Abstract
Serum and glucocorticoid-inducible kinase 1 (SGK1) is an AGC kinase that has been reported to be involved in a variety of physiological and pathological processes. Recent evidence has accumulated that SGK1 acts as an essential Akt-independent mediator of phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway in cancer. SGK1 is overexpressed in several tumors, including prostate cancer, colorectal carcinoma, glioblastoma, breast cancer, and endometrial cancer. The functions of SGK1 include regulating tumor growth, survival, metastasis, autophagy, immunoregulation, calcium (Ca2+) signaling, cancer stem cells, cell cycle, and therapeutic resistance. In this review, we introduce the pleiotropic role of SGK1 in the development and progression of tumors, summarize its downstream targets, and integrate the knowledge provided by preclinical studies that the prospect of SGK1 inhibition as a potential therapeutic approach.
Keywords: autophagy, calcium signaling, chemoresistance, immunoregulation, PI3K signaling, SGK1, therapeutic target
Introduction
Serum and glucocorticoid-inducible kinase 1 (SGK1) is a member of the AGC kinase family of serine/threonine kinases, sharing a large homologous sequence and kinase function with the Akt family,1–3 but it is distinct from Akt that SGK1 does not possess a pleckstrin homology (PH) domain to directly interact with phosphatidylinositol 3,4,5 trisphosphate.4 SGK1 was initially cloned as an immediate-early gene in rat breast tumor cells stimulated by serum and glucocorticoids, and its function is closely related to the phosphorylation of mammalian target of rapamycin (mTOR).1,5,6 The kinase converts into an open conformation for phosphorylation after mTOR-dependent hydrophobic phosphorylation on Ser422, and is fully activated by 3-phosphoinositide-dependent kinase-1 (PDK1).4,7 The consensus phosphorylation site of SGK1 includes serine or threonine residues in the substrate proteins present in an RXRXX(S/T) sequence motif (where X is any amino acid, and R is arginine).1
SGK1 is expressed ubiquitously in most tissues tested to date, and its subcellular localization is associated with the functional state of the cells. SGK1 enters the nucleus after serum stimulation, but may also be located in the cytosol following activation by hypertonic shock or glucocorticoids.1 SGK expression is regulated by a wide spectrum of stimuli, including dehydration, saline ingestion, glucocorticoids, insulin, growth factors, and various cellular stresses such as cell swelling, metabolic acidosis, neuronal excitotoxicity, ultraviolet irradiation, DNA damage, and ischemia.1,8–13 SGK1 is involved in regulating multiple ion channels, membrane transporters, cellular enzymes, and transcription factors, as well as in a variety of physiological processes including memory consolidation, reproductive process, cell growth, proliferation, survival, migration, and apoptosis.1,3,14,15 SGK1 has been previously reported in hypertension, diabetes, hypercoagulability, and autoimmune disease.5,16,17 A growing number of recent studies reveal a role for SGK1 in human tumors such as glioblastoma,18–20 breast,21–25 colorectal,26–28 prostate,29–31 thyroid,32 ovarian,33 myeloma,11 and non-small cell lung cancer34 (Figure 1).
Figure 1.
The functions of SGK1. SGK1 is involved in a broad range of processes of human physiology and pathophysiology.
SGK1, serum and glucocorticoid-inducible kinase 1.
With such extensive studies on SGK1, the understanding of its role in cancer is continuing to improve. In this review, we summarize the functions of SGK1 in tumor growth, survival, metastasis, autophagy, immunoregulation, Ca2+ signaling, cancer stem cells (CSCs), cell cycle, and therapeutic resistance, to clarify the roles of SGK1 cross-talk in different pathways (Figure 2; Table 1).
Figure 2.
Signaling pathway of SGK1 in oncology. SGK1 regulates tumor growth, survival, metastasis, autophagy, immunoregulation, Ca2+ signaling, CSCs, and the cell cycle through phosphorylating different substrates.
CSC, cancer stem cell; SGK1, serum and glucocorticoid-inducible kinase 1.
Table 1.
The substrates and regulation mechanisms of SGK1.
| Target of SGK1 | Phosphorylation sites | Effects after phosphorylation | References |
|---|---|---|---|
| NDRG1 | Thr328, Ser330, Thr346, Thr356, Thr366 | Activates NDRG1 and regulate its cellular distribution | Park et al.35, Weiler et al.36 |
| Sp1 | Ser59 | Enhances the transcriptional factor function of Sp1 | Amato et al.37, Dattilo et al.38 |
| TSC2 | Ser939, Ser981, Ser1130, Ser1132, Thr1462, Ser1798 | Inhibits TSC2 and decrease its negative function on mTOR1 | Castel et al.39 |
| SEK1 | Ser78 | Inactivates SEK1 | Hasna et al.22, Kim et al.40 |
| MDM2 | Ser166 | Activates MDM2 and promote p53 degradation | Amato et al.41 |
| Foxo3a | Thr32, Ser253, Ser315 | Suppresses Foxo3a transcriptional activity | Liu et al.31, Shelly and Herrera42, Brunet et al.43 |
| Foxo1 | Ser256 | Reduces Foxo1 nuclear exclusion | Wu et al.44,45 |
| GSK-3β | Not report | Inhibits GSK-3β and blocks β-catenin phosphorylation | Heikamp et al.46, Norton and Screaton47 |
| Nedd4-2 | Ser221, Thr246, Ser327 | Blocks Nedd4-2 interacting with its substrates and prevents its ubiquitin activity | Snyder48 |
| IKKβ | Ser177, Ser181 | Enhances the activity of IKKβ to phosphorylate IκB | Zhang et al.49 |
| p300 | Ser1834 | Enhances p300 histone acetyltransferase ability | Tai et al.50 |
SGK1, serum and glucocorticoid-inducible kinase 1.
Multiple factors regulate SGK1 in oncology
SGK1 transcription is stimulated by multiple factors, including hormones, cytokines, stress response, and multiple signaling pathways and physiological processes.5 In this review, we focus on stimuli with potential roles in the oncological activities of SGK1.
