Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 1.
Published in final edited form as: Cell Signal. 2020 Apr 20;72:109639. doi: 10.1016/j.cellsig.2020.109639

STAT Signaling in Polycystic Kidney Disease

Sebastian Strubl 1,3,1, Jacob A Torres 1,1, Alison K Spindt 1, Hannah Pellegrini 1, Max C Liebau 2,3, Thomas Weimbs 1,*
PMCID: PMC7269822  NIHMSID: NIHMS1588609  PMID: 32325185

Abstract

The most common form of polycystic kidney disease (PKD) in humans is caused by mutations in the PKD1 gene coding for polycystin1 (PC1). Among the many identified or proposed functions of PC1 is its ability to regulate the activity of transcription factors of the STAT family. Most STAT proteins that have been investigated were found to be aberrantly activated in kidneys in PKD, and some have been shown to be drivers of disease progression. In this review, we focus on the role of signal transducer and activator of transcription (STAT) signaling pathways in various renal cell types in healthy kidneys as compared to polycystic kidneys, on the mechanisms of STAT regulation by PC1 and other factors, and on the possibility to target STAT signaling for PKD therapy.

Introduction

Mutations in numerous genes can lead to polycystic kidney disease (PKD) in humans and animal models. The most common human form, autosomal-dominant PKD (ADPKD), is caused by mutations in the PKD1 orPKD2 genes, and typically leads to slow progression towards end-stage renal failure during adulthood. During disease progression, aberrant proliferation of tubule epithelial cells (TECs) leads to thousands of microscopic and macroscopic cysts that develop in both kidneys. This causes substantial organ enlargement, fibrosis and destruction of the normal renal parenchyma (Bergmann et al., 2018; Cornec-Le Gall et al., 2019; Müller and Benzing, 2018). Polycystin1 (PC1) and polycystin2 (PC2), the products of the PKD1 or PKD2 genes, are the subunits of an apparent cation channel with a poorly understood purpose. It is still controversial, which cations are primarily conducted by this channel, if any (Douguet et al., 2019; Padovano and Caplan, 2018). In addition to their ion channel function, both proteins - either alone or together - also regulate a bewildering number of other signaling functions. This includes the regulation of heterotrimeric G-proteins, mTOR signaling, signal transducer and activator of transcription (STAT) signaling and many more (Bergmann et al., 2018; Ong and Harris, 2015; Torres and Harris, 2019).

The only FDA-approved therapeutic for ADPKD is the vasopressin receptor 2 antagonist tolvaptan which is available to a limited fraction of patients. It’s use is further complicated due to significant side effects and potential toxicities (Chebib and Torres, 2018), poor cost-effectiveness (Erickson et al., 2013) and questions about its efficacy (Anderson, 2020). Tolvaptan does not appear to directly affect a mechanism regulated by the polycystins. It is thought to act via inhibition of cAMP signaling in cyst-lining cells but may also act more indirectly, e.g. by increasing water consumption in patients (Gastel and Torres, 2017).

Much effort has been spent on identifying signaling mechanisms that are directly regulated by the polycystins in an effort to devise strategies for pharmacological intervention to slow disease progression in ADPKD (Torres and Harris, 2019; Weimbs et al., 2018). This review will focus on the role of STAT transcription factors that have been found to be regulated by PC1, are frequently found to be activated in various cell types in ADPKD and PKD animal models, and appear to be drivers of disease progression.

STAT transcription factors

STAT proteins are a family of seven (in mammals) sequence-specific transcription factors that most typically act as signal transducers between growth factor receptors on the plasma membrane and specific genes in the nucleus. They reside in the cytoplasm and oscillate between antiparallel and parallel dimer conformation until the parallel dimer conformation is stabilized by tyrosine phosphorylation (Nast et al., 2019). Canonical STAT activation usually occurs through a series of tyrosine phosphorylations by members of the Janus kinase (JAK) family that are non-covalently bound to those specific receptors (O’Shea et al., 2015). Ligand binding brings the JAK proteins in close proximity allowing for transphosphorylation which releases their intrinsic catalytic activity (O’Shea et al., 2013). Following a phosphorylation series of receptor tails, STATs are recruited through the interaction of their Src-homology-2 (SH2) domain with phosphorylation sites (Liongue and Ward, 2013). Phosphorylation of a single tyrosine residue near the C-terminus of the STAT protein results in release from the receptor and homo- or heterodimerization in parallel arrangement (Zhong et al., 2005) allowing the transcription factors to subsequently enter the nucleus. The translocation event is followed by binding of STATs to specific DNA sequences together with transcriptional cofactors to regulate gene expression (Figure 1 and 2) (Rawlings et al., 2004).

Figure 1. Model of STAT1 and STAT3 signaling in the healthy kidney.

Figure 1.

Binding of certain cytokines and growth factors to their receptors leads to activation of JAKs which in turn phosphorylate intracellular receptor tyrosine residues. STAT1 and STAT3 molecules bind to these residues, which leads to their phosphorylation by JAKs. After dissociation from the receptor, phosphorylated STATs dimerize and translocate to the nucleus resulting in modified target gene expression. Beside JAK, the tyrosine kinase receptor c-Met and the non-receptor associated kinase Src are also capable of STAT3 phosphorylation. STAT1 signaling in the kidney was shown to occur in TECs regulating the macrophage phenotype and, thus, inflammation and fibrosis in the kidney. STAT3 signaling exists in renal TECs and myofibroblasts and seems to mediate a cross talk between these cell types, which regulates myofibroblast attraction and activation.

Figure 2. Model of STAT5 and STAT6 signaling in the healthy kidney.

Figure 2.

Cytokine and growth factor mediated JAK kinase activation can also lead to STAT5 and STAT6 phosphorylation in renal TECs and macrophages. STAT5 and STAT6 dimers translocate to the nucleus and regulate target gene expression. Additionally, the non-receptor associated Src kinase was shown to activate STAT5b. STAT5 signaling seems to mediate inflammation and tissue repair mechanisms and is involved in alternative M2 macrophage polarization upon GM-CSF secretion of damaged renal TECs. STAT6 signaling in the kidney might also be involved in fibrosis and tissue repair mechanisms. Upon IL-4 and IL-13 activation STAT6 signaling was described to primarily drive M2 polarization of macrophages, myofibroblast transformation and accumulation and contributes to fibrosis by regulating the production of ECM proteins.

Besides JAK-associated receptors, several receptors with intrinsic tyrosine kinase activity, such as the epidermal growth factor receptor (EGFR), the platelet-derived growth factor receptor (PDGFR) and the hepatocyte growth factor receptor (c-Met), are able to activate STATs directly (Leaman et al., 1996; Olayioye et al., 1999; Vignais et al., 1996). Non-receptor tyrosine kinases such as Src (Wang et al., 2000) are also able to phosphorylate and activate STAT proteins (Figure 1 and 2)(Silva, 2004).

The three major classes of negative regulators of the JAK/STAT pathway are members of the SOCS (suppressor of cytokine signaling), PIAS (protein inhibitors of activated stats) and PTP (protein tyrosine phosphatases) protein families. SOCS proteins are part of a negative feedback loop as STATs stimulate SOCS gene transcription which then bind to phosphorylated JAK proteins or phosphorylated tyrosine residues on activated cytokine receptors and suppress their activity (Yoshimura et al., 2007). PIAS proteins bind directly to activated STAT dimers thereby preventing STAT-mediated transcription whereas PTPs antagonize signaling by dephosphorylating activated JAKs, phosphorylated receptors or the STATs themselves (O’Sullivan et al., 2007; Shuai, 2006). Besides tyrosine-phosphorylation, several other post-translational modifications critically affect STAT-driven transcription either promoting or suppressing STAT activity. To date, lysine acetylation of STATs has been identified for five out of seven STAT family members (STAT1, STAT2, STAT3, STAT5, STAT6) (Zhuang, 2013) while STAT1, STAT3, STAT4 and STAT5A/B have all been shown to undergo a second phosphorylation event on serine (Ferreli et al., 2017; Villarino et al., 2017). A conserved arginine residue in STAT1, STAT3 and STAT6 has also been shown to be methylated which prevents PIAS effectors from binding to STATs and thereby increases transcription efficiency (Heppler and Frank, 2017). STAT5-dependent transcription was found to be enhanced by the cooperative action of two methyltransferases (Kleinschmidt et al., 2008). STAT1 was also identified to contain five sumoylation sites while one and two sumoylation sites and were identified for STAT3 and STAT5 respectively (Heppler and Frank, 2017; Van Nguyen et al., 2012).

Another layer of complexity of STAT-driven gene expression is due to unphosphorylated STATs that can mediate gene expression in a manner distinct from STATs activated by tyrosine phosphorylation (Cheon et al., 2011). STAT1 and STAT3 genes are themselves targets of activated STAT1/STAT3 proteins. The STAT-driven gene expression of their own genes results in large amounts of unphosphorylated STAT1/STAT3 in the cytoplasm, which are latent unless they collaborate with other proteins to drive gene expression. Unphosphorylated STAT1 (U-STAT1) together with IRF1 results in expression of low molecular mass peptide (LMP) 2 gene (Cheon and Stark, 2009). U-STAT3 has been found to activate a subset of κB-dependent genes by associating with unphosphorylated NF-κB while U-STAT6 can cooperate with p300 to contribute to cyclooxygenase-2 expression (Cui et al., 2007; Yang and Stark, 2008). Further, through the absence of thrombopoietin, uSTAT5 represses megakaryocytic differentiation by colocalizing with CTCF in the nucleus (Park et al., 2016).

Recently, U-STAT2 has been shown to be involved in a novel mechanism leading to STAT3 activation. By interacting with IRF-9 and the p65 subunit of NF-κB, U-STAT2 can lead to increased Interleukin (IL)-6 secretion eventually leading to STAT3 activation (Nan et al., 2018).

Members of the STAT protein family display distinctive biological functions and STAT-STAT interactions (Table 1). To date, all STATs except STAT2 have been implicated in PKD and those will be further discussed below.

Table 1:

STAT functions, activators and interactions

STAT family member Biological function Selection of activators Heterodimerization partners References
STAT1 Growth arrest and apoptosis, immune response Type I IFN
Type II IFN
IL-6,−10,−27,−35
EGF/PDGF/HGF/CSF-1/angiotensin/growth hormone
STAT2
STAT3
STAT4
(Zhang et al., 2017b)
(Ihle, 1996)
STAT2 Antiviral protection IFNα/IFNβ STAT1
STAT6
(Perry et al., 2011)
STAT3 Often oncogenic; protects from apoptosis IL-2,−5,−6,−10,−23,−27
MCSF
GCSF
STAT1
STAT5
(Yu et al., 2009)
STAT4 TH1 differentiation IL-12,−23,−35 STAT1
STAT3
(Wurster et al., 2000)
STAT5A Lymphocyte development
Mammary cell differentiation
IL-2,−7,−15 STAT3
STAT5b
(Creamer et al., 2010)
(Haricharan and Li, 2014)
STAT5B Regulation of sexual dimorphism in liver IL-2,−7,−15 STAT3
STAT5a
(Udy et al., 1997)
STAT6 Lymphocyte development; CD4+ T-cell polarization into Th2 cells; macrophage M2 polarization
IgE class switching
Type I IFN
IL-3,−4,−13
STAT2 (Wurster et al., 2000)
(Takeda et al., 1996)
(Shimoda et al., 1996)

What is the role of STATs in the healthy kidney and in renal pathologies besides PKD?

STAT signaling appears to be generally turned off in normal, adult kidneys, at least as evidenced by the lack of tyrosine-phosphorylation of STATs. Nevertheless, most STATs are highly expressed in normal kidneys suggesting that STAT signaling pathways are “on standby” and can be rapidly activated in response to various stimuli. Renal STAT activation has been most frequently reported in response to renal injury and other renal pathologies and can occur very rapidly as discussed below for each individual STAT protein.

STAT1.

STAT1 is activated in embryonic kidneys by phosphorylation on Y701 and S727 while healthy adult kidneys lack active STAT1 indicating a certain role in kidney development even if Stat1 null mice do not display any renal defects in embryogenesis (Durbin et al., 1996; Meraz et al., 1996; Tao et al., 2018; Wang et al., 2010). In contrast, Socs1- deficient mice exhibit delayed renal tubule organization and unilateral polycystic kidneys. However, a concomitant deletion of Interferon γ (IFNγ) is required to prevent early non-renal death making it difficult to define the specific role of STAT1 in renal development (Metcalf et al., 2002). Beside this, STAT1 is well known in mediating inflammation in several renal diseases like diabetic nephropathy (DN) and lupus nephritis (LN) (Dong et al., 2007; Shi et al., 2010; Zhou et al., 2013). Oxidative stress as a major cause of chronic inflammation and tubular hypertrophy in the kidney seems to mediate its effects by JAK2/STAT1/STAT3 signaling in TECs. Using high glucose treatment as oxidative trigger in TEC culture (LLC-PK1) JAK2/STAT1/STAT3 signaling was shown to be highly activated leading to cell hypertrophy and extracellular matrix (ECM) protein accumulation. Treatment with antioxidants like taurin or N-acetylcysteine blocked JAK2/STAT1 activation and reversed these effects pointing to JAK2/STAT1 as a redox sensitive pathway in TECs (Huang et al., 2007). Furthermore, STAT1 activation contributes to chemokine expression of TECs and macrophage regulation (Figure 1). STAT1 depleted TECs revealed significantly decreased expression of CXCl9, IP-10 and MCP-1 upon acute inflammatory conditions in cell culture. An interesting picture emerged when Stat-1 null mice were subjected to ischemia-reperfusion injury (IRI). During the initial injury phase, STAT1 in TECs appears to be responsible for recruiting macrophages to the injured kidney (Kemmner et al., 2019). Inhibiting STAT1 is therefore beneficial during this phase and lessens the degree of renal damage. However, persistent lack of STAT1 in macrophages leads to their increased M2-polarization and increased fibrosis later after IRI (Kemmner et al., 2019). Therefore, inhibition of STAT1 ultimately leads to worsened progression to chronic kidney disease (CKD). This example highlights the need to be very clear about the cell type that one is studying. Finally, aging human kidneys reveal an increase in STAT1 activity besides other inflammation-associated transcription factors highlighting its contribution in renal inflammation and injury (Figure 1) (Bai et al., 2018; O’Brown et al., 2015; Shi et al., 2010).

STAT3.

STAT3 signaling is known for its important role in inflammation and immunity. For example, STAT3 mediates development, activation and migration of neutrophils, B-lymphocytes, dendritic cells and macrophages throughout different organs upon inflammation (Fielding et al., 2008; Lee et al., 2002; Melillo et al., 2010; Nefedova et al., 2004; Park et al., 2004; Takeda et al., 1999). Overall, STAT3 signaling is thought to be crucial in balancing inflammatory and anti-inflammatory responses (Hillmer et al., 2016). Several kidney diseases including PKD exhibit inflammation and immune cell infiltration (O’Brown et al., 2015; Weimbs et al., 2018). STAT3 activation, for example, was found in macrophages and interstitial fibroblasts facilitating leukocyte infiltration and ECM protein production upon acute kidney injury (AKI) in rats and mice (Bienaimé et al., 2016; Kuratsune et al., 2007; Pang et al., 2010). This indicates that STAT3 signaling in inflammatory and immune cells might likely play a role in development and progression of certain kidney diseases (O’Brown et al., 2015; Weimbs et al., 2018). This section will further focus on the role of STAT3 in renal TECs due to its great importance for PKD.

