGF formation
Olsson T, Hillert J, Ivinson AJ, De Jager PL, Peltonen L, Stewart and fibrosis following DOCA treatment. J Mol Med (Berl) 84(5):
GJ, Hafler DA, Hauser SL, McVean G, Donnelly P, Compston A 396–404. doi:10.1007/s00109-005-0027-z
(2011) Genetic risk and a primary role for cell-mediated immune 67. van Es T, van Puijvelde GH, Ramos OH, Segers FM, Joosten LA,
mechanisms in multiple sclerosis. Nature 476(7359):214–219 van den Berg WB, Michon IM, de Vos P, van Berkel TJ, Kuiper J
53. Schwab M, Lupescu A, Mota M, Mota E, Frey A, Simon P, Mertens (2009) Attenuated atherosclerosis upon IL-17R signaling disruption
PR, Floege J, Luft F, Asante-Poku S, Schaeffeler E, Lang F (2008) in LDLr deficient mice. Biochem Biophys Res Commun 388(2):
Association of SGK1 gene polymorphisms with type 2 diabetes. Cell 261–265. doi:10.1016/j.bbrc.2009.07.152
Physiol Biochem 21(1–3):151–160. doi:10.1159/000113757 68. Voelkl J, Lin Y, Alesutan I, Ahmed MS, Pasham V, Mia S, Gu S,
54. Setiawan I, Henke G, Feng Y, Bohmer C, Vasilets LA, Schwarz W, Feger M, Saxena A, Metzler B, Kuhl D, Pichler BJ, Lang F (2012)
Lang F (2002) Stimulation of Xenopus oocyte Na(+), K(+)ATPase Sgk1 sensitivity of Na(+)/H(+) exchanger activity and cardiac re-
by the serum and glucocorticoid-dependent kinase sgk1. Pflugers modeling following pressure overload. Basic Res Cardiol 107(2):
Arch 444(3):426–431. doi:10.1007/s00424-002-0823-z 236. doi:10.1007/s00395-011-0236-2
55. Shapiro L, Dinarello CA (1995) Osmotic regulation of cytokine 69. von Wowern F, Berglund G, Carlson J, Mansson H, Hedblad B,
synthesis in vitro. Proc Natl Acad Sci U S A 92(26):12230–12234 Melander O (2005) Genetic variance of SGK-1 is associated with
56. Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, Chi H blood pressure, blood pressure change over time and strength of the
(2011) HIF1alpha-dependent glycolytic pathway orchestrates a met- insulin-diastolic blood pressure relationship. Kidney Int 68(5):2164–
abolic checkpoint for the differentiation of TH17 and Treg cells. J 2172. doi:10.1111/j.1523-1755.2005.00672.x
Exp Med 208(7):1367–1376. doi:10.1084/jem.20110278 70. Wagner CA, Ott M, Klingel K, Beck S, Melzig J, Friedrich B, Wild
57. Singh RP, Hasan S, Sharma S, Nagra S, Yamaguchi DT, Wong D, Bh KN, Broer S, Moschen I, Albers A, Waldegger S, Tummler B, Egan
H, Hossain A (2014) Th17 cells in inflammation and autoimmunity. ME, Geibel JP, Kandolf R, Lang F (2001) Effects of the serine/
Autoimmun Rev. doi:10.1016/j.autrev.2014.08.019 threonine kinase SGK1 on the epithelial Na(+) channel (ENaC) and
58. Sundstrom B, Johansson I, Rantapaa-Dahlqvist S (2014) Interaction CFTR: implications for cystic fibrosis. Cell Physiol Biochem 11(4):
between dietary sodium and smoking increases the risk for rheuma- 209–218
toid arthritis: results from a nested case–control study. Rheumatology 71. Waldegger S, Barth P, Raber G, Lang F (1997) Cloning and charac-
(Oxford). doi:10.1093/rheumatology/keu330 terization of a putative human serine/threonine protein kinase tran-
59. Tai DJ, Su CC, Ma YL, Lee EH (2009) SGK1 phosphorylation of scriptionally modified during anisotonic and isotonic alterations of
IkappaB Kinase alpha and p300 Up-regulates NF-kappaB activity cell volume. Proc Natl Acad Sci U S A 94(9):4440–4445
and increases N-Methyl-D-aspartate receptor NR2A and NR2B ex- 72. Waldegger S, Klingel K, Barth P, Sauter M, Rfer ML, Kandolf R,
pression. J Biol Chem 284(7):4073–4089. doi:10.1074/jbc. Lang F (1999) h-sgk serine-threonine protein kinase gene as tran-
M805055200 scriptional target of transforming growth factor beta in human intes-
60. Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, tine. Gastroenterology 116(5):1081–1088
Littman DR, O’Shea JJ (2009) Lymphoid tissue inducer-like cells are 73. Wiig H, Schroder A, Neuhofer W, Jantsch J, Kopp C, Karlsen TV,
an innate source of IL-17 and IL-22. J Exp Med 206(1):35–41. doi: Boschmann M, Goss J, Bry M, Rakova N, Dahlmann A, Brenner S,
10.1084/jem.20072713 Tenstad O, Nurmi H, Mervaala E, Wagner H, Beck FX, Muller DN,
61. Taleb S, Tedgui A, Mallat Z (2014) IL-17 and Th17 cells in athero- Kerjaschki D, Luft FC, Harrison DG, Alitalo K, Titze J (2013) Immune
sclerosis: subtle and contextual roles. Arterioscler Thromb Vasc Biol. cells control skin lymphatic electrolyte homeostasis and blood pres-
doi:10.1161/ATVBAHA.114.303567 sure. J Clin Invest 123(7):2803–2815. doi:10.1172/JCI60113
550 Pflugers Arch - Eur J Physiol (2015) 467:543–550

74. Wilson FC, Icen M, Crowson CS, McEvoy MT, Gabriel SE, Kremers 77. Yang M, Zheng J, Miao Y, Wang Y, Cui W, Guo J, Qiu S, Han Y, Jia
HM (2009) Time trends in epidemiology and characteristics of pso- L, Li H, Cheng J, Du J (2012) Serum-glucocorticoid regulated kinase
riatic arthritis over 3 decades: a population-based study. J Rheumatol 1 regulates alternatively activated macrophage polarization contrib-
36(2):361–367. doi:10.3899/jrheum.080691 uting to angiotensin II-induced inflammation and cardiac fibrosis.
75. Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, Regev A, Kuchroo Arterioscler Thromb Vasc Biol 32(7):1675–1686. doi:10.1161/
VK (2013) Induction of pathogenic TH17 cells by inducible salt-sensing ATVBAHA.112.248732
kinase SGK1. Nature 496(7446):513–517. doi:10.1038/nature11984 78. Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F,
76. Xi X, Liu S, Shi H, Yang M, Qi Y, Wang J, Du J (2014) Serum- Gattorno M, Monticelli S, Lanzavecchia A, Sallusto F (2012)
glucocorticoid regulated kinase 1 regulates macrophage recruitment Pathogen-induced human TH17 cells produce IFN-gamma or IL-10
and activation contributing to monocrotaline-induced pulmonary arte- and are regulated by IL-1beta. Nature 484(7395):514–518. doi:10.
rial hypertension. Cardiovasc Toxicol. doi:10.1007/s12012-014-9260-4 1038/nature10957
Copyright of Pflugers Archiv European Journal of Physiology is the property of Springer
Science & Business Media B.V. and its content may not be copied or emailed to multiple sites
or posted to a listserv without the copyright holder's express written permission. However,
users may print, download, or email articles for individual use.

Potrebbero piacerti anche