SGK1 is known to be activated by insulin via phosphatidylinositol 3-kinase (PI3K) signaling. SGK1 is phosphorylated by mTOR complex 2 (mTORC2) at Ser422, which offers a docking site for PDK1. PDK1 then fully activates SGK1 by phosphorylating Thr256 in the activation loop of its catalytic domain.7
In addition to mTORC2 and PDK1, SGK1 activation is also related to other cofactors through transcriptional or posttranslational regulation. During DNA damage, SGK1 is significantly induced in a p53-dependent manner via extracellular signal-regulated kinase 1/2 (ERK1/2).8 Interleukin-2 (IL-2) also induces SGK1 protein expression, possibly via transcriptional activation and posttranscriptional phosphorylation.51 Androgen receptor (AR) is a pivotal ligand-activated transcription factor and contributes to the development and progression of prostate cancer. Following androgen stimulation, AR activates the response element motif (5′-CGGACAaaaTGTTCT-3′) at −1159/−1145 region in SGK1 promoter and upregulates SGK1 expression.30 The glucocorticoid receptor (GR) is another transcription factor, which shares an almost identical consensus DNA-binding motif with AR.52 The glucocorticoid receptor thus regulates SGK1 expression and enhances cell survival in prostate cancer and triple-negative breast cancer via a similar mechanism.53,54 β2-microglobulin upregulates phosphorylated SGK1/SGK1 level and promotes cell growth and survival in estrogen receptor-negative and HER2-negative breast cancer through the SGK1/Bcl-2 pathway.55
Functions and mechanisms of SGK1 in oncology
Growth, survival, and metastasis
SGK1 expression is elevated in several tumors, including prostate cancer, colorectal carcinoma, glioblastoma, breast cancer, and endometrial cancer. SGK1 expression is also associated with tumor growth, survival, and metastasis.20,26,49,56,57
The PI3K/Akt/mTOR signaling pathway is abnormally activated in most cancers, and has been considered as a promising therapeutic target.58 Akt is a well-known classic effector of PI3K-mediated activity and phosphorylates numerous substrates involved in cell growth, proliferation, metabolism, survival, and glucose metabolism.6 However, growing evidence has recently pointed to the existence of additional effectors of PI3K, that is SGK1 plays a critical role downstream of PI3K.6,23
SGK1 is required for PI3K-activation-related cancer cell proliferation, while the depletion of SGK1 reduces the proliferation and viability of cancer cells in a wide variety of malignancies, including glioblastoma, colon, prostate, thyroid, and endometrial cancers.19,31,32,59,60 Combined targeting of SGK1 and Akt suppresses cell growth more efficiently than inhibiting either PI3K or Akt alone.32 The activity of mTORC1 is regulated through the tuberous sclerosis complex (TSC)/Ras homolog enriched in brain (RHEB)/mTORC1 axis. SGK1 maintains the activity of mTORC1 by phosphorylating and inhibiting its negative regulator TSC2.39
In addition to mTOR-mediated survival effects, SGK1 blocks apoptosis by preventing the de-attachment-induced dephosphorylation of Foxo3a (previously known as FKHRL1).42 SGK1 suppresses Foxo3a transcriptional activity by phosphorylating its regulatory sites at Thr32 and Ser315, thus hinders Foxo3a-induced cell cycle arrest and apoptosis.43 SGK1 is not only activated by AR but also regulates AR-mediated gene expression. SGK1 overexpression enhances intracellular AR (iAR) transactivation and promotes cell survival, even in the absence of androgen stimulation.30 Membrane AR (mAR) has also demonstrated a strong iAR-independent tumor-inhibition effect. Inhibition of SGK1 enhances mAR-dependent apoptosis of breast cancer cells.61 Moreover, the pro-survival and anti-apoptosis functions of SGK1 can also be implemented by inhibiting SEK1 binding to JUNK1 and MEKK1 via phosphorylation of SEK1 on Ser78,40 upregulating oncogenic β-catenin,1,26 activating nuclear factor (NF)-κB transcriptional activity,49,62 promoting p53 degradation,41 and increasing glucose uptake and ATP genesis.63
SGK1 level is significantly higher in mesenchymal-subtype lung adenocarcinoma samples, based on RNA-seq data from The Cancer Genome Atlas (TCGA) database.64 SGK1 inhibition attenuates epithelial-mesenchymal transition and metastasis of prostate cancer cells, while overexpression of SGK1 promotes their invasion and migration.65 Similar results are observed in glioblastoma, colorectal, and hepatocellular carcinoma cells. Inhibition of SGK1 decreases the mesenchymal markers N-cadherin, vimentin, and focal adhesion kinase, and reduces the cell motility and invasion abilities.18 As noted previously, mAR demonstrates strong antioxidant and antitumorigenic effects, which are mediated by vinculin phosphorylation and actin reorganization. Transfection with a constitutively active SGK1 mutant effectively dephosphorylates the cell-adhesion protein vinculin and enhances cell motility.66 Interestingly, SGK1 has been shown to reduce cell migration and invasion. Lee et al. found that SGK1 favored colorectal cell differentiation and inhibited colorectal cancer metastasis in an orthotopic xenograft model.28 Godbole et al. found that SGK1 expression was increased under progesterone treatment in breast cancer, which upregulated neuroblastoma-derived Myc downstream regulator gene 1 (NDRG1) via the AP-1 network. Increased NDRG1 expression reduced the activation of multiple cellular kinases and cell migration.21 These inconsistent views may be due to the different models used, suggesting that SGK1 performs specific functions in different situations.
Autophagy
Autophagy is a crucial process in response to anti-tumor therapeutic stresses and is cytotoxic in certain circumstances. PI3K/mTOR and AMP-activated protein kinase are the central signaling pathways regulating autophagy, and SGK1 plays an intermediary role in that progress.67 Inhibition of SGK1 by GSK650394 or shSGK1 promotes autophagy-dependent cell apoptosis via the mTOR-Foxo3a pathway, which is cytotoxic as the cell viability is restored by autophagy inhibitors.31 Dual inhibition of mTOR and SGK1 enhances autophagy activation, thereby increasing the cytotoxic effect and inhibiting the progression of prostate cancer cells.31,65 A similar manifestation of cytotoxic autophagy is also observed in endometrial cancer cells56 and glioblastoma multiforme cells after treatment with SGK1 inhibitors.68
The conversion of LC3-I to LC3-II is considered as a marker of autophagy,69 while GRP78, which is usually activated by endoplasmic reticulum stress, has an anti-apoptotic function and plays an important part in anti-radiation and chemotherapy.68 SGK1 overexpression increases phosphorylation of endogenous Foxo3a at Thr32 and Ser257, and triggers the translocation of Foxo3a from the nucleus to the cytoplasm, preventing LC3-I/LC3-II conversion.31 Treatment with SGK1 inhibitor leads to the conversion of LC3-I to LC3-II and decreases GRP78 and Bcl-2 expression, thereby releasing Beclin1 and inducing autophagy.31,56
Immunoregulation
The balance between Th1/Th2 cells is an important factor in anti-tumor immune responses.70 SGK1 regulates the fate of T helper cells by acting as a positive regulator of Th2 differentiation and a negative regulator of Th1 differentiation.46 SGK1 phosphorylates glycogen synthase kinase 3β (GSK-3β) in CD4+ helper T cells, thus blocking the degradation of β-catenin.47 β-catenin, in turn, inhibits downstream targets associated with the Th1 phenotype by increasing the accumulation of T cell factor 1 (TCF1).46 Moreover, SGK1 also phosphorylates Nedd4-2 at Ser221, Thr246, and Ser327, which inhibits the ubiquitination of JunB. The stability of JunB contributes to the expression of IL-4 and GATA-3, which is required by the Th2 cell lineage.48 SGK1 deletion removes the inhibition of GSK-3β and Nedd4-2, thus promoting the Th1 and inhibiting the Th2 phenotype.47
Th1 cells function as the main anti-tumor effector cells and are associated with prolonged survival in different types of tumors.71,72 Heikamp et al. generated T-Sgk1−/− mice in which SGK1 was selectively deleted in CD4+ helper precursor cells, and these mice demonstrated decreased IL-4 and inappropriate IFN-γ release.46 Injection of B16 melanoma cells into wild-type and T-Sgk1−/− mice resulted in increased IFN-γ production and robust immunologic rejection of the tumors.46,73 This suggests that SGK1 inhibitors might have therapeutic value as an immunoadjuvant to tumor therapy by enhancing the Th1-mediated immune response.