STAT3 is highly activated in embryonic rat kidneys (phosphorylation on Y705) and in TECs of early postnatal mice (Talbot et al., 2011; Wang et al., 2010) suggesting a certain role in renal development and maturation. Stat3 null mice have been unhelpful in this regard due to early embryonic lethality (Takeda et al., 1997). With the end of renal proliferation in mice, around day 14, STAT3 activity is strongly down regulated in mature mouse kidneys (Talbot et al., 2011). However, STAT3 expression itself remains high in healthy adult kidneys indicating that it is ready to be activated as shown in the aging human kidney and in several forms of renal insults including tubulointerstitial fibrosis or glomerulosclerosis (Du et al., 2018; Estrada et al., 2018; O’Brown et al., 2015; Talbot et al., 2011). Active STAT3 is localized to a broad spectrum of cells depending on the type of kidney injury. While animal models of DN, focal segmental glomerulosclerosis (FSGS) and rapidly progressive glomerulonephritis (RPGN) reveal STAT3 activation in glomerular cells (Wang et al., 2002), settings of tubulointerstitial fibrosis and AKI exhibit STAT3 activation predominantly in tubular, interstitial and endothelial cells (Bienaimé et al., 2016; Dube et al., 2017; Kuratsune et al., 2007; Pang et al., 2010). Using ureteral obstruction (UUO) as a model of renal fibrosis, STAT3 activation was found between day 1 and day 7 after the onset of obstruction in mice and rats (Kuratsune et al., 2007; Pang et al., 2010). Here, STAT3 activation was localized to both interstitial cells and TECs (Kuratsune et al., 2007). STAT3 inhibition with S3I-201 inhibited renal fibrosis and inflammation in the murine model (Pang et al., 2010). Similarly, 75% nephrectomy, a slower model of CKD, led to STAT3 activation in TECs and interstitial cells after about 6 weeks and coinciding with features of CKD including fibrosis. Remarkably, genetic inactivation of STAT3 specifically in TECs of mice alone was sufficient to prevent the progression to fibrosis and other CKD features after 75% nephrectomy. Based on these in vivo studies and additional in vitro experiments, these authors concluded that STAT3 activation in TECs leads to expression of a suite of genes including pro-fibrotic, secreted factors (Figure 1) that then trigger the activation of interstitial myofibroblasts. Consequently, STAT3 is activated in myofibroblasts leading to kidney fibrosis (Bienaimé et al., 2016). This conclusion is consistent with a model showing that necrotic TECs trigger IL-22 secretion in CD45+ phagocytes by releasing TLR4 agonists (Kulkarni et al., 2014). Interestingly, IL-22 receptor expression is exclusively found on TECs while IL-22 is selectively secreted by CD45+ phagocytes in the kidney. However, IL-22 pretreatment in an IRI mouse model increases STAT3 activity in TECs and in contrast ameliorates AKI (Xu et al., 2014). In this model, blocking IL-22 signaling with an IL-22 antibody or by using IL-22 deficient mice led to impaired renal recovery suggesting a protective effect of STAT3 signaling. The same results were achieved by blocking Toll-Like receptor 4 (TLR4) with a TLR4 antibody in the healing phase of this model (Kulkarni et al., 2014). Beside IL-22, pretreatment with IL-6/sIL-6 receptor fusion protein increased STAT3 activity in TECs, too, and strikingly reduced AKI in a mercury-induced AKI mouse model pointing to a similar preventive effect of IL-6/STAT3 trans-signaling (Nechemia-Arbely et al., 2008). Interestingly, endothelial-specific Stat3 deletion in mice increased kidney injury in an IRI model confirming a beneficial effect of STAT3 (Dube et al., 2017). Furthermore, JAK/STAT3 signaling might be required for survivin-mediated recovery effects in mice upon IRI (Chen et al., 2013a). Pretreatment with the STAT3 inhibitor S3I-201 in this model led to decreased survivin expression and reduced AKI recovery (Chen et al., 2013a). In the past, it has been proposed that STAT3/MAPK/ERK1/2 signaling affects fibrosis and inflammatory responses by mediating the transformation of TECs to myofibroblasts, in a process of epithelial-to-mesenchymal transition (EMT) (Iwano et al., 2002; Jain et al., 1998; Kuratsune et al., 2007; Masaki et al., 2003). The results described above, however, show that EMT does not significantly contribute to the development of myofibroblasts in CKD models and rather suggest that these cells arise from resident fibroblast or pericytes and bone marrow-derived circulating monocytes (Bienaimé et al., 2016; Humphreys et al., 2010; LeBleu et al., 2013). This process seems to involve a crosstalk between interstitial cells with epithelial cells like TECs and glomerular cells that is mediated by STAT3 signaling (Bienaimé et al., 2016).

Therefore, the emerging picture is that several renal insults affect TECs first resulting in STAT3 activation. This triggers the secretion of signaling molecules which attract and regulate fibroblasts - in a process that may also involve STAT3 activation in those cells (Figure 1) (Bienaimé et al., 2016; Kulkarni et al., 2014). Additionally, the interstitial cells can communicate back to TECs by further release of cytokines like IL-22 (Kulkarni et al., 2014). In this way STAT3 signaling appears to mediate early inflammatory-like responses upon kidney injury but also induces recovery mechanisms that are important for later stages but can lead to fibrosis if persistently activated.

STAT5.

STAT5 is described to regulate proliferation, survival and apoptosis in immune and cancer cells (Behbod et al., 2003; Cui et al., 2004; Xiong et al., 2009). STAT5 defines a group of two closely related proteins, STAT5A and STAT5B, encoded by separate genes, that can be activated by several cytokines including GH, EGF and IL −2, −3, −5, −7, −9 and −15 via tyrosine phosphorylation (694/699) by members of the JAK and Src family (Figure 2). Interestingly, Src only mediates STAT5b activation (Kazansky et al., 1999). While Stat5a−/− knockout mouse models have not shown any renal alterations, Stat5b−/− deficient mice have exhibited renal pathologies in glomerular structure and proteinuria indicating a distinct role of STAT5b in renal development and the function of healthy kidneys (Cui et al., 2004; Udy et al., 1997; Villarino et al., 2016). Furthermore, a CKD rat model revealed a certain impact of GH/STAT5 signaling in renal diseases. Subtotal nephrectomy, as a CKD model, leads to a decrease in JAK2 and STAT5 activity. Additionally, GH receptor (GHR) levels were reduced beside normal GH serum levels suggesting a possible GH resistance. Notably, GH treatment in these CKD rats significantly increased STAT5 phosphorylation without affecting JAK2 activity. Interestingly, CKD rats showed increased levels of IL-6, pSTAT3 as well as SOCS3 which is known to inhibit GH/JAK2 signaling and thus may contribute to a possible GH resistance (Wiezel et al., 2014). On the other hand, it has been shown that STAT5 is activated upon AKI induction in murine kidneys (Huen et al., 2015; Zhang et al., 2017a). Remarkably, using an IRI mouse model, AKI led to tubular secretion of GM-CSF which in turn seems to induce alternative M2 polarization via STAT5 signaling in macrophages (Figure 2). Blocking GM-CSF with an antibody upon IRI partially decreases STAT5 activation leading to decreased M2 polarization and TEC proliferation. This suggests that STAT5 signaling could contribute to renal injury and repair mechanisms by mediating an alternative way of M2 macrophage polarization in renal macrophages (Huen et al., 2015). Moreover, JAK2/STAT5 signaling seems to mediate the cytoprotective effect of erythropoietin (EPO) on TECs. EPO pretreatment of primary murine TECs in the antimycin A-induced ischemic injury model leads to JAK2 phosphorylation and reduces cytotoxicity in these cells. In contrast, this effect is abolished in primary TECs from mice lacking intact STAT5 signaling (EPOR-HM mice) suggesting that JAK2/STAT5 signaling is required for EPO-mediated cytoprotection in TECs (Breggia et al., 2008). Taken together, STAT5 signaling occurs in renal TECs and macrophages possibly affecting renal diseases by mediating cell survival, inflammation and tissue repair mechanisms (Figure 2).

STAT6.

Renal TECs in adult kidney express STAT6 but exhibit little or no STAT6 activation. However, systemic IL-4 or IL-13 treatment in mice result in rapid (within one hour) and strong STAT6 activation in renal TECs (Olsan et al., 2011, 2018) suggesting that the STAT6 pathway is “on standby” in these cells. Kidney injury by UUO in mice revealed JAK3/STAT6 activation in interstitial cells, too, leading to renal fibrosis with bone marrow derived fibroblast accumulation (Figure 2). Treatment with the JAK3 inhibitor CP690,550 (tofacitinib) in this model led to STAT6 inhibition and reduced these effects. Interestingly, STAT6 deficient mice (Stat6−/− by SH2 domain deletion) in the same UUO model revealed reduced M2 macrophage polarization and myofibroblast transformation (Yan et al., 2015). Thus, a model has been suggested where JAK3/STAT6 signaling mediates myofibroblast accumulation and their transformation from circulating monocyte-derived M2 macrophages (Yan et al., 2015; Yang et al., 2013). Additionally, IL-4-receptor deficiency in mice also inhibited STAT6 activation in a folic acid nephropathy model resulting in a decrease of fibroblast activation in the kidney, again supporting the notion that activation of this pathway contributes to renal fibrosis (Liang et al., 2017). Interestingly, aberrant upregulation of IL-13 was found in renal insults like lupus nephritis (Chen et al., 2001). However, some contrasting results suggest a protective role for STAT6 activation in the kidney. Pretreatment with IL-13 for example reduced tubulointerstitial damage in an IRI model (Sandovici et al., 2008). Moreover, STAT6 null mice (Stat6−/− by first exon coding deletion) revealed increased tubule injury and decreased renal function upon IRI and exhibited stronger apoptosis and inflammation compared to wild type mice in a UUO model (Yokota et al., 2003; Yukawa et al., 2005). These conflicting results in Stat6 null mice might be due to different gene deletion models leading to different phenotypes (Wang et al., 2009). More recent results suggested that EPO treatment in the same UUO model may lead to STAT6 activation protecting mice from AKI by decreasing inflammatory infiltration (Zhang et al., 2018).

In conclusion, JAK3/STAT6 activity was found in TECs and interstitial cells in the kidney primarily in response to IL-4 and IL-13. While IL-4 signaling has been suggested to drive renal fibrosis (Liang et al., 2017) other results showed beneficial effects of IL-13/IL-4 by facilitating tissue repair after renal insults (Sandovici et al., 2008; Zhang et al., 2017a). Again, timing may be crucial in explaining the differences, with STAT6 activation initially playing a beneficial role during injury repair and later playing a detrimental role in fibrosis if the signal is not terminated. Contrasting effects due to STAT6 pathway activation in different renal cell types likely occur and further investigation will be needed to better understand such opposing results (Liang et al., 2017; Zhang et al., 2017a).

The (complicated) mechanisms of STAT regulation by polycystin-1 (PC1)

The loss of functional PC1 leads to ADPKD but “how” is much less clear. PC-1 is a large integral membrane protein with a size exceeding 460 kDa, containing a large extracellular domain consisting of roughly 3,000 amino acids, eleven predicted transmembrane domains and a short cytoplasmic tail of about 200 amino acids (Merrick et al., 2014). It was initially reported that direct association of PC1 with JAK2 leads to STAT1 activation (Bhunia et al., 2002). This mechanism was shown to be dependent on the interaction of PC1 with PC2 and leading to upregulation of p21waf1, a potent inhibitor of cyclin-dependent kinase 2 (Cdk2), resulting in growth arrest.

PC1 undergoes cleavage in both the N-and C-terminal domains. The N-terminal cleavage occurs at the G protein-coupled receptor proteolytic site (GPS) (Qian et al., 2002) while the C-terminal domain undergoes at least 3 cleavages. Those cleavage events have been found to release the entire cytoplasmic tail (~30 kDa) (Bertuccio et al., 2009; Chauvet et al., 2004; Low et al., 2006; Talbot et al., 2011), a smaller fragment corresponding to the C-terminal half of the cytoplasmic tail (~15 kDa) (Low et al., 2006; Talbot et al., 2011) and a larger fragment (~100kDa) containing 6 transmembrane domains and the cytoplasmic tail (Woodward et al., 2010). Our group showed that STAT6 activity is inhibited by membrane-anchored PC1 yet cytoplasmic tail fragments can interact with STAT6 and the transcriptional co-activator P100 and co-activate STAT6-dependent gene expression. This on/off regulation of STAT6 activity appeared to be controlled by changes in apical/tubular fluid flow suggesting a mechanism of mechano-transduction (Low et al., 2006) that was proposed to involve the flow-regulated cleavage (Chauvet et al., 2004) of the cytoplasmic tail of PC1. It was further demonstrated that STAT6 is aberrantly activated in cyst-lining epithelial cells in PKD and that genetic or pharmacological inhibition of STAT6 in a PKD mouse model leads to inhibition of renal cyst growth (Olsan et al., 2011).

PC1 has been ascribed a number of functions relevant to PKD including acting as an adhesion protein (Streets et al., 2009) and regulating microtubule and actin dynamics in response to injury (Castelli et al., 2015). Cleavage of full length PC1 into its C-terminal tail (PC1-CT) appears to be a mechanism of mechanosensation utilizing the STAT3 signaling pathway. Dalagiorgou et al. investigated the PC1-CT/STAT3 axis outside the kidney outlining how PC1-CT/STAT3 contributes to mechanosensation wherein following mechanical stretching of osteoblasts, the JAK2/STAT3 pathway becomes activated to regulate cell differentiation via upregulation of the osteoblastic regulator of differentiation Runx2 (Dalagiorgou et al., 2017). This activation of JAK/STAT3 via PC1 may be mediated by cleavage of the PC1 C-terminal tail (Dalagiorgou et al., 2017). Our group investigated the possible regulation of STAT3 by PC1 and we reported a dual mechanism (Talbot et al., 2011) as well as a third mechanism (Talbot et al., 2014). JAK2 was found to bind to the membrane-proximal half of the cytoplasmic tail of membrane-anchored PC1 causing direct, JAK2-dependent activation of STAT3 (Talbot et al., 2011). Cleavage of the cytoplasmic tail of PC1 resulted in the loss of its ability to activate STAT3 via JAK2. However, the 30 kDa PC1 tail cleavage product (PC1–p30) was able to translocate to the nucleus and co-activate tyrosine-phosphorylated STAT3 (and also STAT1), which has been activated by cytokine signaling (Talbot et al., 2011). PC1–p30 can also interact with the tyrosine kinase Src and directly activate Src leading to phosphorylation and activation of STAT3 (Talbot et al., 2014). While activation of STAT3 by membrane-bound PC1/JAK2 is inhibited by SOCS3, activation of STAT3 by PC1–p30 is insensitive to SOCS3 inhibition. It was further shown that this signaling pathway is amplified by binding of EGF to its receptor and by increased levels of cAMP. By integrating inputs from EGFR and cAMP signaling, PC1–p30 appears to be able to activate STAT3 in a cytokine/JAK2-independent manner by bypassing inhibition by SOCS3 (Talbot et al., 2014). These observations suggest that STAT3 may be a key integrator transmitting extracellular mechanosensation to changes in gene expression.

Together, these results suggest that PC1 regulates STATs through several distinct pathways. First, the membrane-anchored PC1 can activate STAT1 and STAT3 through JAK2-mediated phosphorylation. Second, PC1 C-terminal fragments can co-activate STAT1, STAT3 and STAT6 depending on whether they had previously been activated by canonical signaling. STAT3, however, can also be directly activated by Src through the interaction with PC1–p30. This complicated situation suggests that PC1 may be an important integrator of several incoming signaling pathways, which can cause various downstream effects depending on the cellular situation (e.g. under flow-vs. no-flow conditions)

STAT1 in PKD

STAT1 was the first STAT family member to be investigated in PKD, leading to the conclusion that PC1 may regulate the cell cycle via JAK2-mediated STAT1 regulation (Bhunia et al., 2002). This regulation was suggested to occur in tubule/cyst epithelial cells but has not been studied further. More recently, STAT1 has been investigated in other tissues and cell types involved in PKD. Cardiovascular complications, including hypertension and aneurysms are associated with ADPKD caused by mutations in PC1. Kwak et. al recently reported a link between PC1 and STAT1 in endothelial cells in PKD. PC1 is an atypical G-protein-coupled receptor that can undergo cleavage at a conserved GPS motif near the first transmembrane domain. Kwak et. al found that the inhibitory G protein Gαi3 binds the membrane anchored C-terminal tail (CTT) of PC1, and only upon cleavage of PC1 at the GPS motif will Gαi3 dissociate from the PC1-CTT and activate the classical transient receptor potential channel 4 (TRPC4). The activation of TRPC4 causes an influx of calcium which in turn causes the activation of STAT1 to regulate endothelial cell proliferation and apoptosis, which was suggested to relate to cardiovascular abnormalities in PKD (Kwak et al., 2018). This reported mechanism would appear to be quite indirect and very different from the one reported previously in epithelial cells.

The importance of the dysregulation of immune system components, specifically inflammation, in ADPKD has gained increasing attention. Inflammatory cells such as macrophages are known to accumulate in polycystic kidneys where they differentiate into the M2 phenotype (Swenson-Fields et al., 2013). Furthermore, it has been reported that the alternative complement pathway is aberrantly activated in PKD and that its inhibition ameliorates the disease severity through decreased macrophage infiltration and complement factor B (CFB) expression (Su et al., 2014; Zhou et al., 2012). CFB is over expressed in ADPKD patient kidneys and Cy/+ Han:SPRD rat kidneys and was found to be correlated with an increase in JAK2/STAT1 activation and expression of the 30 kDa PC1-CTT in these cystic kidneys (Wu et al., 2016). The JAK2/STAT1 pathway is known to promote tumor growth primarily through its pro-inflammatory properties (Graziani et al., 2010). Henceforth, it was found that over expression of STAT1 in UCL93 renal epithelial cells causes increased CFB expression and inhibition of STAT1 leads to decreased CFB expression. Illuminating where the PC1-CTT fits into this pathway, over expression of PC1-CTT in renal epithelial cells leads to increased JAK2/STAT1 activation and CFB up regulation (Wu et al., 2016). Macrophage differentiation into the M2 phenotype once in the cystic kidneys was hypothesized to be caused by a soluble factor secreted by renal epithelial cells. Strikingly, CFB promotes macrophage conversion to the M2 phenotype and this conversion is inhibited by inhibition of STAT1 (Wu et al., 2016). These results begin to paint a picture that the PC1-CTT increases CFB through STAT1 activation, which may in turn cause M2 macrophage phenotype conversion. This pathway may play a major role in the inflammatory phenotype seen in ADPKD cystic kidneys. Furthermore, these findings suggest that STAT1 inhibition may be a promising new target in inhibiting M2 macrophage conversion and the subsequent inflammation in ADPKD kidneys.