In addition to the balance between Th1 and Th2 cells, SGK1 can also affect the balance between Th17 and Treg cells. SGK1 is a significant node of IL-23 signaling and promotes IL-23 receptor expression by phosphorylating Foxo1 and reducing its nuclear exclusion.44 The pathogenic effector functions of Th17 cells are enhanced by the IL-23 receptor, and SGK1 is thus involved in IL-23-mediated enhancement of Th17 cell differentiation.74 Foxp3 is also a critical regulator in Treg cell development and function, and its expression is also controlled by Foxo1 activity.45
Th17 cells and Treg cells have been shown to play dual roles in cancer. Th17 cells accelerate tumor progression by promoting angiogenesis and immunosuppression, but also have an anti-tumor effect by recruiting immune cells, stimulating the effect of CD8+ T cells, and promoting transdifferentiation towards Th1 phenotypes.73 Treg cells are generally thought to maintain immunological self-tolerance and suppress anti-tumor responses75–77; however, some reports also suggested that Treg cells might repress inflammation-driven tumorigenesis.78 Besides, the Th17 switch mediated by SGK1 contributes to multiple chronic inflammatory diseases, including ulcerative colitis. Spagnuolo et al. found that the lymphocytes with high SGK1 level downregulated the SGK1 expression in colonic epithelial cells via the SGK1/Th17/Th13 axis and attenuated the cell viability.79 This finding suggests a potential correlation between inflammatory disease and malignancy mediated by SGK1. More studies are therefore needed to clarify the effects of SGK1 in regulating the fate of immune cells, to improve its role as a potential target in cancer immunotherapy.
Ca2+ signaling
Ca2+ is an important intracellular messenger, and Ca2+ wave acts as a code for information transfer, contributing to the regulation of a range of cellular functions.80 Mechanisms for increasing intracellular Ca2+ concentration include their release from intracellular stores and transport across the cell membrane.81,82 The depletion of Ca2+ in the endoplasmic reticulum activates the CRAC channel, resulting in store-operated Ca2+ entry.82
ORAI and STIM are the key proteins that constitute CRAC channel: ORAI forms highly Ca2+-selective ion channels in the plasma membrane and is regulated by STIM, an endoplasmic reticulum-located Ca2+ sensor.83 The most widely studied isoforms are ORAI1 and STIM1, which can be regulated by diverse kinases including SGK1.59,84 Sustained upregulation of store-operated Ca2+ entry by SGK1 through ORAI1 and STIM1 alters the Ca2+ code, which in turn impacts on tumor cell survival, proliferation, migration, and vascularization.85–88
SGK1 enhances ORAI1 by phosphorylating Nedd4-2. Nedd4-2 is an ubiquitin ligase mediating the degradation of many channel proteins including ORAI1, while phosphorylated Nedd4-2 binds to the protein 14-3-3, making it unable to ubiquitinate ORAI1.89 SGK1 also stimulates the expression of ORAI1 and STIM1 by upregulating NF-κB.59,90 SGK1 phosphorylates IκB kinase β at Ser177 and Ser181, leading to the degradation of IκB and NF-κB nuclear translocation.49 SGK1 can also affect NF-κB via phosphorylation of P300 at Ser1834 to enhance NF-κB acetylation.50
Cancer stem cells
CSCs comprise a subgroup of tumor cells with the capacities for self-renewal and plasticity, analogous to stem cells. CSCs have shown an essential role in tumorigenesis, epithelial-mesenchymal transition, and chemoresistance.91,92 SGK1 acts as a major effector of mTORC2 in colorectal CSCs, and SGK1 knockdown significantly decreases the clonogenicity, invasion, and drug sensitivity of colorectal CSCs.93 The CSC marker CD44 is decreased by inhibition of SGK1 in head and neck squamous cell carcinoma and prostate cancer.94,95 Using large-scale RNAi screens, Kulkarni et al. discovered that depletion of SGK1 reduced the survival and increased apoptosis of glioblastoma multiforme stem-like cells (GBM-SCs), as well as patient-derived GBM-SC lines, whereas SGK1 knockdown had little effect on serum glioma lines in vivo and in vitro.20 Undifferentiated GBM-SCs were selectively sensitive while differentiated cells were tolerant of SGK1 inhibition.20
Wnt/β-catenin, Notch, and Hedgehog are typical signaling pathways involved in CSCs.92 However, although SGK1 has been shown to participate in regulating Notch and Hedgehog signaling, its role in CSCs remains unclear.96,97 A recent study indicated that SGK1 mediated Wnt/β-catenin signaling and facilitated increases in the CSCs population among radioresistant prostate cancer cells.94
The development of the CSCs theory provides a reasonable explanation for tumorigenesis, metastasis, and recurrence.92 However, the definition of CSCs remains ambiguous, and CSCs-targeted drugs have not demonstrated successful therapeutic effects in clinical trials. CSCs are known to exhibit extensive cell plasticity and to respond to multiple signals.92,98 Given that SGK1 is involved in cross-talk among different pathways, its role in the regulation of CSCs deserves further investigation.
Cell cycle disorder
The cell-cycle disorder is a hallmark of cancer progression. The cyclin-dependent kinase (CDK) inhibitor p27 oversees the inhibition of the cyclin E-CDK2 complex in the G0 and G1 phase and the activation of cyclin D-CDKs in early G1 phase.99 Furthermore, the reduced level of p27 is shown to be an independent indicator of poor prognosis in patients with breast cancer.100 SGK1 shuttles dynamically between the nucleus (in S and G2/M phases) and the cytoplasm (in G1 phase).101 SGK1 also phosphorylates p27 at Thr157 and hinders nuclear p27 import, resulting in TGF-β resistance and cell cycle deregulation, while SGK1 silencing leads to G2/M arrest and induces caspase-dependent apoptosis.102 Foxo3a interacts with p27 and downregulates its expression after phosphorylation by SGK1.31
The tumor suppressor p53 is essential for preventing tumor formation by inducing cell cycle arrest and apoptosis. SGK1 phosphorylates MDM2 at Ser166 and mediates MDM2-dependent p53 ubiquitination and proteasomal degradation.41,103 Furthermore, p53 degradation occurs not only via MDM2 but also via Nedd4-2, which is reported to link with SGK1 through SEK1.22 SGK1-induced p53 degradation reveals the effect of chronic psychological stress on cell proliferation, survival, and differentiation.103 Interestingly, p53-dependent activation of ERK1/2 in response to DNA damage can also upregulate SGK1.8
Chemo- and radio-resistance
Recent publications have reported that SGK1 can induce chemical and radiological resistance during the treatment of a variety of human tumors.104 The PI3K-AKT-mTOR signaling pathway is known to be one of the most important pathways for cell survival, growth, proliferation, invasion, apoptosis inhibition, and angiogenesis, and inhibitors of this pathway have been widely investigated in preclinical studies and clinical trials. However, resistance to inhibitors targeting this pathway has recently emerged.23,39 SGK1 shares about 54% identity in the catalytic domain with Akt and is indicated as an important Akt-independent effector in PI3K/mTOR pathway.3
SGK1 overexpression is associated with the resistance to Akt inhibitors and temozolomide (TZM) through phosphorylation of NDRG1. Sommer et al. found that Akt-inhibitor-resistant strains of breast cancer cells from two different clinical trials showed significantly elevated SGK1 levels, compared with Akt-inhibitor-sensitive cell lines.23 They also observed a high degree of phosphorylation of NDRG1 in Akt-inhibitor-resistant cell lines.23 NDRG1 reveals a pleiotropic function, as it is downregulated in colon, prostate, and breast cancers, and acts as a metastasis suppressor,25 while it is upregulated in hepatic and gastric cancer and performs a pro-oncogenic function.105 NDRG1 is a specific substrate of SGK1 and can be robustly phosphorylated by SGK1 at Thr328, Ser330, Thr346, Thr356, and Thr366.106 Among these, phosphorylation at Ser330 and Thr346 determine the cellular distribution and activity of NDRG1.35 Weiler et al. reported that SGK1-triggered phosphorylation of NDRG1 at Thr346 was increased in a glioma cell line resistant to TZM alkylating chemotherapy.36 NDRG1 interacts with the DNA-repair enzyme O6-methylguanine-DNA methyltransferase, which is considered being associated with the resistance to alkane chemotherapies, promoting its stability and activity, while treatment with the SGK1 inhibitor EMD638683 overcomes NDRG 1-mediated TZM tolerance.36
In addition, a high level of SGK1 induces resistance to PI3Kα inhibitors. SGK1 does not contain the PH domain, which is essential for Akt in PI3K-dependent membrane translocation recruited by phosphatidylinositol-(3,4,5)-triphosphate (PIP3), thus SGK1 is PIP3 independent and remains active even if PIP3 levels are negligible.4 Castel et al. demonstrated that SGK1 could sustain Akt-independent mTORC1 activation under the circumstance of PI3Kα inhibition.39
SGK1 can also induce paclitaxel resistance via modulation of RAN-binding protein 1 (RANBP1) expression. Paclitaxel administration results in an increase in prometaphase-blocked cells in SGK1-silenced RKO colon carcinoma cells, while this cell subset is decreased by SGK1 overexpression at the same paclitaxel concentration. Amato et al. found that Sp1 was phosphorylated by SGK1 at Ser59 and thus enhanced RANBP1 expression.37 SGK1 inhibition significantly reduces nuclear transport by Sp1-dependent reduction of RANBP1 expression.37,38 Cells with RANBP1 interference results in an increased response to taxol-induced apoptosis, compared with those with normal or high RANBP1 levels.107 A preclinical study of ovarian cancer xenografts model indicated that co-treatment of SGK1 inhibitor and paclitaxel significantly reduces the RANBP1 expression and restores paclitaxel sensitivity in paclitaxel-resistant murine.108
The inhibition of SGK1 also increases the sensitivity to radiation treatment in vitro. For instance, the administration of an SGK1 inhibitor significantly increases the apoptosis after radiotherapy in colon carcinoma,60 while the incubation of testosterone albumin conjugates with an SGK1 inhibitor enhances radiation-induced apoptosis in breast cancer.61 SGK1 inhibition also significantly increases the effectiveness of radiation by augmenting phosphatidylserine exposure, caspase 3 activity, and DNA fragmentation but decreasing mitochondrial potential.60 The combined use of SGK1 inhibitor and copper-64 shows an additive effect in tremendously increasing the level of human copper transporter 1 (CTR1), thus bringing a higher bioavailability of copper-64 in glioblastomas.109 Co-treatment with SGK1 inhibitor reveals a potential reducing the cumulative toxicity of ionizing radiations, which represents a novel therapeutic alternative in tumors sensitive to radiation therapy. Although corresponding phenomena have been observed, the detailed mechanism by which SGK1 affects radiosensitivity remains to be clarified.
Clinical potential of SGK1 as a therapeutic target
Continuing research into the function of SGK1 has highlighted the clinical potential of this kinase. Under normal physiological conditions, SGK1 is expressed at low levels in several tissues, and a lack of SGK1 in SGK1−/− mice had no significant effect on survival.110 However, SGK1 is upregulated under stress and certain pathological conditions and is thus regarded as a novel therapeutic target.111
Akt, SGK1, and two other SGK isoforms, SGK2 and SGK3, show extensive overlap in terms of downstream targets and thus compensate for each other in gene ablation or depletion studies.112 The discovery of selective SGK1 inhibitors has helped to identify SGK1-specific downstream targets and has provided invaluable tools for further clinical investigations (Table 2).
Table 2.
Summary of frequently or recently reported SGK1 inhibitors.
| Drug | Structures | Mechanism of action | Advantages and limitations | References |
|---|---|---|---|---|
| GSK650394 |
|
Replaces ATP binding to SGK1 thus inhibits kinase activity of SGK1 | Has been commercialized Inhibitory effect is widely recognized Off-target effects are notable |
Sherk et al.29, Isikbay et al.53, Matschke et al.113 |
| EMD638683 |
|
Replaces ATP binding to SGK1 thus inhibits kinase activity of SGK1 | Has been commercialized Slightly higher selectivity Poor cell permeability |
Towhid et al.60, Ackermann et al.114, Schmid et al.115 |
| SI113 |
|
Replaces ATP binding to SGK1 thus inhibits kinase activity of SGK1 | Higher selectivity inhibiting SGK1 with low effect on AKT Not yet commercialized |
Talarico et al.104, Ortuso et al.116 |
| SGK1-inh |
|
Replaces ATP binding to SGK1 thus inhibits kinase activity of SGK1 | Further research is needed | Halland et al.117 |
| QGY-5-114-A |
|
Replaces ATP binding to SGK1 thus inhibits kinase activity of SGK1 | Further research is needed | Liang et al.118 |
SGK1, serum and glucocorticoid-inducible kinase 1.
The heterocyclic indazole derivative later defined as GSK650394 is the first SGK1 inhibitor.29 Berdel et al. demonstrated that GSK650394 decreased the expression of CD44 and HER2, thus suppressing tumor growth and tumorigenicity, while the combined application of GSK650394 and cisplatin was better than cisplatin alone in squamous cell carcinoma of the head and neck.95 Wang et al. found that SGK1 played an antioxidant role through c-JUN-dependent NRF2 expression and activation in cervical cancer. The combination of GSK650394 and melatonin substantially repressed cervical tumors in vivo.119 Besides, GSK650394 has been commercialized and its inhibitory effect is widely recognized, including tumor suppressor functions in pulmonary and prostate cancer.53,113 However, GSK650394 reveals notable off-target effects, that is it equivalently inhibits SGK2 as well and is only about 30-fold more selective for SGK1 than for other targets such as Akt, Rho-associated protein kinase, and Janus kinase isoforms (JAK1, JAK3).104
The benzohydrazide derivative EMD638683 is another commonly used SGK1 inhibitor with a slightly higher selectivity.114 Low-dose EMD638683 promotes radiation-induced apoptosis of colon tumor cells,60 decreases the viability of rhabdomyosarcoma cells, and enhances the tumor-suppressive effect of doxorubicin.115 However, EMD638683 shows poor cell permeability, and it may also inhibit cAMP-dependent protein kinase, mitogen- and stress-activated protein kinase 1, protein kinase C-related kinase, and other SGK isoforms.114
Schenone et al. synthesized SI113 based on a pyrazolo[3,4-d]pyrimidine scaffold.116 SI113 displays high selectivity for inhibiting SGK1 kinase activity by competing with ATP for the binding domain and is much less effective against Akt1.116 Thus, the short-term systemic toxicity of SI113 is low in preclinical observation. In neurospheres, SI113 induces pro-survival autophagy by reducing the phosphorylation of SGK1 and mTOR activity and increasing the AMPK activity. SI113 and quinacrine, an autophagy inhibitor, show a strong synergistic effect in hindering the tumor growth.120 In colon cancer, hepatocellular carcinoma, and glioblastoma multiforme, SI113 is capable of inhibiting cancer cell malignancy by blocking focal adhesion kinase (FAK), EMT, and cytoskeletal organization.18 The tumor-killing effects of radiotherapy and paclitaxel are potentiated by synergy with SI113.121,122
In addition to the three mentioned previously, new inhibitors, including SGK1-inh and QGY-5-114-A, are ceaselessly being designed and synthesized.117,118 However, their effectiveness in tumor cells remains to be further investigated.