STAT3 in PKD

STAT3 dysregulation has been studied intensively within the cancer research field with a number of parallels between STAT3 activity in cancer and PKD. Activation of STAT3 signaling is frequently implicated in tumor angiogenesis, tumor survival, immunomodulation, extracellular matrix remodeling and EMT of precancerous cells (Avalle et al., 2017). Similarly, polycystic kidneys are characterized by increased expression of extracellular matrix proteins (Bello-Reuss et al., 2001; Liu et al., 2012), EMT phenotypes (Togawa et al., 2010), altered cellular growth, apoptosis and changes to immune cell function. The effects of STAT3 in cancer are believed to be contextual as STAT3 may play both pro-oncogenic and tumor suppressor roles dependent on other gene mutations within the cell (Avalle et al., 2017). A contextual role for STAT3 may also be at play in PKD. Our lab has previously reported that polycystic kidneys in multiple rodent models of PKD exhibit increased phospho-tyrosine-STAT3 (pY-STAT3) within cyst lining epithelia (Talbot et al., 2011) with others having reported pY-STAT3 in immune cells within polycystic kidneys (Peda et al., 2016). STAT3 as a driver of disease progression is suggested by the use of putative STAT3 inhibitors in PKD rodent models resulting in decreased cystic disease (Leonhard et al., 2011; Patera et al., 2019; Takakura et al., 2011) with the caveat that these inhibitors have limited specificity and affect more than just cystic epithelial cells. Recently, the role of STAT3 as a driver of cyst progression was tested by cell specific deletion of Stat3 within mouse proximal tubule cells. Cell-specific Stat3 deletion led to modestly diminished cystic kidneys but paradoxically decreased kidney function. The loss of Stat3 was found to induce a strong inflammatory response, recruiting infiltrating macrophages. It was suggested that STAT3 expression in proximal tubules regulates the immune response in PKD by suppression of the pro-inflammatory chemokines CCL5 and CXCL10 (Viau et al., 2019). These findings imply that the role of STAT3 in PKD is more complex than promoting cystogenesis and it is the signaling between the kidney and the immune system that is critical for how kidney function declines. The benefit of STAT3 pharmacological inhibition on cystic disease progression may therefore be due to inactivation of STAT3 in kidney immune cells such as CD8+ T-cells or interstitial macrophages. Both of which have been shown to affect PKD progression (Cassini et al., 2018; Kleczko et al., 2018; Peda et al., 2016; Swenson-Fields et al., 2013; Yang et al., 2018). Under normal conditions, AKI induces recruitment of bone marrow-derived monocytes and proliferation of resident kidney macrophages (Peda et al., 2016), after which these cells undergo a shift to an M2-like phenotype to promote tissue repair. IL-10 is released following kidney injury to promote the stimulation of macrophages to begin the repair process via activation of JAK1, Tyk2, and subsequently STAT3 (Mosser and Zhang, 2008). This repair process in ADPKD may be dysfunctional leading to enhanced cyst growth, cyst expansion and fibrosis (Swenson-Fields et al., 2013). Human ADPKD tissue samples display increased IL-10 expression, enrichment of IL-10 within cyst fluid along with increased STAT3 expression. Stimulation in vitro with ADPKD cell culture media induced the activation of kidney macrophages and blocking IL-10 with an IL-10 function blocking antibody prevented STAT3 activation in these macrophages. Besides the release of IL-10 from cystic epithelia to activate STAT3, multiple factors may play a role in the reprogramming of macrophages, including prostaglandins, cAMP, lipid resolvins and adenosine (Peda et al., 2016). Recently, the transcription factor Interferon Regulatory Factor 5 (IRF5) was found to promote cystogenesis following unilateral nephrectomy in Pkd1-null mice via activation of resident kidney macrophages. IRF5 is known to be expressed in macrophages and regulate the expression of inflammatory cytokines IL-6 and TNF-α. Treatment with an antisense oligonucleotide (ASO) against Irf5 prevented accelerated cyst expansion but not cyst number. Using flow cytometry, the authors concluded that Irf5 ASO affected primarily resident kidney macrophages preventing their release of IL-6 and the subsequent activation of STAT3 in tubule epithelial cells (Zimmerman et al., 2020).

Recently, our lab discovered a role for STAT3 in response to AKI and how this response contributes to cystic disease progression. Torres et. al found that kidneys challenged with calcium oxalate (CaOx) microcrystals led to the activation of PKD-associated pathways in TECs including STAT3, mTOR, and Src. Wild-type rats challenged with CaOx crystals displayed temporary tubule dilation along the entire length of the nephron to allow for the passage of precipitated crystals and coincided with the activation of pY-STAT3 (Torres et al., 2019). Challenging both the Han:SPRD and the PCK rat models of PKD with dietary induced renal crystals led to increased numbers of cysts and worsened disease progression, implicating crystals as triggers for cystogenesis (Torres et al., 2019). In summary, this study suggests that STAT3 activation is required for crystal-induced tubule dilation as a reno-protective mechanism and that this process is inappropriately activated in PKD leading to cystogenesis.

In addition to the activators discussed above, STAT3 activity has also been shown to be modulated by the Renin-Angiotensin-Aldosterone-System (RAAS). RAAS has been well characterized and its involvement in ADPKD extensively studied in the HALT clinical trials testing the efficacy of angiotensin converting enzyme (ACE) inhibitors alone or in combination with angiotensin receptor blockers (ARB) (Schrier et al., 2014; Torres et al., 2012a, 2014). These studies found no beneficial effect for patients with ADPKD. Despite having no beneficial effect in humans, subsequent studies in rodent models have shown that RAAS inhibition can slow PKD progression (Fitzgibbon et al., 2018; Ravichandran et al., 2015; Saigusa et al., 2016). This beneficial effect may be mediated via STAT3 signaling as STAT3 has been shown to be activated by angiotensin II (Ang II) and mediated by TLR4 (Xu et al., 2019). In support of the role of Ang II activation of STAT3, following TLR4 knockdown in rat TECs (NRK-52E), JAK2 and STAT3 activation is diminished with a subsequent reduction of TGF-ß and collagen production (Xu et al., 2019). The lack of efficacy in slowing disease progression in human ADPKD with ACE and ARB inhibitors may indicate that Ang II activity plays a role early in the disease and therefore inhibition at later stages provides no benefit.

Taken together, these data present a compelling case for the importance of STAT3 signaling in the pathogenesis of PKD.

Src in PKD

Multiple publications have linked activation of the tyrosine kinase Src to pathophysiology of PKD and STAT3 activation. Src-mediated activation of STAT3-dependent transcription was initially identified in models of Src-mediated tumorigenesis. Src induces phosphorylation of Y705 of STAT3 thus promoting its dimerization and nuclear translocation (Silva, 2004). A link between Src and PKD proteins was initially suggested when PC1 was found to form a complex with focal adhesion proteins and that Src could induce tyrosine phosphorylation of the cytoplasmic tail of PC1 (Li et al., 1999; Wilson et al., 1999). Importantly, Src activity correlates with disease severity in both orthologous and non-orthologous mouse models of ARPKD although the mechanisms underlying Src activation remain incompletely understood (Elliott et al., 2011; Sweeney et al., 2008). Pharmacological inhibition of Src attenuates the phenotype in various PKD models (Sweeney et al., 2008). Tesevatinib, a multi-tyrosine kinase inhibitor that affects EGFR and Src activity, has been shown to ameliorate progression of PKD in rodent models and is currently in first clinical trials for both ARPKD and ADPKD (Sweeney et al., 2017)(ClinicalTrials.gov identifiers NCT03096080 and NCT03203642). The efficacy and safety of Bosutinib, an oral dual Src/Bcr-Abl tyrosine kinase inhibitor, was recently evaluated in a phase 2 trial in ADPKD patients in early stages. The annual rate of kidney enlargement was reduced in bosutinib-treated patients but no beneficial effect on kidney function was detected, and adverse effects were common leading to high rate of discontinuation (Tesar et al., 2017).

As already described above, PC1 regulates STAT3 activity via several distinct mechanisms including one pathway involving Src. Our lab has reported that the cleaved, soluble cytoplasmic tail of PC1 induces Src-dependent activation of STAT3 (Talbot et al., 2011, 2014). The same tail fragment of PC1 also increased the activating effect of EGFR on the Src-STAT3 axis (Talbot et al., 2014). Importantly, hyperactivation and apical mislocalization of EGFR have been described in various PKD models (Orellana et al., 1995; Richards et al., 1998). Patients with severe ARPKD courses and increased expression of EGFR have been described (Arbeiter et al., 2008). Conversely, mutations in Egfr that result in reduced tyrosine kinase activity can attenuate the PKD phenotype in a rodent model of PKD (Richards et al., 1998). Pharmacological inhibition of the EGFR tyrosine kinase results in a less pronounced phenotype in a murine PKD model (Sweeney et al., 2003). Taken together, the data suggests that EGF/EGFR signaling seems to contribute to the development and severity of a PKD phenotype.

In addition to EGFR, increased intracellular cAMP has been shown to further enhance the activating effect of the cleaved PC1 tail on the Src-STAT3 axis (Talbot et al., 2014). This effect could be inhibited by the protein kinase A (PKA) inhibitor H89 suggesting that PKA is involved in this process (Talbot et al., 2014). PKA activation has been associated with both hepatic and renal phenotypes including Src activation in an orthologous PKD animal model (Banales et al., 2008; Ye et al., 2017). Intracellular cAMP levels can be increased by activation of the vasopressin V2 receptor (V2R), which has been described in PKD and is thought to drive cystic progression (Gattone et al., 2003; Wang et al., 2008). These landmark experimental studies paved the way for translation of basic research into the clinic and led to the approval of the V2R antagonist Tolvaptan as the first drug therapy for ADPKD in adult patients in certain disease stages (Gastel and Torres, 2017; Torres et al., 2012b, 2017). A clinical trial for safety and tolerability of Tolvaptan in children with ADPKD is ongoing (Schaefer et al., 2019).

In summary, there is ample evidence for activation of Src and Src-associated signaling in PKD. However, it is not completely clear whether STAT3 is the most critical downstream effector of Src, EGFR and cAMP signaling that drives cystic progression or whether other pathways are more important.

STAT5 in PKD

The research group of Albert Ong reported that STAT5 signaling may play an important role in aberrant cell proliferation in ADPKD. Inhibition of STAT5 by siRNA or a small molecule inhibitor (VWR 573108) reduced cell proliferation and mitosis as well as cyst growth in PKD cell lines. Furthermore, STAT5 activation was found to be significantly increased in cyst lining cells of two murine ADPKD models (Pkd1nl/nl mice, Pkd1fl/fl, Ksp-CreERT2 mice) peaking at 4 weeks of age and decreasing after week 10 (Fragiadaki et al., 2017). However, it remains unknown whether STAT5 is excessively activated in human ADPKD kidneys, too.

This altered STAT5 activation might be mediated by increased growth hormone/receptor (GH-GHR) signaling since GHR signals via STAT5 and since Pkd1nl/nl mice exhibit aberrantly elevated GH serum levels. Moreover, using a GH overexpressing mouse model, GH seems to induce STAT5 activation independently of Pkd1 expression status (Fragiadaki et al., 2017). Interestingly, this mouse model was previously shown to display renal tubular dilatations (Friedbichler et al., 2012). STAT5 was found to be activated and localized in these dilated tubules beside increased expression of Cyclin D1. Additionally, GH stimulation of human PKD1 wild type TECs (UCL93) revealed increased binding of STAT5 to the CyclinD1 promoter suggesting that GH-induced STAT5 activation promotes proliferation mediated by CyclinD1 (Fragiadaki et al., 2017). However, it remains to be elucidated whether GH levels are also elevated in human ADPKD.

Taken together, this is the first evidence that the GH-STAT5 axis could affect the pathogenesis of ADPKD. However, additional upstream activators may be considered as an alternative driving force such as EGF/EGFR known to be elevated in PKD kidneys and to mediate STAT5 signaling (Groner et al., 2000; Sweeney and Avner, 2006). Furthermore, analyzing the specific STAT5 isoform as well as the involvement of activating kinases could lead to more insights into its role in ADPKD.

STAT6 in PKD

Our group first investigated the role of STAT6 in PKD. The initial study suggested that PC1 suppresses STAT6 activity under normal conditions but that cessation of apical fluid flow triggers cleavage of the cytoplasmic tail of PC1 allowing nuclear translocation of the PC1 tail and STAT6 in complex with the co-activator P100 (Low et al., 2006). Building on these initial findings, we then reported STAT6 over expression in cysts in ADPKD human kidneys and high levels of activated STAT6 in the cyst-lining cells of two different PKD mouse models (Olsan et al., 2011). Genetic removal or pharmacologic inhibition of STAT6 in the bpk mouse model led to a significant decrease in cyst size and improvement in kidney function. Furthermore, the cytokine and potent activator of STAT6, IL-13, was found to be significantly over expressed by cyst lining cells and accumulates in cyst fluid, while the IL-13 receptor is also over expressed in cyst lining cells (Olsan et al., 2011). These results suggested that a positive feedback loop between IL-13, its receptor and STAT6 leads to persistent activation of this pathway in cystic epithelial cells.

STAT6 activation in TECs may play a role in the pathology of PKD via activation of fibrosis pathways and dysregulation of immune system components. Fibrosis is a major factor in the severity of ADPKD, and it has been shown that IL-13 can activate fibrosis pathways in the lung and liver (Kaviratne et al., 2004; Wynn, 2004). We reported that IL-13 and STAT6 activity mediate the expression and upregulation of periostin, galectin-3 and IL-24 in various forms of PKD (Olsan et al., 2018). Periostin has previously been shown to be highly overexpressed in renal tissue of human ADPKD and ARPKD patients, and in multiple mouse models of PKD, where it appears to be secreted by cyst lining cells, accelerate cyst growth and promote fibrosis (Raman et al., 2018; Wallace, 2019; Wallace et al., 2008, 2014; Watanabe et al., 2019). Periostin expression increases after IL-13 treatment in kidney epithelial cells, and Stat6 gene ablation in bpk mice leads to a substantial reduction of periostin expression down to the level in wild-type kidneys (Olsan et al., 2018). Periostin may contribute to renal cyst growth in an auto/paracrine fashion in response to STAT6 signaling via IL-13 activation in cyst lining cells. Galectin-3 is a β-galactoside-binding lectin that is expressed during kidney development, has been shown to be upregulated and promote fibrosis after kidney injury, is known to play a role in CKD, chronic heart failure, and may contribute to inflammatory processes in Systemic Lupus Erythematosus Nephritis (Chen and Kuo, 2016). Furthermore, pharmacological inhibition of galectin-3 has been shown to reduce renal fibrosis in renal damage associated with obesity and aortic stenosis (Martinez-Martinez et al., 2016). Galectin-3 has also been found to localize to cystic epithelial cells in both the cpk mouse model and in human ARPKD, suggesting galectin-3 contributes to the fibrosis seen in PKD (Chiu et al., 2006; Winyard). Galectin-3 expression increases after IL-13 treatment in kidney epithelial cells and after injecting IL-13 in vivo (Olsan et al., 2018). This suggests that in cystic epithelial cells, IL-13 activation of STAT6 may cause activation of galectin-3, which in turn promotes the fibrosis seen in PKD kidneys. IL-24 has been shown to play a role in tissue integrity, wound healing, the response of epithelial cells to infections (Ouyang et al., 2011), tissue remodeling in the kidney in response to UOO in newborn mice (Castelli et al., 2015), and plays a role in the pathophysiology of irritable bowel syndrome and Crohn’s Disease in humans (Fonseca-Camarillo et al., 2014). IL-24 expression is upregulated by both IL-13 treatment and PC1–p30 overexpression in mouse and human kidney cells (Olsan et al., 2018) suggesting that it may play a role in epithelial cell dysregulation that leads to cyst formation.

The IL-13/STAT6 pathway is known to orchestrate a complex set of cellular responses and to play a role in multiple human diseases including allergic asthma (Antczak et al., 2016). When IL-13/STAT6 signaling is constitutively activated, this pathway appears to promote renal cyst growth and disease progression in PKD. While their exact roles remain largely unclear, a commonality in the possible functions of periostin, galectin-3 and IL-24 are their roles in regulating innate immune responses. Macrophages are a crucial component of the innate immune system, have recently been strongly implicated in the pathogenesis of PKD (Cassini et al., 2018; Karihaloo et al., 2011; Song et al., 2017; Swenson-Fields et al., 2013; Viau et al., 2018; Weimbs, 2018; Yang et al., 2018; Zimmerman et al., 2019, 2020) and may be involved in STAT6-mediated effects. Interestingly, M2 macrophages are IL-4 and IL-13 secreting (and responding) cells (Van Dyken and Locksley, 2013). In orthologous mouse models of both PKD1 and PKD2, a significant abundance of M2 macrophages was found in the interstitial space surrounding cysts and contribute to disease progression (Cassini et al., 2018; Karihaloo et al., 2011). In addition, it has been shown that galectin-3 expression and secretion by macrophages is critical in the activation of renal fibroblasts that leads to the promotion of renal fibrosis (Henderson et al., 2008). Moreover, it was found that following AKI there is an increase in periostin expression from TECs that drive macrophage proliferation and expression of regenerative factors (Kormann et al., 2019). Together, these findings may begin to paint a picture in which macrophages and tubule/cyst cells stimulate each other via IL-13 secretion and STAT6 activation leading to increased cyst proliferation and fibrosis. Furthermore, macrophage secretion of galectin-3 is critical in activation of renal fibroblasts (Henderson et al., 2008) and may play a role in the fibrosis seen in PKD. Altogether, these findings suggest that the IL-13/STAT6 pathway and its downstream targets, including periostin, galectin-3 and IL-24, may be promising therapeutic targets for PKD therapy.

JAK inhibitors

Janus kinases (JAKs) as the essential signaling mediators downstream of pro-inflammatory cytokines and many other growth factors have been pharmacologically targeted for the treatment of inflammatory diseases and cancers (Winthrop, 2017). JAK inhibitors (also called jakinibs) have been associated with numerous undesired effects such as an increased risk for infection as well as anemia and leukopenia due to signals from hematopoietic growth factors including erythropoietin transduced by JAK2 being inhibited (Schwartz et al., 2017).

To date, several JAK inhibitors have been FDA approved (See Table 2 for a comprehensive list of JAK inhibitors). Among the early JAK inhibitors are Tofacitinib (November 2012), which potently inhibits JAK1 and JAK3 and to a lesser degree also inhibits JAK2 (Kontzias et al., 2012, Danese et al., 2015) and Ruxolitinib, a selective inhibitor of JAK1/2 (Mesa et al., 2012). Second generation Janus kinase inhibitors such as Baricitinib (June 2018), Fedratinib (August 2019) and Upadacitinib (August 2019) have emerged more recently as more specific compounds. Most of the JAK inhibitors are approved for the treatment of myelofibrosis, rheumatoid arthritis (RA) or as immunosuppressants with none of them approved for treatment of renal diseases. Since different STAT pathways are aberrantly activated in ADPKD, targeting JAK proteins may be promising, although long-term side effects of these relatively broad-acting drugs will likely be a significant concern for this slowly-progressive disease requiring long-term therapy (Weimbs et al., 2018).

Table 2:

Pharmacological approaches to STAT inhibition

Molecule Effect Target(s) Indication (Clinical Trial Phase) Ref
JAK
Ruxolitinib Direct inhibition of JAKs JAK1, JAK2 Approved for treatment of polycythemiavera and myelofibrosis (MF) (Verstovsek et al., 2010)
Tofacitinib Direct inhibition of JAKs JAK1, JAK3 Treatment of Rheumatoid Arthritis (RA) and ulcerative colitis (Kremer et al., 2009)
Baricitinib Direct inhibition of JAKs JAK1, JAK2 Treatment of RA (Taylor et al., 2017)
VX-509 Direct inhibition of JAK3 JAK3 Treatment of RA (Genovese et al., 2016)
Fedratinib Direct inhibition of JAK2 JAK2 Treatment of MF (Harrison et al., 2017)
Upadacitinib Direct inhibition of JAK1 JAK1 Treatment of RA (Genovese et al., 2018)
STAT3
Triptolide Inhibits phosphorylation of JAK2/ STAT3 JAK/STAT3 pathway Phase 3 trial for HIV, Uncontrolled trial in PKD (Jing et al., 2018)
AZ505 Inhibits STAT3 phosphorylation SMYD2 Not Applicable (Ferguson et al., 2011)
Anti-IL10 Inhibition of macrophages Macrophages Not Applicable (Peda et al., 2016)
Pyrimethamine STAT3 dimerization STAT3 Treatment for Toxoplasmosis, (Takakura et al., 2011)
Curcumin Decreased STAT3 phosphorylation Wnt, TNF-a, MAPK, HIF1, notch-1 and mTOR and STAT3 Phase 3 trial planned for CKD Phase 2 Trial for AKI Phase 4 trial planned for PKD (Leonhard et al., 2011)
Nifuroxazide Reduced renal macrophage infiltration, TNF-a, IL-18, Decreased STAT3 phosphorylation Jak2/ STAT3, Tyk2 Treatment of colitis and diarrhea (Said et al., 2018)
Imatinib Inhibits STAT3 phosphorylation Tyr-Kinases Treatment of Chronic myeologenous leukemia (CML), gastrointestinal stromal tumors (GISTs) (Zaki et al., 2018)
Crizotinib Inhibits STAT3 phosphorylation c-Met Treatment of non-small cell lung cancers,
Phase 2 trials for renal cancers
(Mesarosova et al., 2017)
Stevia/Stevioside Inhibits STAT3 phosphorylation, Inhibits ERK1/2 phosphorylation Not known Several trials for diabetes mellitus (DM) and obesity (Potočnjak et al., 2017)
Cryptotanshinone Inhibits STAT3 phosphorylation SH2 Domain of STAT3 Trial planned for polycystic ovary syndrom (Chen et al., 2017)
Cinnamaldehyde Inhibits STAT3 phosphorylation JAKs Trials planned for DM and obesity (Huang et al., 2015)
Paclitaxel Inhibits nuclear translocation Tubulin Treatment of breast, ovarian and lung cancer (Zhang et al., 2015)
Resveratrol Inhibits pY-STAT3 and acetyl-STAT3 Sirtuin 1 Several trials for CKD and diabetic nephropathy (Ni et al., 2014b)
MS-275 Inhibits STAT3 and Akt phosphorylation HDAC1 Phase 1 trial for CML, acute myeloid leukemia (AML), and breast cancer Phase 2 trial for renal cancer (Tang et al., 2014)
Oligodeoxynucleotidedecoys Decreased STAT3 translation STAT3 mRNA Not Applicable (Miklossy et al., 2013)
Antisense oligonucleotide Decreased STAT3 translation STAT3 mRNA Not Applicable (Miklossy et al., 2013)
STAT5
Pimozide Decreased STAT5 phosphorylation Not known Approved for treatment of Tourette Syndrome (Nelson et al., 2011)
E804 (Indirubin derivatives) Decreased STAT5 phosphorylation and DNA binding activity Most likely Tyr kinases Not Applicable (Nam et al., 2012)
Chromone based nicotinyl hydrazine Weakly inhibit STAT5 phosphorylation STAT5 Not Applicable (Müller et al., 2008)
Stafib-2 STAT5b SH2 domain inhibitor STAT5 Not Applicable (Elumalai et al., 2017)
AC-3-19 STAT5 SH2 domain inhibitor STAT5 Not Applicable (Cumaraswamy et al., 2014)
AC-4-130 STAT5 SH2 domain inhibitor STAT5 Not Applicable (Wingelhofer et al., 2018)
STAT6
Teriflunomide Inhibits Phosphorylation of STAT6 Various Protein Kinases Approved for treatment of Multiple Sclerosis (Bar-Or et al., 2014; Olsan et al., 2011)
AS1517499 Inhibits Phosphorylation of STAT6 STAT6 Not Applicable (Chiba et al., 2009)
Niflumic acid Inhibits Phosphorylation of STAT6 JAK2/ STAT6 Commonly used for treatment of Rheumatoid Arthritis (Nakano et al., 2006)
YM-341619 Inhibits Phosphorylation of STAT6 STAT6 Not Applicable (Ohga et al., 2008)
PM-43I STAT5/STAT5 SH2 Domains STAT5/STAT6 Not Applicable (Knight et al., 2018)
Tralokinumab IL-13 neutralizing monoclonal antibody IL-13 Positive phase 2b study for AD, currently in phase 3 study for AD Wollenberg et al., 2019
Lebrikizumab IL-13 neutralizing monoclonal antibody IL-13 Positive phase 2b results for Atopic Dermatitis (Corren et al., 2011; Hanania et al., 2015; Noonan et al., 2013; Scheerens et al., 2014; Simpson et al., 2018)

Ruxolitinib is a JAK1/2 inhibitor and is approved for treatment of polycythemia vera and myelofibrosis (Verstovsek et al., 2012). Renal dysfunction is common in patients with primary myelofibrosis (PMF). A recent study showed that Ruxolitinib significantly improved renal function in patients with PMF (Strati et al., 2019) and discontinuation of the drug treatment resulted in worsening of renal function in PMF patients (Tefferi Pardanani, 2011). It is unknown how Ruxolitinib may affect renal function in PMF and, to date, there have been no studies to investigate Ruxolitinib as a treatment for renal disease.

The spice compound cinnamaldehyde, has also been shown to inhibit JAK/STAT3 activation following advanced glycation end-product (AGE) stimulation of the AGE specific receptor (RAGE) in HK2 cells. Following AGE stimulation for 48 hours with co-treatment of cinnamaldehyde, decreased pY-STAT3 and increased expression of SOCS3 levels were observed, and after 5 days of co-treatment a decrease in collagen 4 expression (Huang et al., 2015).

Recently, Patera et al. treated human and murine derived PKD cell lines in vitro with the JAK inhibitor Tofacitinib and reported inhibition of STAT3 tyrosine phosphorylation that they attributed to JAK2 inhibition (Patera et al., 2019). These authors also reported high JAK2 expression levels in cystic kidneys in the Pkd1nl/nl mouse model. However, the level of activation of JAK2 was not investigated in that study and no in vivo experiments were conducted with Tofacitinib. These authors’ hypothesis that STAT3 activation in PKD cysts may be due to JAK2 activity contradicts previous results by our group showing that the level of tyrosine-phosphorylation of JAK2 is not increased in two different PKD mouse models, and that STAT3 activation in PKD is independent of JAKs and mediated by Src instead (Talbot et al., 2014). Altogether, it is currently unclear whether JAK inhibitors may be beneficial in PKD.

STAT3 Inhibitors

STAT3 inhibition has been intensely studied as a therapeutic approach because of STAT3’s immune modulating effects, and in the cancer field because of its involvement in cancer cell transformation, apoptosis and proliferation (Beebe et al., 2018). Similarly, these effects of STAT3 are beginning to be tested in PKD and the inhibitors used to do so are growing in number. STAT3 can be targeted in a number of ways and each method attempted has both pros and cons in execution. Many of the compounds developed to target STAT3 act upstream of its activation targeting either cytokine or growth factors/receptors (e.g. GP130, IL-6R, IL-10R, EGFR, PDGF, FGFR) or GPCR type receptors. Blockade of activating cytokines (e.g. HGF, EGF, IL-5, IL-6, IL-10, interferons, Bone Morphogenic Proteins (BMP), Leukemia Inducing Factor (LIF)) inhibits their respective receptor activity. Alternatively, inhibition of the non-receptor tyrosine kinases Src and Abl, and receptor tyrosine kinases (e.g. c-Met, EGFR, FGFR, etc.) may also prevent STAT3 activation. As would be expected, inhibition of many of these kinases may have profound effects outside of STAT3 inhibition as each has numerous downstream effectors. Some of the compounds that inhibit these tyrosine kinases and subsequently STAT3 include the JAK and Src inhibitors (discussed above), triptolide (Jing et al., 2018), imatinib (Gleevac) (Zaki et al., 2018), and the Met inhibitor crizotinib (Mesarosova et al., 2017) (See Table 2 for a more comprehensive list of STAT3 inhibitors). Because STAT3 has pleiotropic effects, targeting specificity becomes paramount to avoid potentially life-threatening complications. Direct inhibition of STAT3 can be achieved by blocking its dimerization, activation by kinases, translocation via the SH2 domain, or by blocking DNA binding via the DNA binding domain (DBD). Compounds that target STAT3 dimerization such as S3I-201 (Takakura et al., 2011) have been tested in PKD. Outside of PKD, the natural product cryptotanshinone (Chen et al., 2013b, 2017) has been tested with some success. An alternative to STAT3 inhibition may be targeting translational and transcriptional regulation of STAT3. The use of antisense oligonucleotides and oligodeoxynucleotide decoys have also been tested in the cancer field (Miklossy et al., 2013). There may be other compounds that can be used for the inhibition of STAT3 as demonstrated by the repurposing of compounds for use as STAT3 inhibitors such as the antibiotic nifuroxazide which was found to inhibit STAT3 in diabetic rats, reducing macrophage infiltration, fibrosis and decreased mRNA and protein levels of TNF-α and IL-18 (Said et al., 2018).

Natural products are an exciting area of research for the treatment of PKD and specifically those that affect STAT3 signaling. The Indian spice turmeric, and its active component, curcumin (Leonhard et al., 2011), have already begun to be tested in clinical trials for the treatment of ADPKD (ClinicalTrials.gov Identifier: NCT02494141). Recently, other natural products have begun to show promise as STAT3 inhibitors. Stevioside, a component of the sugar substitute stevia was tested in the Pkd1flox/+:Pkhd1-Cre mouse model and shown to inhibit cyst growth (Yuajit et al., 2014) and has been shown to inhibit STAT3 following cisplatin induced kidney injury, preventing both Erk and STAT3 phosphorylation (Potočnjak et al., 2017). The observed effect on cyst growth by stevioside may also involve activation of AMPK followed by inhibition of mTOR, and degradation of CFTR channels (Yuajit et al., 2014). With the increased need for specific STAT3 inhibitors, the list of available compounds for this purpose will undoubtedly continue to expand.

STAT5 inhibitors

Inhibition of STAT5 may be a promising approach in ADPKD but, so far, specific STAT5 inhibitors have been primarily developed for hematopoietic cancer therapy but mostly have not reached the clinic yet (See table 2 for a comprehensive list of STAT5 inhibitors) (Wingelhofer et al., 2018).

Pimozide is, so far, the only STAT5 inhibitor that is approved by the FDA for neuroleptic treatment. It showed a suppression of STAT5 activity in chronic myeloid leukemia (CML) cells of which the mechanism has not been fully clarified yet (Nelson et al., 2011).

Stafib-2, a small molecule inhibitor, and AC-3–19, a SH2 domain inhibitor, showed more specific STAT5 inhibition. Stafib-2 for example has been shown to selectively inhibit STAT5b activation in human leukemia cells without affecting STAT5a activity (Cumaraswamy et al., 2014; Elumalai et al., 2017). However, their effect was not sufficient to justify clinical trials (Wingelhofer et al., 2018). More recently, AC-4–130 another SH2 domain inhibitor of STAT5 inhibited proliferation of primary human acute myeloid leukemia cells and, furthermore, suppressed growth of an in vivo tumor xenograft in mice indicating a better chance for transfer into clinical studies (Wingelhofer et al., 2018).

Beside small molecule inhibitors, STAT5 activation is additionally approached by activation of peroxisome proliferator-activated receptor γ (PPARγ). The PPARγ activator and antidiabetic drug pioglitazone, for example, seems to be capable of decreasing STAT5 phosphorylation in CML cells (Prost et al., 2015).

Finally, histone deacetylase 6 (HDAC6) has been proposed as another target that may reduce STAT5 transcription and could therefore reduce cyst growth in PKD models (Cebotaru et al., 2016; Pinz et al., 2015).

STAT6/IL-13 inhibitors

Targeting IL-13/STAT6 for PKD therapy is a potentially very promising approach because it may be possible to repurpose several drugs that have already been developed for other indications (See Table 2 for a comprehensive list of STAT6 inhibitors).

The immunosuppressant teriflunomide, the active metabolite of leflunomide, is a broad range tyrosine kinase inhibitor that is approved for treatment of multiple sclerosis (Bar-Or et al., 2014). Our lab has previously shown that teriflunomide inhibits STAT6 activation and ameliorates disease progression in bpk mice (Olsan et al., 2011). Teriflunomide itself may not be viable for PKD therapy because of its many unwanted effects due to inhibition of additional molecular targets. Another small molecule drug reported to inhibit the STAT6 pathway is niflumic acid. Niflumic acid is a chloride channel inhibitor classified as a non-steroidal anti-inflammatory (NSAID) used for treatment of RA. It is thought to suppress IL-13 induced asthma phenotypes in vivo via inhibition of the calcium activated chloride channel, mCLCA3 which subsequently may inhibit JAK/STAT6 activation (Nakano et al., 2006).