Increasing evidence suggests that SGK1 inhibitors represent a novel tumor therapy approach, extending traditional strategies. On that basis, the structure and function of inhibitors need to be improved, and in vivo pharmacodynamic and pharmacokinetic data are required to conduct clinical trials.
Conclusion
SGK1 is a pleiotropic factor involved in several physiological and pathological processes and has shown important effects on oncology. As an essential Akt-independent mediator of the PI3K/mTOR signaling pathway, SGK1 is transcriptionally and posttranslationally affected by various factors, including hormones, cytokines, and stress response. SGK1 is involved in regulating tumor growth, survival, metastasis, autophagy, immunoregulation, Ca2+ signaling, cancer stem cells, cell cycle, and mediates the therapeutic resistance. SGK1 inhibition is regarded as a potential tumor therapeutic approach, but further pharmacological investigations are needed to advance clinical trials of SGK1-targeted therapies.
Footnotes
Author contributions: All authors contributed to the writing and editing of this manuscript. All authors read and approved the final manuscript.
Conflict of interest statement: The authors declare that there is no conflict of interest.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was supported by grants from the National Natural Science Foundation of China (No. 81772639; 81802475; 81972258; 81974376); Natural Science Foundation of Beijing (No. 7192157); CAMS Innovation Fund for Medical Sciences (CIFMS) (No.2016-I2M-1-001); China Postdoctoral Science Foundation (No.198831); National Key R&D Program of China (2018YFE0118600); Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019XK320001).
Contributor Information
Ruizhe Zhu, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Gang Yang, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Zhe Cao, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Kexin Shen, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Lianfang Zheng, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Jianchun Xiao, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Lei You, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
Taiping Zhang, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing 100730, China.
References
- 1. Lang F, Bohmer C, Palmada M, et al. (Patho)physiological significance of the serum- and glucocorticoid-inducible kinase isoforms. Physiol Rev 2006; 86: 1151–1178. [DOI] [PubMed] [Google Scholar]
- 2. Leroux AE, Schulze JO, Biondi RM. AGC kinases, mechanisms of regulation and innovative drug development. Semin Cancer Biol 2018; 48: 1–17. [DOI] [PubMed] [Google Scholar]
- 3. Loffing J, Flores SY, Staub O. Sgk kinases and their role in epithelial transport. Annu Rev Physiol 2006; 68: 461–490. [DOI] [PubMed] [Google Scholar]
- 4. Pearce LR, Komander D, Alessi DR. The nuts and bolts of AGC protein kinases. Nat Rev Mol Cell Biol 2010; 11: 9–22. [DOI] [PubMed] [Google Scholar]
- 5. Lang F, Stournaras C. Serum and glucocorticoid inducible kinase, metabolic syndrome, inflammation, and tumor growth. Hormones (Athens) 2013; 12: 160–171. [DOI] [PubMed] [Google Scholar]
- 6. Di Cristofano A. SGK1: the dark side of PI3K signaling. Curr Top Dev Biol 2017; 123: 49–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Yan L, Mieulet V, Lamb RF. mTORC2 is the hydrophobic motif kinase for SGK1. Biochem J 2008; 416: e19–e21. [DOI] [PubMed] [Google Scholar]
- 8. You H, Jang Y, You-Ten AI, et al. p53-dependent inhibition of FKHRL1 in response to DNA damage through protein kinase SGK1. Proc Natl Acad Sci USA 2004; 101: 14057–14062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Inoue K, Leng T, Yang T, et al. Role of serum- and glucocorticoid-inducible kinases in stroke. J Neurochem 2016; 138: 354–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Tang C, Zelenak C, Volkl J, et al. Hydration-sensitive gene expression in brain. Cell Physiol Biochem 2011; 27: 757–768. [DOI] [PubMed] [Google Scholar]
- 11. Fagerli UM, Ullrich K, Stuhmer T, et al. Serum/glucocorticoid-regulated kinase 1 (SGK1) is a prominent target gene of the transcriptional response to cytokines in multiple myeloma and supports the growth of myeloma cells. Oncogene 2011; 30: 3198–3206. [DOI] [PubMed] [Google Scholar]
- 12. Mizuno H, Nishida E. The ERK MAP kinase pathway mediates induction of SGK (serum- and glucocorticoid-inducible kinase) by growth factors. Genes Cells 2001; 6: 261–268. [DOI] [PubMed] [Google Scholar]
- 13. Pasham V, Rotte A, Gu S, et al. Upregulation of intestinal NHE3 following saline ingestion. Kidney Blood Press Res 2013; 37: 48–57. [DOI] [PubMed] [Google Scholar]
- 14. Lang F, Shumilina E. Regulation of ion channels by the serum- and glucocorticoid-inducible kinase SGK1. FASEB J 2013; 27: 3–12. [DOI] [PubMed] [Google Scholar]
- 15. Lou Y, Hu M, Mao L, et al. Involvement of serum glucocorticoid-regulated kinase 1 in reproductive success. FASEB J 2017; 31: 447–456. [DOI] [PubMed] [Google Scholar]
- 16. Burgon J, Robertson AL, Sadiku P, et al. Serum and glucocorticoid-regulated kinase 1 regulates neutrophil clearance during inflammation resolution. J Immunol 2014; 192: 1796–1805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Lang F, Gorlach A, Vallon V. Targeting SGK1 in diabetes. Expert Opin Ther Targets 2009; 13: 1303–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Abbruzzese C, Matteoni S, Persico M, et al. The small molecule SI113 hinders epithelial-to-mesenchymal transition and subverts cytoskeletal organization in human cancer cells. J Cell Physiol 2019; 234: 22529–22542. [DOI] [PubMed] [Google Scholar]
- 19. Abbruzzese C, Catalogna G, Gallo E, et al. The small molecule SI113 synergizes with mitotic spindle poisons in arresting the growth of human glioblastoma multiforme. Oncotarget 2017; 8: 110743–110755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Kulkarni S, Goel-Bhattacharya S, Sengupta S, et al. A large-scale RNAi screen identifies SGK1 as a key survival kinase for GBM stem cells. Mol Cancer Res 2018; 16: 103–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Godbole M, Togar T, Patel K, et al. Up-regulation of the kinase gene SGK1 by progesterone activates the AP-1-NDRG1 axis in both PR-positive and -negative breast cancer cells. J Biol Chem 2018; 293: 19263–19276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Hasna J, Hague F, Rodat-Despoix L, et al. Orai3 calcium channel and resistance to chemotherapy in breast cancer cells: the p53 connection. Cell Death Differ 2018; 25: 691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Sommer EM, Dry H, Cross D, et al. Elevated SGK1 predicts resistance of breast cancer cells to Akt inhibitors. Biochem J 2013; 452: 499–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Hall BA, Kim TY, Skor MN, et al. Serum and glucocorticoid-regulated kinase 1 (SGK1) activation in breast cancer: requirement for mTORC1 activity associates with ER-alpha expression. Breast Cancer Res Treat 2012; 135: 469–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Salis O, Okuyucu A, Bedir A, et al. Antimetastatic effect of fluvastatin on breast and hepatocellular carcinoma cells in relation to SGK1 and NDRG1 genes. Tumour Biol 2016; 37: 3017–3024. [DOI] [PubMed] [Google Scholar]
- 26. Lang F, Perrotti N, Stournaras C. Colorectal carcinoma cells—regulation of survival and growth by SGK1. Int J Biochem Cell Biol 2010; 42: 1571–1575. [DOI] [PubMed] [Google Scholar]
- 27. Zhu J, Zhang R, Yang D, et al. Knockdown of long non-coding RNA XIST inhibited doxorubicin resistance in colorectal cancer by upregulation of miR-124 and downregulation of SGK1. Cell Physiol Biochem 2018; 51: 113–128. [DOI] [PubMed] [Google Scholar]
- 28. Lee LYW, Woolley C, Starkey T, et al. Serum- and glucocorticoid-induced kinase Sgk1 directly promotes the differentiation of colorectal cancer cells and restrains metastasis. Clin Cancer Res 2019; 25: 629–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Sherk AB, Frigo DE, Schnackenberg CG, et al. Development of a small-molecule serum- and glucocorticoid-regulated kinase-1 antagonist and its evaluation as a prostate cancer therapeutic. Cancer Res 2008; 68: 7475–7483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Shanmugam I, Cheng G, Terranova PF, et al. Serum/glucocorticoid-induced protein kinase-1 facilitates androgen receptor-dependent cell survival. Cell Death Differ 2007; 14: 2085–2094. [DOI] [PubMed] [Google Scholar]
- 31. Liu W, Wang X, Liu Z, et al. SGK1 inhibition induces autophagy-dependent apoptosis via the mTOR-Foxo3a pathway. Br J Cancer 2017; 117: 1139–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Orlacchio A, Ranieri M, Brave M, et al. SGK1 is a critical component of an AKT-independent pathway essential for PI3K-mediated tumor development and maintenance. Cancer Res 2017; 77: 6914–6926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Melhem A, Yamada SD, Fleming GF, et al. Administration of glucocorticoids to ovarian cancer patients is associated with expression of the anti-apoptotic genes SGK1 and MKP1/DUSP1 in ovarian tissues. Clin Cancer Res 2009; 15: 3196–3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Tang Z, Shen Q, Xie H, et al. Serum and glucocorticoid-regulated kinase 1 (SGK1) is a predictor of poor prognosis in non-small cell lung cancer, and its dynamic pattern following treatment with SGK1 inhibitor and gamma-ray irradiation was elucidated. Oncol Rep 2018; 39: 1505–1515. [DOI] [PubMed] [Google Scholar]
- 35. Park KC, Menezes SV, Kalinowski DS, et al. Identification of differential phosphorylation and sub-cellular localization of the metastasis suppressor, NDRG1. Biochim Biophys Acta Mol Basis Dis 2018; 1864: 2644–2663. [DOI] [PubMed] [Google Scholar]
- 36. Weiler M, Blaes J, Pusch S, et al. mTOR target NDRG1 confers MGMT-dependent resistance to alkylating chemotherapy. Proc Natl Acad Sci USA 2014; 111: 409–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Amato R, Scumaci D, D’Antona L, et al. Sgk1 enhances RANBP1 transcript levels and decreases taxol sensitivity in RKO colon carcinoma cells. Oncogene 2013; 32: 4572–4578. [DOI] [PubMed] [Google Scholar]
- 38. Dattilo V, D’Antona L, Talarico C, et al. SGK1 affects RAN/RANBP1/RANGAP1 via SP1 to play a critical role in pre-miRNA nuclear export: a new route of epigenomic regulation. Sci Rep 2017; 7: 45361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Castel P, Ellis H, Bago R, et al. PDK1-SGK1 signaling sustains AKT-independent mTORC1 activation and confers resistance to PI3Kα inhibition. Cancer Cell 2016; 30: 229–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Kim MJ, Chae JS, Kim KJ, et al. Negative regulation of SEK1 signaling by serum- and glucocorticoid-inducible protein kinase 1. EMBO J 2007; 26: 3075–3085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Amato R, D’Antona L, Porciatti G, et al. Sgk1 activates MDM2-dependent p53 degradation and affects cell proliferation, survival, and differentiation. J Mol Med (Berl) 2009; 87: 1221–1239. [DOI] [PubMed] [Google Scholar]
- 42. Shelly C, Herrera R. Activation of SGK1 by HGF, Rac1 and integrin-mediated cell adhesion in MDCK cells: PI-3K-dependent and -independent pathways. J Cell Sci 2002; 115: 1985–1993. [DOI] [PubMed] [Google Scholar]
- 43. Brunet A, Park J, Tran H, et al. Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol 2001; 21: 952–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Wu C, Yosef N, Thalhamer T, et al. Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature 2013; 496: 513–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Wu C, Chen Z, Xiao S, et al. SGK1 governs the reciprocal development of Th17 and regulatory T cells. Cell Rep 2018; 22: 653–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Heikamp EB, Patel CH, Collins S, et al. The AGC kinase SGK1 regulates TH1 and TH2 differentiation downstream of the mTORC2 complex. Nat Immunol 2014; 15: 457–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Norton M, Screaton RA. SGK1: master and commander of the fate of helper T cells. Nat Immunol 2014; 15: 411–413. [DOI] [PubMed] [Google Scholar]
- 48. Snyder PM. Down-regulating destruction: phosphorylation regulates the E3 ubiquitin ligase Nedd4-2. Sci Signal 2009; 2: pe41. [DOI] [PubMed] [Google Scholar]
- 49. Zhang L, Cui R, Cheng X, et al. Antiapoptotic effect of serum and glucocorticoid-inducible protein kinase is mediated by novel mechanism activating IκB kinase. Cancer Res 2005; 65: 457–464. [PubMed] [Google Scholar]
- 50. Tai DJ, Su CC, Ma YL, et al. SGK1 phosphorylation of IκB kinase α and p300 up-regulates NF-κB activity and increases N-methyl-D-aspartate receptor NR2A and NR2B expression. J Biol Chem 2009; 284: 4073–4089. [DOI] [PubMed] [Google Scholar]
- 51. Amato R, Menniti M, Agosti V, et al. IL-2 signals through Sgk1 and inhibits proliferation and apoptosis in kidney cancer cells. J Mol Med (Berl) 2007; 85: 707–721. [DOI] [PubMed] [Google Scholar]
- 52. Shaffer PL, Jivan A, Dollins DE, et al. Structural basis of androgen receptor binding to selective androgen response elements. Proc Natl Acad Sci USA 2004; 101: 4758–4763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Isikbay M, Otto K, Kregel S, et al. Glucocorticoid receptor activity contributes to resistance to androgen-targeted therapy in prostate cancer. Horm Cancer 2014; 5: 72–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Skor MN, Wonder EL, Kocherginsky M, et al. Glucocorticoid receptor antagonism as a novel therapy for triple-negative breast cancer. Clin Cancer Res 2013; 19: 6163–6172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Chai D, Li K, Du H, et al. β2-microglobulin has a different regulatory molecular mechanism between ER+ and ER− breast cancer with HER2−. BMC Cancer 2019; 19: 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Conza D, Mirra P, Cali G, et al. The SGK1 inhibitor SI113 induces autophagy, apoptosis, and endoplasmic reticulum stress in endometrial cancer cells. J Cell Physiol 2017; 232: 3735–3743. [DOI] [PubMed] [Google Scholar]