Additional small molecule inhibitors of STAT6 have been developed (Miklossy et al., 2013). Of note are three that have been developed for asthma therapy and have shown promising preclinical results, AS1517499, YM-341619, and PM-43I. AS1517499 and YM-341619 both inhibit phosphorylation of STAT6 and PM-43I targets the STAT5/STAT6 Src Homology 2 (SH2) domain (Knight et al., 2018). AS1517499 ameliorates antigen-induced bronchial hypercontractility in mice (Chiba et al., 2009) and blocks IL-13 induced STAT6 phosphorylation in human esophageal fibroblasts and esophageal epithelial cells with minimal cellular toxicity (Cheng et al., 2016). YM-341619 inhibits IL-4 induced STAT6 dependent gene expression and has been shown to inhibit allergen induced Th2 responses in vitro and airway hyperresponsiveness in vivo (Ohga et al., 2008). The most recently developed inhibitor is PM-43I, which was shown to inhibit STAT5 and STAT6 dependent allergic airway disease and reversed preexisting allergic airway disease in mice with no long-term toxicity (Knight et al., 2018).

Another strategy for inhibition of STAT6 is targeting its upstream activator, the IL-4/−13 receptor or the cytokines IL-4 or IL-13. Several anti-IL-13 monoclonal antibodies have been developed and undergone human studies including lebrikizumab and tralokinumab. Lebrikizumab was initially developed to treat asthma and has undergone 11 clinical trials with over 4,000 patients, but after two phase III trials that had conflicting results is being repurposed as a potential novel treatment for atopic dermatitis (AD); (Corren et al., 2011; Hanania et al., 2015; Noonan et al., 2013; Scheerens et al., 2014). Lebrikizumab recently underwent a randomized, placebo-controlled, phase II study in adults with moderate to severe AD and yielded promising results. This study led to significant improvement in symptoms and was well tolerated in these patients (Simpson et al., 2018). Tralokinumab has had a similar path as lebrikizumab. Tralokinumab, an IL-13 neutralizing antibody, was tested in phase 3 studies for asthma but gave disappointing results (Wollenberg et al., 2019; Panettieri et al., 2018) and is also being re-purposed for treating AD. In phase 2b studies for treatment of AD, Tralokinumab treatment was associated with sustained and early improvements in AD symptoms while giving a promising safety and tolerability profile (Wollenberg et al., 2019). Tralokinumab is currently undergoing phase 3 studies for AD (ClinicalTrials.gov Identifiers: NCT03587805, NCT0376157, NCT03556592).

Both of these antibodies are of the IgG isotype and our lab has shown that IgG antibodies do not effectively reach the lumen of renal cysts (Olsan et al., 2015) and would therefore not be expected to be effective for PKD therapy since IL-13 appears to signal mainly via the luminal surface of cyst cells (Olsan et al., 2011). However, our lab has discovered that antibodies in dimeric IgA format are targeted to renal cyst lumens via transcytosis by the polymeric immunoglobulin receptor (pIgR) that is highly expressed on cysts (Olsan et al., 2015). This mechanism can potentially be exploited to target inhibitory antibodies - against IL-13 or other luminal targets - specifically to cysts in PKD which may make effective, long-term therapy possible while avoiding extra-renal effects (Weimbs et al., 2018).

Conclusion

Overall, STAT signaling features a large number of different STAT proteins which are activated by an even larger number of upstream regulators leading to a very broad spectrum of possible downstream effects that, on top of everything else, are also often highly cell-type specific.

Research revealed direct connections between PC1 and STAT signaling, which underlines the significance of STAT signaling in PKD. The general theme is that STAT signaling is generally inactive in healthy adult kidneys but that all components of STAT signaling are generally well expressed. This suggests that the various STAT signaling pathways are generally on standby in the kidney and are meant to quickly respond to changes in circumstances. The main factors that seems to lead to STAT activation are different forms of renal insults leading to injury and necessitating fast immune responses followed by slower injury repair responses. Different renal cell types, especially TECs and immune cells, appear to closely communicate with each other - generally via growth factors and cytokines - to orchestrate activation or downregulation of the various STAT pathways between them. In PKD, many of these processes seem to be disrupted leading to permanent activation of STAT signaling in epithelial and immune cells as well as myofibroblasts. This then appears to lead to a persistent state of a futile repair program that is characterized by epithelial proliferation, fibrosis and a persistent involvement of the immune system. Pharmacological approaches to attempt to disrupt individual STAT signaling pathways may lead to breaking the cycles of cross-activation between renal cells and may be promising for PKD therapy. Since numerous drugs affecting STAT signaling have been, or are being, developed - primarily for cancer therapy - repurposing these drugs for PKD therapy is a timely proposition.

Highlights.

  • Most STAT signaling pathways are activated in kidneys in PKD

  • STAT signaling is regulated by polycystin-1, the protein affected by mutations in ADPKD

  • STAT signaling pathways can be activated in different renal cell types leading to different effects

  • Inhibitors of STAT signaling may be promising for PKD therapy

Acknowledgments

This work was supported by a grant from the National Institutes of Health (NIH; DK109563) and a gift from the Amy P. Goldman Foundation to TW. JAT is the Jarrett Postdoctoral Fellow courtesy of a gift from the Jarrett Family Fund for work in TW’s laboratory. SS is supported by a postdoctoral fellowship of the German Research Foundation (DFG) for work in TWś laboratory. MCL is supported by the German Federal Ministry for Education and Research (NEOCYST consortium; BMBF grants: 01GM1515E and 01GM1903B).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

DECLARATION OF INTERESTS

TW and JAT are inventors on patent applications by UCSB related to PKD. TW serves on the scientific advisory board of, and receives research funding from, Chinook Therapeutics, Inc. TW, JAT and AS are shareholders of Santa Barbara Nutrients, Inc, and serve in managerial positions and/or on the board of directors. Representing the University Hospital of Cologne, MCL serves as a member of an advisory board for Otsuka Pharmaceuticals.

References

  1. Anderson CL (2020). Doubts about the efficacy of tolvaptan for polycystic kidney disease. Clin. Nephrol [DOI] [PubMed] [Google Scholar]
  2. Antczak A, Domańska-Senderowska D, Górski P, Pastuszak-Lewandoska D, Nielepkowicz-Goździńska A, Szewczyk K, Kurmanowska Z, Kiszałkiewicz J, and Brzeziańska-Lasota E (2016). Analysis of changes in expression of IL-4/IL-13/STAT6 pathway and correlation with the selected clinical parameters in patients with atopic asthma. Int J Immunopathol Pharmacol 29, 195–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Avalle L, Camporeale A, Camperi A, and Poli V (2017). STAT3 in cancer: A double edged sword. Cytokine 98, 42–50. [DOI] [PubMed] [Google Scholar]
  4. Bai J, Wu L, Chen X, Wang L, Li Q, Zhang Y, Wu J, Cai G, and Chen X (2018). Suppressor of Cytokine Signaling-1/STAT1 Regulates Renal Inflammation in Mesangial Proliferative Glomerulonephritis Models. Front. Immunol 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Banales JM, Masyuk TV, Bogert PS, Huang BQ, Gradilone SA, Lee S-O, Stroope AJ, Masyuk AI, Medina JF, and LaRusso NF (2008). Hepatic Cystogenesis Is Associated with Abnormal Expression and Location of Ion Transporters and Water Channels in an Animal Model of Autosomal Recessive Polycystic Kidney Disease. Am J Pathol 173, 1637–1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bar-Or A, Pachner A, Menguy-Vacheron F, Kaplan J, and Wiendl H (2014). Teriflunomide and Its Mechanism of Action in Multiple Sclerosis. Drugs 74, 659–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Beebe JD, Liu J-Y, and Zhang J-T (2018). Two decades of research in discovery of anticancer drugs targeting STAT3, how close are we? Pharmacology & Therapeutics 191, 74–91. [DOI] [PubMed] [Google Scholar]
  8. Behbod F, Nagy ZS, Stepkowski SM, Karras J, Johnson CR, Jarvis WD, and Kirken RA (2003). Specific inhibition of Stat5a/b promotes apoptosis of IL-2-responsive primary and tumor-derived lymphoid cells. J. Immunol 171, 3919–3927. [DOI] [PubMed] [Google Scholar]
  9. Bello-Reuss E, Holubec K, and Rajaraman S (2001). Angiogenesis in autosomal-dominant polycystic kidney disease. Kidney International 60, 37–45. [DOI] [PubMed] [Google Scholar]
  10. Bergmann C, Guay-Woodford LM, Harris PC, Horie S, Peters DJM, and Torres VE (2018). Polycystic kidney disease. Nat Rev Dis Primers 4, 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bertuccio CA, Chapin HC, Cai Y, Mistry K, Chauvet V, Somlo S, and Caplan MJ (2009). Polycystin-1 C-terminal Cleavage Is Modulated by Polycystin-2 Expression. J Biol Chem 284, 21011–21026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bhunia AK, Piontek K, Boletta A, Liu L, Qian F, Xu P-N, Germino FJ, and Germino GG (2002). PKD1 Induces p21waf1 and Regulation of the Cell Cycle via Direct Activation of the JAK-STAT Signaling Pathway in a Process Requiring PKD2. Cell 109, 157–168. [DOI] [PubMed] [Google Scholar]
  13. Bienaimé F, Muorah M, Yammine L, Burtin M, Nguyen C, Baron W, Garbay S, Viau A, Broueilh M, Blanc T, et al. (2016). Stat3 Controls Tubulointerstitial Communication during CKD. JASN 27, 3690–3705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Breggia AC, Wojchowski DM, and Himmelfarb J (2008). JAK2/Y343/STAT5 signaling axis is required for erythropoietin-mediated protection against ischemic injury in primary renal tubular epithelial cells. Am. J. Physiol. Renal Physiol 295, F1689–1695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bullock SL, Johnson TM, Bao Q, Hughes RC, Winyard PJ, and Woolf AS (2001). Galectin-3 modulates ureteric bud branching in organ culture of the developing mouse kidney. J. Am. Soc. Nephrol 12, 515–523. [DOI] [PubMed] [Google Scholar]
  16. Cassini MF, Kakade VR, Kurtz E, Sulkowski P, Glazer P, Torres R, Somlo S, and Cantley LG (2018). Mcp1 Promotes Macrophage-Dependent Cyst Expansion in Autosomal Dominant Polycystic Kidney Disease. JASN 29, 2471–2481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Castelli M, De Pascalis C, Distefano G, Ducano N, Oldani A, Lanzetti L, and Boletta A (2015). Regulation of the microtubular cytoskeleton by Polycystin-1 favors focal adhesions turnover to modulate cell adhesion and migration. BMC Cell Biol 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cebotaru L, Liu Q, Yanda MK, Boinot C, Outeda P, Huso DL, Watnick T, Guggino WB, and Cebotaru V (2016). Inhibition of histone deacetylase 6 activity reduces cyst growth in polycystic kidney disease. Kidney Int 90, 90–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chauvet V, Tian X, Husson H, Grimm DH, Wang T, Hieseberger T, Igarashi P, Bennett AM, Ibraghimov-Beskrovnaya O, Somlo S, et al. (2004). Mechanical stimuli induce cleavage and nuclear translocation of the polycystin-1 C terminus. J Clin Invest 114, 1433–1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen S-C, and Kuo P-L (2016). The Role of Galectin-3 in the Kidneys. IJMS 17, 565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chen J, Chen J-K, Conway EM, and Harris RC (2013a). Survivin mediates renal proximal tubule recovery from AKI. J. Am. Soc. Nephrol 24, 2023–2033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chen W, Lu Y, Chen G, and Huang S (2013b). Molecular evidence of cryptotanshinone for treatment and prevention of human cancer. Anticancer Agents Med Chem 13, 979–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chen Z, Zhu R, Zheng J, Chen C, Huang C, Ma J, Xu C, Zhai W, and Zheng J (2017). Cryptotanshinone inhibits proliferation yet induces apoptosis by suppressing STAT3 signals in renal cell carcinoma. Oncotarget 8, 50023–50033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cheng E, Zhang X, Wilson KS, Wang DH, Park JY, Huo X, Yu C, Zhang Q, Spechler SJ, and Souza RF (2016). JAK-STAT6 Pathway Inhibitors Block Eotaxin-3 Secretion by Epithelial Cells and Fibroblasts from Esophageal Eosinophilia Patients: Promising Agents to Improve Inflammation and Prevent Fibrosis in EoE. PLoS ONE 11, e0157376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cheon H, and Stark GR (2009). Unphosphorylated STAT1 prolongs the expression of interferon-induced immune regulatory genes. Proc Natl Acad Sci U S A 106, 9373–9378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cheon H, Yang J, and Stark GR (2011). The Functions of Signal Transducers and Activators of Transcriptions 1 and 3 as Cytokine-Inducible Proteins. J Interferon Cytokine Res 31, 33–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chiba Y, Todoroki M, Nishida Y, Tanabe M, and Misawa M (2009). A Novel STAT6 Inhibitor AS1517499 Ameliorates Antigen-Induced Bronchial Hypercontractility in Mice. Am J Respir Cell Mol Biol 41, 516–524. [DOI] [PubMed] [Google Scholar]
  28. Chiu MG, Johnson TM, Woolf AS, Dahm-Vicker EM, Long DA, Guay-Woodford L, Hillman KA, Bawumia S, Venner K, Hughes RC, et al. (2006). Galectin-3 Associates with the Primary Cilium and Modulates Cyst Growth in Congenital Polycystic Kidney Disease. The American Journal of Pathology 169, 1925–1938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Cornec-Le Gall E, Alam A, and Perrone RD (2019). Autosomal dominant polycystic kidney disease. Lancet 393, 919–935. [DOI] [PubMed] [Google Scholar]
  30. Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, Arron JR, Harris JM, Scheerens H, Wu LC, Su Z, et al. (2011). Lebrikizumab Treatment in Adults with Asthma. N Engl J Med 365, 1088–1098. [DOI] [PubMed] [Google Scholar]
  31. Creamer BA, Sakamoto K, Schmidt JW, Triplett AA, Moriggl R, and Wagner K-U (2010). Stat5 Promotes Survival of Mammary Epithelial Cells through Transcriptional Activation of a Distinct Promoter in Akt1. Mol Cell Biol 30, 2957–2970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Cui X, Zhang L, Luo J, Rajasekaran A, Hazra S, Cacalano N, and Dubinett SM (2007). Unphosphorylated STAT6 contributes to constitutive cyclooxygenase-2 expression in human non-small cell lung cancer. Oncogene 26, 4253–4260. [DOI] [PubMed] [Google Scholar]
  33. Cui Y, Riedlinger G, Miyoshi K, Tang W, Li C, Deng C-X, Robinson GW, and Hennighausen L (2004). Inactivation of Stat5 in mouse mammary epithelium during pregnancy reveals distinct functions in cell proliferation, survival, and differentiation. Mol. Cell. Biol 24, 8037–8047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Cumaraswamy AA, Lewis AM, Geletu M, Todic A, Diaz DB, Cheng XR, Brown CE, Laister RC, Muench D, Kerman K, et al. (2014). Nanomolar-Potency Small Molecule Inhibitor of STAT5 Protein. ACS Med Chem Lett 5, 1202–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Dalagiorgou G, Piperi C, Adamopoulos C, Georgopoulou U, Gargalionis AN, Spyropoulou A, Zoi I, Nokhbehsaim M, Damanaki A, Deschner J, et al. (2017). Mechanosensor polycystin-1 potentiates differentiation of human osteoblastic cells by upregulating Runx2 expression via induction of JAK2/STAT3 signaling axis. Cell. Mol. Life Sci 74, 921–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Dong J, Wang QX, Zhou CY, Ma XF, and Zhang YC (2007). Activation of the STAT1 signalling pathway in lupus nephritis in MRL/lpr mice. Lupus 16, 101–109. [DOI] [PubMed] [Google Scholar]
  37. Douguet D, Patel A, and Honoré E (2019). Structure and function of polycystins: insights into polycystic kidney disease. Nature Reviews Nephrology 15, 412–422. [DOI] [PubMed] [Google Scholar]
  38. Du Y, Zhang W, Liu S, Feng X, Gao F, and Liu Q (2018). S3I-201 ameliorates tubulointerstitial lesion of the kidneys in MRL/lpr mice. Biochem. Biophys. Res. Commun 503, 177–180. [DOI] [PubMed] [Google Scholar]
  39. Dube S, Matam T, Yen J, Mang HE, Dagher PC, Hato T, and Sutton TA (2017). Endothelial STAT3 Modulates Protective Mechanisms in a Mouse Ischemia-Reperfusion Model of Acute Kidney Injury. J Immunol Res 2017, 4609502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Durbin JE, Hackenmiller R, Simon MC, and Levy DE (1996). Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease. Cell 84, 443–450. [DOI] [PubMed] [Google Scholar]
  41. Elliott J, Zheleznova NN, and Wilson PD (2011). c-Src inactivation reduces renal epithelial cell-matrix adhesion, proliferation, and cyst formation. Am J Physiol Cell Physiol 301, C522–C529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Elumalai N, Berg A, Rubner S, Blechschmidt L, Song C, Natarajan K, Matysik J, and Berg T (2017). Rational development of Stafib-2: a selective, nanomolar inhibitor of the transcription factor STAT5b. Sci Rep 7, 819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Erickson KF, Chertow GM, and Goldhaber-Fiebert JD (2013). Cost-Effectiveness of Tolvaptan in Autosomal Dominant Polycystic Kidney Disease. Ann Intern Med 159, 382–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Estrada CC, Paladugu P, Guo Y, Pace J, Revelo MP, Salant DJ, Shankland SJ, D’Agati VD, Mehrotra A, Cardona S, et al. (2018). Krüppel-like factor 4 is a negative regulator of STAT3-induced glomerular epithelial cell proliferation. JCI Insight 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Ferguson AD, Larsen NA, Howard T, Pollard H, Green I, Grande C, Cheung T, Garcia-Arenas R, Cowen S, Wu J, et al. (2011). Structural basis of substrate methylation and inhibition of SMYD2. Structure 19, 1262–1273. [DOI] [PubMed] [Google Scholar]
  46. Ferreli C, Lai C, August S, Buggy Y, Kumar P, Brownlow N, Parker P, Friedmann PS, Ardern-Jones M, Pickard C, et al. (2017). STAT4 expression and activation is increased during mitosis in vitro and in vivo in skin- and mucosa-derived cell types: implications in neoplastic and inflammatory skin diseases. Journal of the European Academy of Dermatology and Venereology 31, 1663–1673. [DOI] [PubMed] [Google Scholar]
  47. Fielding CA, McLoughlin RM, McLeod L, Colmont CS, Najdovska M, Grail D, Ernst M, Jones SA, Topley N, and Jenkins BJ (2008). IL-6 regulates neutrophil trafficking during acute inflammation via STAT3. J. Immunol 181, 2189–2195. [DOI] [PubMed] [Google Scholar]
  48. Fitzgibbon WR, Dang Y, Bunni MA, Baicu CF, Zile MR, Mullick AE, and Saigusa T (2018). Attenuation of accelerated renal cystogenesis in Pkd1 mice by renin-angiotensin system blockade. Am. J. Physiol. Renal Physiol 314, F210–F218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Fonseca-Camarillo G, Furuzawa-Carballeda J, Granados J, and Yamamoto-Furusho JK (2014). Expression of interleukin (IL)-19 and IL-24 in inflammatory bowel disease patients: a cross-sectional study: Expression of IL-19 and IL-24 in IBD patients. Clin Exp Immunol 177, 64–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Fragiadaki M, Lannoy M, Themanns M, Maurer B, Leonhard WN, Peters DJM, Moriggl R, and Ong ACM (2017). STAT5 drives abnormal proliferation in autosomal dominant polycystic kidney disease. Kidney Int 91, 575–586. [DOI] [PubMed] [Google Scholar]
  51. Friedbichler K, Themanns M, Mueller KM, Schlederer M, Kornfeld J-W, Terracciano LM, Kozlov AV, Haindl S, Kenner L, Kolbe T, et al. (2012). Growth-hormone-induced signal transducer and activator of transcription 5 signaling causes gigantism, inflammation, and premature death but protects mice from aggressive liver cancer. Hepatology 55, 941–952. [DOI] [PubMed] [Google Scholar]
  52. van Gastel MDA, and Torres VE (2017). Polycystic Kidney Disease and the Vasopressin Pathway. ANM 70, 43–50. [DOI] [PubMed] [Google Scholar]
  53. Gattone VH, Wang X, Harris PC, and Torres VE (2003). Inhibition of renal cystic disease development and progression by a vasopressin V2 receptor antagonist. Nat Med 9, 1323–1326. [DOI] [PubMed] [Google Scholar]
  54. Genovese MC, van Vollenhoven RF, Pacheco-Tena C, Zhang Y, and Kinnman N (2016). VX-509 (Decernotinib), an Oral Selective JAK-3 Inhibitor, in Combination With Methotrexate in Patients With Rheumatoid Arthritis. Arthritis & Rheumatology (Hoboken, N.J.) 68, 46–55. [DOI] [PubMed] [Google Scholar]
  55. Genovese MC, Fleischmann R, Combe B, Hall S, Rubbert-Roth A, Zhang Y, Zhou Y, Mohamed M-EF, Meerwein S, and Pangan AL (2018). Safety and efficacy of upadacitinib in patients with active rheumatoid arthritis refractory to biologic disease-modifying anti-rheumatic drugs (SELECT-BEYOND): a double-blind, randomised controlled phase 3 trial. The Lancet 391, 2513–2524. [DOI] [PubMed] [Google Scholar]
  56. Graziani A, Poteser M, Heupel W-M, Schleifer H, Krenn M, Drenckhahn D, Romanin C, Baumgartner W, and Groschner K (2010). Cell-Cell Contact Formation Governs Ca 2+ Signaling by TRPC4 in the Vascular Endothelium: EVIDENCE FOR A REGULATORY TRPC4-β-CATENIN INTERACTION. J. Biol. Chem 285, 4213–4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Groner B, Fritsche M, Stöcklin E, Berchtold S, Merkle C, Moriggl R, and Pfitzner E (2000). Regulation of the trans-activation potential of STAT5 through its DNA-binding activity and interactions with heterologous transcription factors. Growth Hormone & IGF Research 10, S15–S20. [DOI] [PubMed] [Google Scholar]
  58. Hanania NA, Noonan M, Corren J, Korenblat P, Zheng Y, Fischer SK, Cheu M, Putnam WS, Murray E, Scheerens H, et al. (2015). Lebrikizumab in moderate-to-severe asthma: pooled data from two randomised placebo-controlled studies. Thorax 70, 748–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Haricharan S, and Li Y (2014). STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol 382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Harrison CN, Schaap N, Vannucchi AM, Kiladjian J-J, Tiu RV, Zachee P, Jourdan E, Winton E, Silver RT, Schouten HC, et al. (2017). Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): a single-arm, open-label, non-randomised, phase 2, multicentre study. The Lancet Haematology 4, e317–e324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Henderson NC, Mackinnon AC, Farnworth SL, Kipari T, Haslett C, Iredale JP, Liu F-T, Hughes J, and Sethi T (2008). Galectin-3 Expression and Secretion Links Macrophages to the Promotion of Renal Fibrosis. The American Journal of Pathology 172, 288–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Heppler LN, and Frank DA (2017). Targeting Oncogenic Transcription Factors: Therapeutic Implications of Endogenous STAT Inhibitors. Trends in Cancer 3, 816–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Hillmer EJ, Zhang H, Li HS, and Watowich SS (2016). STAT3 signaling in immunity. Cytokine Growth Factor Rev 31, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Huang J-S, Chuang L-Y, Guh JY, Huang Y-J, and Hsu M-S (2007). Antioxidants attenuate high glucose-induced hypertrophic growth in renal tubular epithelial cells. American Journal of Physiology. Renal Physiology 293, NaN–NaN. [DOI] [PubMed] [Google Scholar]
  65. Huang J-S, Lee Y-H, Chuang L-Y, Guh J-Y, and Hwang J-Y (2015). Cinnamaldehyde and Nitric Oxide Attenuate Advanced Glycation End Products-Induced the JAK/STAT Signaling in Human Renal Tubular Cells. Journal of Cellular Biochemistry 116, 1028–1038. [DOI] [PubMed] [Google Scholar]
  66. Huen SC, Huynh L, Marlier A, Lee Y, Moeckel GW, and Cantley LG (2015). GM-CSF Promotes Macrophage Alternative Activation after Renal Ischemia/Reperfusion Injury. J. Am. Soc. Nephrol 26, 1334–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Humphreys BD, Lin S-L, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, Valerius MT, McMahon AP, and Duffield JS (2010). Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am. J. Pathol 176, 85–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Ihle JN (1996). STATs: Signal Transducers and Activators of Transcription. Cell 84, 331–334. [DOI] [PubMed] [Google Scholar]
  69. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, and Neilson EG (2002). Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Invest 110, 341–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Jain N, Zhang T, Fong SL, Lim CP, and Cao X (1998). Repression of Stat3 activity by activation of mitogen-activated protein kinase (MAPK). Oncogene 17, 3157–3167. [DOI] [PubMed] [Google Scholar]
  71. Jing Y, Wu M, Zhang D, Chen D, Yang M, Mei S, He L, Gu J, Qi N, Fu L, et al. (2018). Triptolide delays disease progression in an adult rat model of polycystic kidney disease through the JAK2-STAT3 pathway. American Journal of Physiology-Renal Physiology 315, F479–F486. [DOI] [PubMed] [Google Scholar]
  72. Karihaloo A, Koraishy F, Huen SC, Lee Y, Merrick D, Caplan MJ, Somlo S, and Cantley LG (2011). Macrophages Promote Cyst Growth in Polycystic Kidney Disease. JASN 22, 1809–1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Kaviratne M, Hesse M, Leusink M, Cheever AW, Davies SJ, McKerrow JH, Wakefield LM, Letterio JJ, and Wynn TA (2004). IL-13 Activates a Mechanism of Tissue Fibrosis That Is Completely TGF-β Independent. J Immunol 173, 4020–4029. [DOI] [PubMed] [Google Scholar]
  74. Kazansky AV, Kabotyanski EB, Wyszomierski SL, Mancini MA, and Rosen JM (1999). Differential effects of prolactin and src/abl kinases on the nuclear translocation of STAT5B and STAT5A. J. Biol. Chem 274, 22484–22492. [DOI] [PubMed] [Google Scholar]