- 57. Yemelyanov A, Bhalla P, Yang X, et al. Differential targeting of androgen and glucocorticoid receptors induces ER stress and apoptosis in prostate cancer cells: a novel therapeutic modality. Cell Cycle 2012; 11: 395–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Fruman DA, Chiu H, Hopkins BD, et al. The PI3K pathway in human disease. Cell 2017; 170: 605–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Lang F, Pelzl L, Hauser S, et al. To die or not to die SGK1-sensitive ORAI/STIM in cell survival. Cell Calcium 2018; 74: 29–34. [DOI] [PubMed] [Google Scholar]
- 60. Towhid ST, Liu GL, Ackermann TF, et al. Inhibition of colonic tumor growth by the selective SGK inhibitor EMD638683. Cell Physiol Biochem 2013; 32: 838–848. [DOI] [PubMed] [Google Scholar]
- 61. Liu G, Honisch S, Liu G, et al. Inhibition of SGK1 enhances mAR-induced apoptosis in MCF-7 breast cancer cells. Cancer Biol Ther 2015; 16: 52–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Schmid E, Xuan NT, Zahir N, et al. Serum- and glucocorticoid-inducible kinase 1 sensitive NF-κB signaling in dendritic cells. Cell Physiol Biochem 2014; 34: 943–954. [DOI] [PubMed] [Google Scholar]
- 63. Mason JA, Davison-Versagli CA, Leliaert AK, et al. Oncogenic Ras differentially regulates metabolism and anoikis in extracellular matrix-detached cells. Cell Death Differ 2016; 23: 1271–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Greenawalt EJ, Edmonds MD, Jain N, et al. Targeting of SGK1 by miR-576-3p inhibits lung adenocarcinoma migration and invasion. Mol Cancer Res 2019; 17: 289–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Liu W, Wang X, Wang Y, et al. SGK1 inhibition-induced autophagy impairs prostate cancer metastasis by reversing EMT. J Exp Clin Cancer Res 2018; 37: 73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Schmidt EM, Gu S, Anagnostopoulou V, et al. Serum- and glucocorticoid-dependent kinase-1-induced cell migration is dependent on vinculin and regulated by the membrane androgen receptor. FEBS J 2012; 279: 1231–1242. [DOI] [PubMed] [Google Scholar]
- 67. Sui X, Chen R, Wang Z, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 2013; 4: e838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Talarico C, Dattilo V, D’Antona L, et al. SI113, a SGK1 inhibitor, potentiates the effects of radiotherapy, modulates the response to oxidative stress and induces cytotoxic autophagy in human glioblastoma multiforme cells. Oncotarget 2016; 7: 15868–15884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kouroku Y, Fujita E, Tanida I, et al. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ 2007; 14: 230–239. [DOI] [PubMed] [Google Scholar]
- 70. Nishimura T, Iwakabe K, Sekimoto M, et al. Distinct role of antigen-specific T helper type 1 (Th1) and Th2 cells in tumor eradication in vivo. J Exp Med 1999; 190: 617–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Knutson KL, Disis ML. Tumor antigen-specific T helper cells in cancer immunity and immunotherapy. Cancer Immunol Immunother 2005; 54: 721–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Narendra BL, Reddy KE, Shantikumar S, et al. Immune system: a double-edged sword in cancer. Inflamm Res 2013; 62: 823–834. [DOI] [PubMed] [Google Scholar]
- 73. Asadzadeh Z, Mohammadi H, Safarzadeh E, et al. The paradox of Th17 cell functions in tumor immunity. Cell Immunol 2017; 322: 15–25. [DOI] [PubMed] [Google Scholar]
- 74. Aggarwal S, Ghilardi N, Xie MH, et al. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J Biol Chem 2003; 278: 1910–1914. [DOI] [PubMed] [Google Scholar]
- 75. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res 2017; 27: 109–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol 2014; 27: 1–7. [DOI] [PubMed] [Google Scholar]
- 77. Joshi NS, Akama-Garren EH, Lu Y, et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity 2015; 43: 579–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Wolf D, Sopper S, Pircher A, et al. Treg(s) in cancer: friends or foe? J Cell Physiol 2015; 230: 2598–2605. [DOI] [PubMed] [Google Scholar]
- 79. Spagnuolo R, Dattilo V, D’Antona L, et al. Deregulation of SGK1 in ulcerative colitis: a paradoxical relationship between immune cells and colonic epithelial cells. Inflamm Bowel Dis 2018; 24: 1967–1977. [DOI] [PubMed] [Google Scholar]
- 80. Parkash J, Asotra K. Calcium wave signaling in cancer cells. Life Sci 2010; 87: 587–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Putney JW., Jr. Capacitative calcium entry: sensing the calcium stores. J Cell Biol 2005; 169: 381–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Parekh AB, Putney JW., Jr. Store-operated calcium channels. Physiol Rev 2005; 85: 757–810. [DOI] [PubMed] [Google Scholar]
- 83. Derler I, Jardin I, Romanin C. Molecular mechanisms of STIM/Orai communication. Am J Physiol Cell Physiol 2016; 310: C643–C662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Lang F, Eylenstein A, Shumilina E. Regulation of Orai1/STIM1 by the kinases SGK1 and AMPK. Cell Calcium 2012; 52: 347–354. [DOI] [PubMed] [Google Scholar]
- 85. Qu B, Al-Ansary D, Kummerow C, et al. ORAI-mediated calcium influx in T cell proliferation, apoptosis and tolerance. Cell Calcium 2011; 50: 261–269. [DOI] [PubMed] [Google Scholar]
- 86. Prevarskaya N, Skryma R, Shuba Y. Calcium in tumour metastasis: new roles for known actors. Nat Rev Cancer 2011; 11: 609–618. [DOI] [PubMed] [Google Scholar]
- 87. Roderick HL, Cook SJ. Ca2+ signalling checkpoints in cancer: remodelling Ca2+ for cancer cell proliferation and survival. Nat Rev Cancer 2008; 8: 361–375. [DOI] [PubMed] [Google Scholar]
- 88. Prevarskaya N, Ouadid-Ahidouch H, Skryma R, et al. Remodelling of Ca2+ transport in cancer: how it contributes to cancer hallmarks? Philos Trans R Soc Lond B Biol Sci 2014; 369: 20130097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Eylenstein A, Gehring EM, Heise N, et al. Stimulation of Ca2+-channel Orai1/STIM1 by serum- and glucocorticoid-inducible kinase 1 (SGK1). FASEB J 2011; 25: 2012–2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Sukkar B, Hauser S, Pelzl L, et al. Inhibition of lithium sensitive Orai1/STIM1 expression and store operated Ca2+ entry in chorea-acanthocytosis neurons by NF-κB inhibitor Wogonin. Cell Physiol Biochem 2018; 51: 278–289. [DOI] [PubMed] [Google Scholar]
- 91. Putzer BM, Solanki M, Herchenroder O. Advances in cancer stem cell targeting: how to strike the evil at its root. Adv Drug Deliv Rev 2017; 120: 89–107. [DOI] [PubMed] [Google Scholar]
- 92. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med 2017; 23: 1124–1134. [DOI] [PubMed] [Google Scholar]
- 93. Francipane MG, Lagasse E. Selective targeting of human colon cancer stem-like cells by the mTOR inhibitor Torin-1. Oncotarget 2013; 4: 1948–1962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Chen F, Chen X, Ren Y, et al. Radiation-induced glucocorticoid receptor promotes CD44+ prostate cancer stem cell growth through activation of SGK1-Wnt/β-catenin signaling. J Mol Med (Berl) 2019; 97: 1169–1182. [DOI] [PubMed] [Google Scholar]
- 95. Berdel HO, Yin H, Liu JY, et al. Targeting serum glucocorticoid-regulated kinase-1 in squamous cell carcinoma of the head and neck: a novel modality of local control. PLoS One 2014; 9: e113795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Mo JS, Ann EJ, Yoon JH, et al. Serum- and glucocorticoid-inducible kinase 1 (SGK1) controls Notch1 signaling by downregulation of protein stability through Fbw7 ubiquitin ligase. J Cell Sci 2011; 124: 100–112. [DOI] [PubMed] [Google Scholar]
- 97. Anacker C, Cattaneo A, Musaelyan K, et al. Role for the kinase SGK1 in stress, depression, and glucocorticoid effects on hippocampal neurogenesis. Proc Natl Acad Sci USA 2013; 110: 8708–8713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Ahmad G, Amiji MM. Cancer stem cell-targeted therapeutics and delivery strategies. Expert Opin Drug Deliv 2017; 14: 997–1008. [DOI] [PubMed] [Google Scholar]
- 99. Chu I, Sun J, Arnaout A, et al. p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2. Cell 2007; 128: 281–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Alkarain A, Jordan R, Slingerland J. p27 deregulation in breast cancer: prognostic significance and implications for therapy. J Mammary Gland Biol Neoplasia 2004; 9: 67–80. [DOI] [PubMed] [Google Scholar]
- 101. Buse P, Tran SH, Luther E, et al. Cell cycle and hormonal control of nuclear-cytoplasmic localization of the serum- and glucocorticoid-inducible protein kinase, Sgk, in mammary tumor cells. A novel convergence point of anti-proliferative and proliferative cell signaling pathways. J Biol Chem 1999; 274: 7253–7263. [DOI] [PubMed] [Google Scholar]
- 102. Hong F, Larrea MD, Doughty C, et al. mTOR-raptor binds and activates SGK1 to regulate p27 phosphorylation. Mol Cell 2008; 30: 701–711. [DOI] [PubMed] [Google Scholar]
- 103. Feng Z, Liu L, Zhang C, et al. Chronic restraint stress attenuates p53 function and promotes tumorigenesis. Proc Natl Acad Sci USA 2012; 109: 7013–7018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Talarico C, Dattilo V, D’Antona L, et al. SGK1, the new player in the game of resistance: chemo-radio molecular target and strategy for inhibition. Cell Physiol Biochem 2016; 39: 1863–1876. [DOI] [PubMed] [Google Scholar]
- 105. Chua MS, Sun H, Cheung ST, et al. Overexpression of NDRG1 is an indicator of poor prognosis in hepatocellular carcinoma. Mod Pathol 2007; 20: 76–83. [DOI] [PubMed] [Google Scholar]
- 106. Murray JT, Campbell DG, Morrice N, et al. Exploitation of KESTREL to identify NDRG family members as physiological substrates for SGK1 and GSK3. Biochem J 2004; 384: 477–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Rensen WM, Roscioli E, Tedeschi A, et al. RanBP1 downregulation sensitizes cancer cells to taxol in a caspase-3-dependent manner. Oncogene 2009; 28: 1748–1758. [DOI] [PubMed] [Google Scholar]
- 108. D’Antona L, Dattilo V, Catalogna G, et al. In preclinical model of ovarian cancer, the SGK1 inhibitor SI113 counteracts the development of paclitaxel resistance and restores drug sensitivity. Transl Oncol 2019; 12: 1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Catalogna G, Talarico C, Dattilo V, et al. The SGK1 kinase inhibitor SI113 sensitizes theranostic effects of the 64CuCl2 in human glioblastoma multiforme cells. Cell Physiol Biochem 2017; 43: 108–119. [DOI] [PubMed] [Google Scholar]
- 110. Wulff P, Vallon V, Huang DY, et al. Impaired renal Na+ retention in the sgk1-knockout mouse. J Clin Invest 2002; 110: 1263–1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Lang F, Gorlach A. Heterocyclic indazole derivatives as SGK1 inhibitors, WO2008138448. Expert Opin Ther Pat 2010; 20: 129–135. [DOI] [PubMed] [Google Scholar]
- 112. Bruhn MA, Pearson RB, Hannan RD, et al. AKT-independent PI3-K signaling in cancer—emerging role for SGK3. Cancer Manag Res 2013; 5: 281–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Matschke J, Wiebeck E, Hurst S, et al. Role of SGK1 for fatty acid uptake, cell survival and radioresistance of NCI-H460 lung cancer cells exposed to acute or chronic cycling severe hypoxia. Radiat Oncol 2016; 11: 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Ackermann TF, Boini KM, Beier N, et al. EMD638683, a novel SGK inhibitor with antihypertensive potency. Cell Physiol Biochem 2011; 28: 137–146. [DOI] [PubMed] [Google Scholar]
- 115. Schmid E, Stagno MJ, Yan J, et al. Serum and glucocorticoid inducible kinase 1-sensitive survival, proliferation and migration of rhabdomyosarcoma cells. Cell Physiol Biochem 2017; 43: 1301–1308. [DOI] [PubMed] [Google Scholar]
- 116. Ortuso F, Amato R, Artese A, et al. In silico identification and biological evaluation of novel selective serum/glucocorticoid-inducible kinase 1 inhibitors based on the pyrazolo-pyrimidine scaffold. J Chem Inf Model 2014; 54: 1828–1832. [DOI] [PubMed] [Google Scholar]
- 117. Halland N, Schmidt F, Weiss T, et al. Discovery of N-[4-(1H-pyrazolo[3,4-b]pyrazin-6-yl)-phenyl]-sulfonamides as highly active and selective SGK1 inhibitors. ACS Med Chem Lett 2015; 6: 73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Liang X, Lan C, Zhou J, et al. Development of a new analog of SGK1 inhibitor and its evaluation as a therapeutic molecule of colorectal cancer. J Cancer 2017; 8: 2256–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Wang M, Xue Y, Shen L, et al. Inhibition of SGK1 confers vulnerability to redox dysregulation in cervical cancer. Redox Biol 2019; 24: 101225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Matteoni S, Abbruzzese C, Matarrese P, et al. The kinase inhibitor SI113 induces autophagy and synergizes with quinacrine in hindering the growth of human glioblastoma multiforme cells. J Exp Clin Cancer Res 2019; 38: 202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Talarico C, D’Antona L, Scumaci D, et al. Preclinical model in HCC: the SGK1 kinase inhibitor SI113 blocks tumor progression in vitro and in vivo and synergizes with radiotherapy. Oncotarget 2015; 6: 37511–37525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. D’Antona L, Amato R, Talarico C, et al. SI113, a specific inhibitor of the Sgk1 kinase activity that counteracts cancer cell proliferation. Cell Physiol Biochem 2015; 35: 2006–2018. [DOI] [PubMed] [Google Scholar]