  75. Kemmner S, Bachmann Q, Steiger S, Lorenz G, Honarpisheh M, Foresto-Neto O, Wang S, Carbajo-Lozoya J, Alt V, Schulte C, et al. (2019). STAT1 regulates macrophage number and phenotype and prevents renal fibrosis after ischemia-reperfusion injury. Am. J. Physiol. Renal Physiol 316, F277–F291. [DOI] [PubMed] [Google Scholar]
  76. Kleczko EK, Marsh KH, Tyler LC, Furgeson SB, Bullock BL, Altmann CJ, Miyazaki M, Gitomer BY, Harris PC, Weiser-Evans MCM, et al. (2018). CD8+ T cells modulate autosomal dominant polycystic kidney disease progression. Kidney Int 94, 1127–1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kleinschmidt MA, Streubel G, Samans B, Krause M, and Bauer U-M (2008). The protein arginine methyltransferases CARM1 and PRMT1 cooperate in gene regulation. Nucleic Acids Res 36, 3202–3213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Knight JM, Mandal P, Morlacchi P, Mak G, Li E, Madison M, Landers C, Saxton B, Felix E, Gilbert B, et al. (2018). Small molecule targeting of the STAT5/6 Src homology 2 (SH2) domains to inhibit allergic airway disease. J. Biol. Chem 293, 10026–10040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kontzias A, Kotlyar A, Laurence A, Changelian P, and O’Shea JJ (2012). Jakinibs: A New Class of Kinase Inhibitors in Cancer and Autoimmune Disease. Curr Opin Pharmacol 12, 464–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kormann R, Kavvadas P, Placier S, Vandermeersch S, Dorison A, Dussaule J-C, Chadjichristos CE, Prakoura N, and Chatziantoniou C (2019). Periostin Promotes Cell Proliferation and Macrophage Polarization to Drive Repair after AKI JASN ASN.2019020113. [DOI] [PMC free article] [PubMed]
  81. Kremer JM, Bloom BJ, Breedveld FC, Coombs JH, Fletcher MP, Gruben D, Krishnaswami S, Burgos-Vargas R, Wilkinson B, Zerbini CAF, et al. (2009). The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo-controlled phase IIa trial of three dosage levels of CP-690,550 versus placebo. Arthritis Rheum 60, 1895–1905. [DOI] [PubMed] [Google Scholar]
  82. Kulkarni OP, Hartter I, Mulay SR, Hagemann J, Darisipudi MN, Kumar Vr S, Romoli S, Thomasova D, Ryu M, Kobold S, et al. (2014). Toll-like receptor 4-induced IL-22 accelerates kidney regeneration. J. Am. Soc. Nephrol 25, 978–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kuratsune M, Masaki T, Hirai T, Kiribayashi K, Yokoyama Y, Arakawa T, Yorioka N, and Kohno N (2007). Signal transducer and activator of transcription 3 involvement in the development of renal interstitial fibrosis after unilateral ureteral obstruction. Nephrology (Carlton) 12, 565–571. [DOI] [PubMed] [Google Scholar]
  84. Kwak M, Hong C, Myeong J, Park EYJ, Jeon J-H, and So I (2018). Gαi-mediated TRPC4 activation by polycystin-1 contributes to endothelial function via STAT1 activation. Sci Rep 8, 3480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Leaman D, Pisharody S, W Flickinger T, Commane M, Schlessinger J, M Kerr I, Levy D, and R Stark G (1996). Roles of JAKs in Activation of STATs and Stimulation of c-fos Gene Expression by Epidermal Growth Factor. Molecular and Cellular Biology 16, 369–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. LeBleu VS, Taduri G, O’Connell J, Teng Y, Cooke VG, Woda C, Sugimoto H, and Kalluri R (2013). Origin and function of myofibroblasts in kidney fibrosis. Nat. Med 19, 1047–1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Lee C, Raz R, Gimeno R, Gertner R, Wistinghausen B, Takeshita K, DePinho RA, and Levy DE (2002). STAT3 is a negative regulator of granulopoiesis but is not required for G-CSF-dependent differentiation. Immunity 17, 63–72. [DOI] [PubMed] [Google Scholar]
  88. Leonhard WN, van der Wal A, Novalic Z, Kunnen SJ, Gansevoort RT, Breuning MH, de Heer E, and Peters DJM (2011). Curcumin inhibits cystogenesis by simultaneous interference of multiple signaling pathways: in vivo evidence from a Pkd1-deletion model. American Journal of Physiology-Renal Physiology 300, F1193–F1202. [DOI] [PubMed] [Google Scholar]
  89. Li H-P, Geng L, Burrow CR, and Wilson PD (1999). Identification of Phosphorylation Sites in the PKD1-Encoded Protein C-Terminal Domain. Biochemical and Biophysical Research Communications 259, 356–363. [DOI] [PubMed] [Google Scholar]
  90. Liang H, Zhang Z, Yan J, Wang Y, Hu Z, Mitch WE, and Wang Y (2017). The IL-4 receptor α has a critical role in bone marrow-derived fibroblast activation and renal fibrosis. Kidney Int 92, 1433–1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Liongue C, and Ward AC (2013). Evolution of the JAK-STAT pathway. JAK-STAT 2, e22756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Liu B, Li C, Liu Z, Dai Z, and Tao Y (2012). Increasing extracellular matrix collagen level and MMP activity induces cyst development in polycystic kidney disease. BMC Nephrol 13, 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Low SH, Vasanth S, Larson CH, Mukherjee S, Sharma N, Kinter MT, Kane ME, Obara T, and Weimbs T (2006). Polycystin-1, STAT6, and P100 function in a pathway that transduces ciliary mechanosensation and is activated in polycystic kidney disease. Dev. Cell 10, 57–69. [DOI] [PubMed] [Google Scholar]
  94. Martinez-Martinez E, Ibarrola J, Calvier L, Fernandez-Celis A, Leroy C, Cachofeiro V, Rossignol P, and Lopez-Andres N (2016). Galectin-3 Blockade Reduces Renal Fibrosis in Two Normotensive Experimental Models of Renal Damage. PLoS One 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Masaki T, Foti R, Hill PA, Ikezumi Y, Atkins RC, and Nikolic-Paterson DJ (2003). Activation of the ERK pathway precedes tubular proliferation in the obstructed rat kidney. Kidney Int 63, 1256–1264. [DOI] [PubMed] [Google Scholar]
  96. Melillo JA, Song L, Bhagat G, Blazquez AB, Plumlee CR, Lee C, Berin C, Reizis B, and Schindler C (2010). Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function. J. Immunol 184, 2638–2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Meraz MA, White JM, Sheehan KC, Bach EA, Rodig SJ, Dighe AS, Kaplan DH, Riley JK, Greenlund AC, Campbell D, et al. (1996). Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK-STAT signaling pathway. Cell 84, 431–442. [DOI] [PubMed] [Google Scholar]
  98. Merrick D, Bertuccio CA, Chapin HC, Lal M, Chauvet V, and Caplan MJ (2014). Polycystin-1 cleavage and the regulation of transcriptional pathways. Pediatr Nephrol 29, 505–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Mesa RA, Yasothan U, and Kirkpatrick P (2012). Ruxolitinib [DOI] [PubMed]
  100. Mesarosova L, Ochodnicky P, Leemans JC, Florquin S, Krenek P, and Klimas J (2017). High glucose induces HGF-independent activation of Met receptor in human renal tubular epithelium. Journal of Receptors and Signal Transduction 37, 535–542. [DOI] [PubMed] [Google Scholar]
  101. Metcalf D, Mifsud S, Di Rago L, Nicola NA, Hilton DJ, and Alexander WS (2002). Polycystic kidneys and chronic inflammatory lesions are the delayed consequences of loss of the suppressor of cytokine signaling-1 (SOCS-1). Proc. Natl. Acad. Sci. U.S.A 99, 943–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Miklossy G, Hilliard TS, and Turkson J (2013). Therapeutic modulators of STAT signalling for human diseases. Nat Rev Drug Discov 12, 611–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Mosser DM, and Zhang X (2008). Interleukin-10: new perspectives on an old cytokine. Immunological Reviews 226, 205–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Müller R-U, and Benzing T (2018). Management of autosomal-dominant polycystic kidney disease-state-of-the-art. Clin Kidney J 11, i2–i13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Nakano T, Inoue H, Fukuyama S, Matsumoto K, Matsumura M, Tsuda M, Matsumoto T, Aizawa H, and Nakanishi Y (2006). Niflumic Acid Suppresses Interleukin-13-induced Asthma Phenotypes. Am J Respir Crit Care Med 173, 1216–1221. [DOI] [PubMed] [Google Scholar]
  106. Nan J, Wang Y, Yang J, and Stark GR (2018). IRF9 and unphosphorylated STAT2 cooperate with NF-κB to drive IL6 expression. Proc Natl Acad Sci USA 115, 3906–3911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Nast R, Staab J, and Meyer T (2019). Gene Activation by the Cytokine-Driven Transcription Factor STAT1. Gene Regulation [Google Scholar]
  108. Nechemia-Arbely Y, Barkan D, Pizov G, Shriki A, Rose-John S, Galun E, and Axelrod JH (2008). IL-6/IL-6R axis plays a critical role in acute kidney injury. J. Am. Soc. Nephrol 19, 1106–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Nefedova Y, Huang M, Kusmartsev S, Bhattacharya R, Cheng P, Salup R, Jove R, and Gabrilovich D (2004). Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J. Immunol 172, 464–474. [DOI] [PubMed] [Google Scholar]
  110. Nelson EA, Walker SR, Weisberg E, Bar-Natan M, Barrett R, Gashin LB, Terrell S, Klitgaard JL, Santo L, Addorio MR, et al. (2011). The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood 117, 3421–3429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Nims N, Vassmer D, and Maser RL (2003). Transmembrane Domain Analysis of Polycystin-1, the Product of the Polycystic Kidney Disease-1 (PKD1) Gene: Evidence for 11 Membrane-Spanning Domains. Biochemistry 42, 13035–13048. [DOI] [PubMed] [Google Scholar]
  112. Noonan M, Korenblat P, Mosesova S, Scheerens H, Arron JR, Zheng Y, Putnam WS, Parsey MV, Bohen SP, and Matthews JG (2013). Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. Journal of Allergy and Clinical Immunology 132, 567–574.e12. [DOI] [PubMed] [Google Scholar]
  113. O’Brown ZK, Van Nostrand EL, Higgins JP, and Kim SK (2015). The Inflammatory Transcription Factors NFκB, STAT1 and STAT3 Drive Age-Associated Transcriptional Changes in the Human Kidney. PLoS Genet 11, e1005734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Ohga K, Kuromitsu S, Takezawa R, Numazaki M, Ishikawa J, Nagashima S, and Shimizu Y (2008). YM-341619 suppresses the differentiation of spleen T cells into Th2 cells in vitro, eosinophilia, and airway hyperresponsiveness in rat allergic models. European Journal of Pharmacology 590, 409–416. [DOI] [PubMed] [Google Scholar]
  115. Olayioye MA, Beuvink I, Horsch K, Daly JM, and Hynes NE (1999). ErbB Receptor-induced Activation of Stat Transcription Factors Is Mediated by Src Tyrosine Kinases. J. Biol. Chem 274, 17209–17218. [DOI] [PubMed] [Google Scholar]
  116. Olsan EE, Mukherjee S, Wulkersdorfer B, Shillingford JM, Giovannone AJ, Todorov G, Song X, Pei Y, and Weimbs T (2011). Signal transducer and activator of transcription-6 (STAT6) inhibition suppresses renal cyst growth in polycystic kidney disease. Proceedings of the National Academy of Sciences 108, 18067–18072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Olsan EE, Matsushita T, Rezaei M, and Weimbs T (2015). Exploitation of the Polymeric Immunoglobulin Receptor for Antibody Targeting to Renal Cyst Lumens in Polycystic Kidney Disease. J. Biol. Chem 290, 15679–15686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Olsan EE, West JD, Torres JA, Doerr N, and Weimbs T (2018). Identification of targets of IL-13 and STAT6 signaling in polycystic kidney disease. American Journal of Physiology-Renal Physiology 315, F86–F96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Ong ACM, and Harris PC (2015). A polycystin-centric view of cyst formation and disease: the polycystins revisited. Kidney Int 88, 699–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Orellana SA, Sweeney WE, Neff CD, and Avner ED (1995). Epidermal growth factor receptor expression is abnormal in murine polycystic kidney. Kidney International 47, 490–499. [DOI] [PubMed] [Google Scholar]
  121. O’Shea JJ, Holland SM, and Staudt LM (2013). JAKs and STATs in immunity, immunodeficiency, and cancer. N. Engl. J. Med 368, 161–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, and Laurence A (2015). The JAK-STAT Pathway: Impact on Human Disease and Therapeutic Intervention. Annu. Rev. Med 66, 311–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. O’Sullivan LA, Liongue C, Lewis RS, Stephenson SEM, and Ward AC (2007). Cytokine receptor signaling through the Jak-Stat-Socs pathway in disease. Mol. Immunol 44, 2497–2506. [DOI] [PubMed] [Google Scholar]
  124. Ouyang W, Rutz S, Crellin NK, Valdez PA, and Hymowitz SG (2011). Regulation and Functions of the IL-10 Family of Cytokines in Inflammation and Disease. Annu. Rev. Immunol 29, 71–109. [DOI] [PubMed] [Google Scholar]
  125. Padovano V, and Caplan MJ (2018). The Polycystin Complex Reveals Its Complexity. Biochemistry 57, 6917–6918. [DOI] [PubMed] [Google Scholar]
  126. Panettieri RA, Sjöbring U, Péterffy A, Wessman P, Bowen K, Piper E, Colice G, and Brightling CE (2018). Tralokinumab for severe, uncontrolled asthma (STRATOS 1 and STRATOS 2): two randomised, double-blind, placebo-controlled, phase 3 clinical trials. The Lancet Respiratory Medicine 6, 511–525. [DOI] [PubMed] [Google Scholar]
  127. Pang M, Ma L, Gong R, Tolbert E, Mao H, Ponnusamy M, Chin YE, Yan H, Dworkin LD, and Zhuang S (2010). A novel STAT3 inhibitor, S3I-201, attenuates renal interstitial fibroblast activation and interstitial fibrosis in obstructive nephropathy. Kidney Int 78, 257–268. [DOI] [PubMed] [Google Scholar]
  128. Park HJ, Li J, Hannah R, Biddie S, Leal-Cervantes AI, Kirschner K, Flores Santa Cruz D, Sexl V, Göttgens B, and Green AR (2016). Cytokine-induced megakaryocytic differentiation is regulated by genome-wide loss of a uSTAT transcriptional program. EMBO J 35, 580–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Park S-J, Nakagawa T, Kitamura H, Atsumi T, Kamon H, Sawa S-I, Kamimura D, Ueda N, Iwakura Y, Ishihara K, et al. (2004). IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J. Immunol 173, 3844–3854. [DOI] [PubMed] [Google Scholar]
  130. Patera F, Cudzich-Madry A, Huang Z, and Fragiadaki M (2019). Renal expression of JAK2 is high in polycystic kidney disease and its inhibition reduces cystogenesis. Sci Rep 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Peda JD, Salah SM, Wallace DP, Fields PE, Grantham CJ, Fields TA, and Swenson-Fields KI (2016). Autocrine IL-10 activation of the STAT3 pathway is required for pathological macrophage differentiation in polycystic kidney disease. Dis Model Mech 9, 1051–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Perry ST, Buck MD, Lada SM, Schindler C, and Shresta S (2011). STAT2 Mediates Innate Immunity to Dengue Virus in the Absence of STAT1 via the Type I Interferon Receptor. PLoS Pathog 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Pinz S, Unser S, Buob D, Fischer P, Jobst B, and Rascle A (2015). Deacetylase inhibitors repress STAT5-mediated transcription by interfering with bromodomain and extraterminal (BET) protein function. Nucleic Acids Res 43, 3524–3545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Potočnjak I, Broznić D, Kindl M, Kropek M, Vladimir-Knežević S, and Domitrović R (2017). Stevia and stevioside protect against cisplatin nephrotoxicity through inhibition of ERK1/2, STAT3, and NF-κB activation. Food and Chemical Toxicology 107, 215–225. [DOI] [PubMed] [Google Scholar]
  135. Prost S, Relouzat F, Spentchian M, Ouzegdouh Y, Saliba J, Massonnet G, Beressi J-P, Verhoeyen E, Raggueneau V, Maneglier B, et al. (2015). Erosion of the chronic myeloid leukaemia stem cell pool by PPARγ agonists. Nature 525, 380–383. [DOI] [PubMed] [Google Scholar]
  136. Qian F, Boletta A, Bhunia AK, Xu H, Liu L, Ahrabi AK, Watnick TJ, Zhou F, and Germino GG (2002). Cleavage of polycystin-1 requires the receptor for egg jelly domain and is disrupted by human autosomal-dominant polycystic kidney disease 1-associated mutations. Proceedings of the National Academy of Sciences 99, 16981–16986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Raman A, Parnell SC, Zhang Y, Reif GA, Dai Y, Khanna A, Daniel E, White C, Vivian JL, and Wallace DP (2018). Periostin overexpression in collecting ducts accelerates renal cyst growth and fibrosis in polycystic kidney disease. American Journal of Physiology-Renal Physiology 315, F1695–F1707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Ravichandran K, Ozkok A, Wang Q, Mullick AE, and Edelstein CL (2015). Antisense-mediated angiotensinogen inhibition slows polycystic kidney disease in mice with a targeted mutation in Pkd2. American Journal of Physiology - Renal Physiology 308, F349–F357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Rawlings JS, Rosler KM, and Harrison DA (2004). The JAK/STAT signaling pathway. Journal of Cell Science 117, 1281–1283. [DOI] [PubMed] [Google Scholar]
  140. Richards WG, Sweeney WE, Yoder BK, Wilkinson JE, Woychik RP, and Avner ED (1998). Epidermal growth factor receptor activity mediates renal cyst formation in polycystic kidney disease. J Clin Invest 101, 935–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Said E, Zaitone SA, Eldosoky M, and Elsherbiny NM (2018). Nifuroxazide, a STAT3 inhibitor, mitigates inflammatory burden and protects against diabetes-induced nephropathy in rats. Chemico-Biological Interactions 281, 111–120. [DOI] [PubMed] [Google Scholar]
  142. Saigusa T, Dang Y, Mullick AE, Yeh ST, Zile MR, Baicu CF, and Bell PD (2016). Suppressing angiotensinogen synthesis attenuates kidney cyst formation in a Pkd1 mouse model. FASEB J 30, 370–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Sandovici M, Henning RH, van Goor H, Helfrich W, de Zeeuw D, and Deelman LE (2008). Systemic gene therapy with interleukin-13 attenuates renal ischemia-reperfusion injury. Kidney Int 73, 1364–1373. [DOI] [PubMed] [Google Scholar]
  144. Schaefer F, Mekahli D, Emma F, Gilbert RD, Bockenhauer D, Cadnapaphornchai MA, Shi L, Dandurand A, Sikes K, and Shoaf SE (2019). Tolvaptan use in children and adolescents with autosomal dominant polycystic kidney disease: rationale and design of a two-part, randomized, double-blind, placebo-controlled trial. Eur J Pediatr 178, 1013–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Scheerens H, Arron JR, Zheng Y, Putnam WS, Erickson RW, Choy DF, Harris JM, Lee J, Jarjour NN, and Matthews JG (2014). The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge. Clin Exp Allergy 44, 38–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Schrier RW, Abebe KZ, Perrone RD, Torres VE, Braun WE, Steinman TI, Winklhofer FT, Brosnahan G, Czarnecki PG, Hogan MC, et al. (2014). Blood pressure in early autosomal dominant polycystic kidney disease. N. Engl. J. Med 371, 2255–2266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, and O’Shea JJ (2017). JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov 16, 843–862. [DOI] [PubMed] [Google Scholar]
  148. Shi Y, Du C, Zhang Y, Ren Y, Hao J, Zhao S, Yao F, and Duan H (2010). Suppressor of cytokine signaling-1 ameliorates expression of MCP-1 in diabetic nephropathy. Am. J. Nephrol 31, 380–388. [DOI] [PubMed] [Google Scholar]
  149. Shimoda K, van Deursen J, Sangster MY, Sarawar SR, Carson RT, Tripp RA, Chu C, Quelle FW, Nosaka T, Vignali DA, et al. (1996). Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene. Nature 380, 630–633. [DOI] [PubMed] [Google Scholar]
  150. Shuai K (2006). Regulation of cytokine signaling pathways by PIAS proteins. Cell Research 16, 196–202. [DOI] [PubMed] [Google Scholar]
  151. Silva CM (2004). Role of STATs as downstream signal transducers in Src family kinase-mediated tumorigenesis. Oncogene 23, 8017–8023. [DOI] [PubMed] [Google Scholar]
  152. Simpson EL, Flohr C, Eichenfield LF, Bieber T, Sofen H, Taïeb A, Owen R, Putnam W, Castro M, DeBusk K, et al. (2018). Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). Journal of the American Academy of Dermatology 78, 863–871.e11. [DOI] [PubMed] [Google Scholar]
  153. Song CJ, Zimmerman KA, Henke SJ, and Yoder BK (2017). Inflammation and Fibrosis in Polycystic Kidney Disease In Kidney Development and Disease, Miller RK, ed. (Cham: Springer International Publishing; ), pp. 323–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Strati P, Abdelrahim M, Selamet U, Page VD, Pierce SA, Verstovsek S, and Abudayyeh A (2019). Ruxolitinib therapy is associated with improved renal function in patients with primary myelofibrosis. Ann Hematol 98, 1611–1616. [DOI] [PubMed] [Google Scholar]
  155. Streets AJ, Wagner BE, Harris PC, Ward CJ, and Ong ACM (2009). Homophilic and heterophilic polycystin 1 interactions regulate E-cadherin recruitment and junction assembly in MDCK cells. J Cell Sci 122, 1410–1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Sweeney WE, and Avner ED (2006). Molecular and cellular pathophysiology of autosomal recessive polycystic kidney disease (ARPKD). Cell Tissue Res 326, 671–685. [DOI] [PubMed] [Google Scholar]
  157. Sweeney WE, Hamahira K, Sweeney J, Garcia-Gatrell M, Frost P, and Avner ED (2003). Combination treatment of PKD utilizing dual inhibition of EGF-receptor activity and ligand bioavailability. Kidney International 64, 1310–1319. [DOI] [PubMed] [Google Scholar]
  158. Sweeney WE, von Vigier RO, Frost P, and Avner ED (2008). Src Inhibition Ameliorates Polycystic Kidney Disease. J Am Soc Nephrol 19, 1331–1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Sweeney WE, Frost P, and Avner ED (2017). Tesevatinib ameliorates progression of polycystic kidney disease in rodent models of autosomal recessive polycystic kidney disease. World J Nephrol 6, 188–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Swenson-Fields KI, Vivian CJ, Salah SM, Peda JD, Davis BM, van Rooijen N, Wallace DP, and Fields TA (2013). Macrophages promote polycystic kidney disease progression. Kidney International 83, 855–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Takakura A, Nelson EA, Haque N, Humphreys BD, Zandi-Nejad K, Frank DA, and Zhou J (2011). Pyrimethamine inhibits adult polycystic kidney disease by modulating STAT signaling pathways. Hum Mol Genet 20, 4143–4154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Takeda K, Tanaka T, Shi W, Matsumoto M, Minami M, Kashiwamura S, Nakanishi K, Yoshida N, Kishimoto T, and Akira S (1996). Essential role of Stat6 in IL-4 signalling. Nature 380, 627–630. [DOI] [PubMed] [Google Scholar]
  163. Takeda K, Noguchi K, Shi W, Tanaka T, Matsumoto M, Yoshida N, Kishimoto T, and Akira S (1997). Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc. Natl. Acad. Sci. U.S.A 94, 3801–3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Takeda K, Clausen BE, Kaisho T, Tsujimura T, Terada N, Förster I, and Akira S (1999). Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity 10, 39–49. [DOI] [PubMed] [Google Scholar]
  165. Talbot JJ, Shillingford JM, Vasanth S, Doerr N, Mukherjee S, Kinter MT, Watnick T, and Weimbs T (2011). Polycystin-1 regulates STAT activity by a dual mechanism. Proc. Natl. Acad. Sci. U.S.A 108, 7985–7990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Talbot JJ, Song X, Wang X, Rinschen MM, Doerr N, LaRiviere WB, Schermer B, Pei YP, Torres VE, and Weimbs T (2014). The Cleaved Cytoplasmic Tail of Polycystin-1 Regulates Src-Dependent STAT3 Activation. J Am Soc Nephrol 25, 1737–1748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Tao J, Mariani L, Eddy S, Maecker H, Kambham N, Mehta K, Hartman J, Wang W, Kretzler M, and Lafayette RA (2018). JAK-STAT signaling is activated in the kidney and peripheral blood cells of patients with focal segmental glomerulosclerosis. Kidney Int 94, 795–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Taylor PC, Keystone EC, van der Heijde D, Weinblatt ME, del Carmen Morales L, Reyes Gonzaga J, Yakushin S, Ishii T, Emoto K, Beattie S, et al. (2017). Baricitinib versus Placebo or Adalimumab in Rheumatoid Arthritis. New England Journal of Medicine 376, 652–662. [DOI] [PubMed] [Google Scholar]
  169. Tefferi A, and Pardanani A (2011). Serious Adverse Events During Ruxolitinib Treatment Discontinuation in Patients With Myelofibrosis. Mayo Clin Proc 86, 1188–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Tesar V, Ciechanowski K, Pei Y, Barash I, Shannon M, Li R, Williams JH, Levisetti M, Arkin S, and Serra A (2017). Bosutinib versus Placebo for Autosomal Dominant Polycystic Kidney Disease. J. Am. Soc. Nephrol 28, 3404–3413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Togawa H, Nakanishi K, Mukaiyama H, Hama T, Shima Y, Sako M, Miyajima M, Nozu K, Nishii K, Nagao S, et al. (2010). Epithelial-to-mesenchymal transition in cyst lining epithelial cells in an orthologous PCK rat model of autosomal-recessive polycystic kidney disease. American Journal of Physiology-Renal Physiology 300, F511–F520. [DOI] [PubMed] [Google Scholar]
  172. Torres VE, and Harris PC (2019). Progress in the understanding of polycystic kidney disease. Nature Reviews Nephrology 15, 70–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Torres JA, Rezaei M, Broderick C, Lin L, Wang X, Hoppe B, Benjamin D Cowley J, Savica V, Torres VE, Khan S, et al. (2019). Crystal deposition triggers tubule dilation that accelerates cystogenesis in polycystic kidney disease. J Clin Invest [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Torres VE, Chapman AB, Perrone RD, Bae KT, Abebe KZ, Bost JE, Miskulin DC, Steinman TI, Braun W, Winklhofer FT, et al. (2012a). The HALT Polycystic Kidney Disease Trials – Analysis of baseline parameters. Kidney Int 81, 577–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Torres VE, Chapman AB, Devuyst O, Gansevoort RT, Grantham JJ, Higashihara E, Perrone RD, Krasa HB, Ouyang J, and Czerwiec FS (2012b). Tolvaptan in Patients with Autosomal Dominant Polycystic Kidney Disease. New England Journal of Medicine 367, 2407–2418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Torres VE, Abebe KZ, Chapman AB, Schrier RW, Braun WE, Steinman TI, Winklhofer FT, Brosnahan G, Czarnecki PG, Hogan MC, et al. (2014). Angiotensin blockade in late autosomal dominant polycystic kidney disease. N. Engl. J. Med 371, 2267–2276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Torres VE, Chapman AB, Devuyst O, Gansevoort RT, Perrone RD, Koch G, Ouyang J, McQuade RD, Blais JD, Czerwiec FS, et al. (2017). Tolvaptan in Later-Stage Autosomal Dominant Polycystic Kidney Disease. N. Engl. J. Med 377, 1930–1942. [DOI] [PubMed] [Google Scholar]
  178. Udy GB, Towers RP, Snell RG, Wilkins RJ, Park SH, Ram PA, Waxman DJ, and Davey HW (1997). Requirement of STAT5b for sexual dimorphism of body growth rates and liver gene expression. Proc. Natl. Acad. Sci. U.S.A 94, 7239–7244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Van Dyken SJ, and Locksley RM (2013). Interleukin-4- and Interleukin-13-Mediated Alternatively Activated Macrophages: Roles in Homeostasis and Disease. Annu. Rev. Immunol 31, 317–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Van Nguyen T, Angkasekwinai P, Dou H, Lin F-M, Lu L-S, Cheng J, Chin YE, Dong C, and Yeh ETH (2012). SUMO-Specific Protease 1 Is Critical for Early Lymphoid Development through Regulation of STAT5 Activation. Mol Cell 45, 210–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, et al. (2012). A Double-Blind Placebo-Controlled Trial of Ruxolitinib for Myelofibrosis. N Engl J Med 366, 799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Viau A, Bienaimé F, Lukas K, Todkar AP, Knoll M, Yakulov TA, Hofherr A, Kretz O, Helmstädter M, Reichardt W, et al. (2018). Cilia-localized LKB 1 regulates chemokine signaling, macrophage recruitment, and tissue homeostasis in the kidney. EMBO J 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Vignais ML, Sadowski HB, Watling D, Rogers NC, and Gilman M (1996). Platelet-derived growth factor induces phosphorylation of multiple JAK family kinases and STAT proteins. Mol Cell Biol 16, 1759–1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Villarino A, Laurence A, Robinson GW, Bonelli M, Dema B, Afzali B, Shih H-Y, Sun H-W, Brooks SR, Hennighausen L, et al. (2016). Signal transducer and activator of transcription 5 (STAT5) paralog dose governs T cell effector and regulatory functions. ELife 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Villarino AV, Kanno Y, and O’Shea JJ (2017). Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol 18, 374–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Wallace DP (2019). Periostin in the Kidney In Periostin, Kudo A, ed. (Singapore: Springer Singapore; ), pp. 99–112. [Google Scholar]
  187. Wallace DP, Quante MT, Reif GA, Nivens E, Ahmed F, Hempson SJ, Blanco G, and Yamaguchi T (2008). Periostin induces proliferation of human autosomal dominant polycystic kidney cells through α V -integrin receptor. American Journal of Physiology-Renal Physiology 295, F1463–F1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Wallace DP, White C, Savinkova L, Nivens E, Reif GA, Pinto CS, Raman A, Parnell SC, Conway SJ, and Fields TA (2014). Periostin promotes renal cyst growth and interstitial fibrosis in polycystic kidney disease. Kidney International 85, 845–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Wang H, Yang Y, Sharma N, Tarasova NI, Timofeeva OA, Winkler-Pickett RT, Tanigawa S, and Perantoni AO (2010). STAT1 activation regulates proliferation and differentiation of renal progenitors. Cell. Signal 22, 1717–1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Wang J, Ouyang C, Chen X, Fu B, Lu Y, and Hong Q (2008). STAT3 inhibits apoptosis of human renal tubular epithelial cells induced by ATP depletion/recovery. Nephron Exp. Nephrol 108, e11–18. [DOI] [PubMed] [Google Scholar]
  191. Wang X, Shaw S, Amiri F, Eaton DC, and Marrero MB (2002). Inhibition of the Jak/STAT signaling pathway prevents the high glucose-induced increase in tgf-beta and fibronectin synthesis in mesangial cells. Diabetes 51, 3505–3509. [DOI] [PubMed] [Google Scholar]
  192. Wang Y, Evans JT, Rodriguez F, Fields P, Mueller C, Chitnis T, Khoury SJ, and Bynoe MS (2009). A tale of two STAT6 knock out mice in the induction of experimental autoimmune encephalomyelitis. J. Neuroimmunol 206, 76–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Wang Y-Z, Wharton W, Garcia R, Kraker A, Jove R, and Pledger WJ (2000). Activation of Stat3 preassembled with platelet-derived growth factor β receptors requires Src kinase activity. Oncogene 19, 2075–2085. [DOI] [PubMed] [Google Scholar]
  194. Watanabe EH, Amaral AG, and Onuchic LF (2019). Periostin and polycystic kidney disease: more pieces in the puzzle. American Journal of Physiology-Renal Physiology 316, F159–F161. [DOI] [PubMed] [Google Scholar]
  195. Weimbs T (2018). Are Cyst-Associated Macrophages in Polycystic Kidney Disease the Equivalent to TAMs in Cancer? JASN 29, 2447–2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Weimbs T, Shillingford JM, Torres J, Kruger SL, and Bourgeois BC (2018). Emerging targeted strategies for the treatment of autosomal dominant polycystic kidney disease. Clinical Kidney Journal 11, i27–i38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Wiezel D, Assadi MH, Landau D, Troib A, Kachko L, Rabkin R, and Segev Y (2014). Impaired renal growth hormone JAK/STAT5 signaling in chronic kidney disease. Nephrol. Dial. Transplant 29, 791–799. [DOI] [PubMed] [Google Scholar]
  198. Wilson PD, Geng L, Li X, and Burrow CR (1999). The PKD1 gene product, “polycystin-1,” is a tyrosine-phosphorylated protein that colocalizes with alpha2beta1-integrin in focal clusters in adherent renal epithelia. Lab. Invest 79, 1311–1323. [PubMed] [Google Scholar]
  199. Wingelhofer B, Maurer B, Heyes EC, Cumaraswamy AA, Berger-Becvar A, de Araujo ED, Orlova A, Freund P, Ruge F, Park J, et al. (2018). Pharmacologic inhibition of STAT5 in acute myeloid leukemia. Leukemia 32, 1135–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Winthrop KL (2017). The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol 13, 234–243. [DOI] [PubMed] [Google Scholar]
  201. Winyard JD Epithelial Galectin-3 During Human Nephrogenesis and Childhood Cystic Diseases. Journal of the American Society of Nephrology 11. [DOI] [PubMed] [Google Scholar]
  202. Wollenberg A, Howell MD, Guttman-Yassky E, Silverberg JI, Kell C, Ranade K, Moate R, and van der Merwe R (2019). Treatment of atopic dermatitis with tralokinumab, an anti-IL-13 mAb. Journal of Allergy and Clinical Immunology 143, 135–141. [DOI] [PubMed] [Google Scholar]
  203. Woodward OM, Li Y, Yu S, Greenwell P, Wodarczyk C, Boletta A, Guggino WB, and Qian F (2010). Identification of a Polycystin-1 Cleavage Product, P100, That Regulates Store Operated Ca2+ Entry through Interactions with STIM1. PLoS One 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Wu M, Chen M, Jing Y, Gu J, Mei S, Yao Q, Zhou J, Yang M, Sun L, Wang W, et al. (2016). The C-terminal tail of polycystin-1 regulates complement factor B expression by signal transducer and activator of transcription 1. American Journal of Physiology-Renal Physiology 310, F1284–F1294. [DOI] [PubMed] [Google Scholar]
  205. Wurster AL, Tanaka T, and Grusby MJ (2000). The biology of Stat4 and Stat6. Oncogene 19, 2577–2584. [DOI] [PubMed] [Google Scholar]
  206. Wynn TA (2004). Fibrotic disease and the TH1/TH2 paradigm. Nat Rev Immunol 4, 583–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Xiong H, Su W-Y, Liang Q-C, Zhang Z-G, Chen H-M, Du W, Chen Y-X, and Fang J-Y (2009). Inhibition of STAT5 induces G1 cell cycle arrest and reduces tumor cell invasion in human colorectal cancer cells. Lab. Invest 89, 717–725. [DOI] [PubMed] [Google Scholar]
  208. Xu M-J, Feng D, Wang H, Guan Y, Yan X, and Gao B (2014). IL-22 ameliorates renal ischemia-reperfusion injury by targeting proximal tubule epithelium. J. Am. Soc. Nephrol 25, 967–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Xu Z, Zou C, Yu W, Xu S, Huang L, Khan Z, Wang J, Liang G, and Wang Y (2019). Inhibition of STAT3 activation mediated by toll-like receptor 4 attenuates angiotensin II-induced renal fibrosis and dysfunction. British Journal of Pharmacology 0, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Yan J, Zhang Z, Yang J, Mitch WE, and Wang Y (2015). JAK3/STAT6 Stimulates Bone Marrow-Derived Fibroblast Activation in Renal Fibrosis. J. Am. Soc. Nephrol 26, 3060–3071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Yang J, and Stark GR (2008). Roles of unphosphorylated STATs in signaling. Cell Res 18, 443–451. [DOI] [PubMed] [Google Scholar]
  212. Yang J, Lin S-C, Chen G, He L, Hu Z, Chan L, Trial J, Entman ML, and Wang Y (2013). Adiponectin promotes monocyte-to-fibroblast transition in renal fibrosis. J. Am. Soc. Nephrol 24, 1644–1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Yang Y, Chen M, Zhou J, Lv J, Song S, Fu L, Chen J, Yang M, and Mei C (2018). Interactions between Macrophages and Cyst-Lining Epithelial Cells Promote Kidney Cyst Growth in Pkd1 -Deficient Mice. JASN 29, 2310–2325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Ye H, Wang X, Constans MM, Sussman CR, Chebib FT, Irazabal MV, Young WF, Harris PC, Kirschner LS, and Torres VE (2017). The regulatory 1α subunit of protein kinase A modulates renal cystogenesis. Am J Physiol Renal Physiol 313, F677–F686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Yokota N, Burne-Taney M, Racusen L, and Rabb H (2003). Contrasting roles for STAT4 and STAT6 signal transduction pathways in murine renal ischemia-reperfusion injury. Am. J. Physiol. Renal Physiol 285, F319–325. [DOI] [PubMed] [Google Scholar]
  216. Yoshimura A, Naka T, and Kubo M (2007). SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol 7, 454–465. [DOI] [PubMed] [Google Scholar]
  217. Yu H, Pardoll D, and Jove R (2009). STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 9, 798–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Yuajit C, Muanprasat C, Gallagher A-R, Fedeles SV, Kittayaruksakul S, Homvisasevongsa S, Somlo S, and Chatsudthipong V (2014). Steviol retards renal cyst growth through reduction of CFTR expression and inhibition of epithelial cell proliferation in a mouse model of polycystic kidney disease. Biochemical Pharmacology 88, 412–421. [DOI] [PubMed] [Google Scholar]
  219. Yukawa K, Kishino M, Goda M, Liang X-M, Kimura A, Tanaka T, Bai T, Owada-Makabe K, Tsubota Y, Ueyama T, et al. (2005). STAT6 deficiency inhibits tubulointerstitial fibrosis in obstructive nephropathy. Int. J. Mol. Med 15, 225–230. [PubMed] [Google Scholar]
  220. Zaki OS, Safar MM, Ain-Shoka AA, and Rashed LA (2018). A Novel Role of a Chemotherapeutic Agent in a Rat Model of Endotoxemia: Modulation of the STAT-3 Signaling Pathway. Inflammation 41, 20–32. [DOI] [PubMed] [Google Scholar]
  221. Zhang J, Zhao D, Na N, Li H, Miao B, Hong L, and Huang Z (2018). Renoprotective effect of erythropoietin via modulation of the STAT6/MAPK/NF-κB pathway in ischemia/reperfusion injury after renal transplantation. Int. J. Mol. Med 41, 25–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Zhang M-Z, Wang X, Wang Y, Niu A, Wang S, Zou C, and Harris RC (2017a). IL-4/IL-13-mediated polarization of renal macrophages/dendritic cells to an M2a phenotype is essential for recovery from acute kidney injury. Kidney International 91, 375–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Zhang Y, Chen Y, Yun H, Liu Z, Su M, and Lai R (2017b). STAT1β enhances STAT1 function by protecting STAT1α from degradation in esophageal squamous cell carcinoma. Cell Death & Disease 8, e3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Zhong M, Henriksen MA, Takeuchi K, Schaefer O, Liu B, ten Hoeve J, Ren Z, Mao X, Chen X, Shuai K, et al. (2005). Implications of an antiparallel dimeric structure of nonphosphorylated STAT1 for the activation-inactivation cycle. PNAS 102, 3966–3971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Zhou H, Hasni SA, Perez P, Tandon M, Jang S-I, Zheng C, Kopp JB, Austin H, Balow JE, Alevizos I, et al. (2013). miR-150 promotes renal fibrosis in lupus nephritis by downregulating SOCS1. J. Am. Soc. Nephrol 24, 1073–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Zhuang S (2013). Regulation of STAT Signaling by Acetylation. Cell Signal 25, 1924–1931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Zimmerman KA, Song CJ, Li Z, Lever JM, Crossman DK, Rains A, Aloria EJ, Gonzalez NM, Bassler JR, Zhou J, et al. (2019). Tissue-Resident Macrophages Promote Renal Cystic Disease. JASN 30, 1841–1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Zimmerman KA, Huang J, He L, Revell DZ, Li Z, Hsu J-S, Fitzgibbon WR, Hazard ES, Hardiman G, Mrug M, et al. (2020). Interferon Regulatory Factor 5 in Resident Macrophage Promotes Polycystic Kidney Disease. Kidney360 10.34067/KID.0001052019. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES