Signaling Pathways in Glioblastoma Cancer Stem Cells: A Role of Stat3 as a Potential Therapeutic Target

Review Article

Austin J Cancer Clin Res 2015;2(2): 1030.

Signaling Pathways in Glioblastoma Cancer Stem Cells: A Role of Stat3 as a Potential Therapeutic Target

Hiroshi Kanno1,2*, Shigeta Miyake1,2 and Satoshi Nakanowatari2

1Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Japan

2Department of Neurosurgery, Yokosuka City Hospital, Japan

*Corresponding author: Hiroshi Kanno, Department of Neurosurgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.

Received: February 02, 2015; Accepted: March 26,2015; Published: April 03, 2015

Abstract

Glioblastoma cancer stem cells (GCSCs) play an important role in proliferation, invasion, progression, immune evasion, and resistance to radiation in glioblastoma. Their signaling pathways including receptor tyrosine kinase, Akt, MARK, Wnt, Notch, Hedgehog, and JAK/STAT pathways are complicated, but STAT-3 is a convergence point in several important signaling pathways and contributes to the tumor progression by promoting cell proliferation, cell cycle progression, the inhibition of apoptosis, and tissue invasion. Therefore, STAT- 3 is a candidate of therapeutic target of GCSCs. STAT3 is activated through tyrosine phosphorylation by various cytokines and growth factors. STAT-3 is tyrosine phosphorylated by three types of kinases such as receptor tyrosine kinases, JAK family members, and oncogenic kinases including Src and Bcl- Alb. Tyrosine phosphorylated STAT-3 dimerizes and translocates to the nucleus. Active STAT-3 dimers bind to consequences in the promoters of genes such as Bcl-2, Bcl-xL, Mcl-1, and cyclin D1. After induction of target gene expression, multiple STAT-3-endogenous negative regulators such as SOCS3, VHL, and PIAS3 attenuate STAT-3 signaling, and similarly STAT-3 exogenous negative regulators such as pharmacologic JAK inhibitors, dobesilate, and decoy oligonucleotides attenuate STAT-3 activity. In GCSCs, STAT-3 plays a role as a molecular hub in several important signaling pathways that control proliferation, cell cycle progression, anti-apoptosis, invasion, angiogenesis and immune evasion. Therefore, STAT-3 has great potential as a therapeutic target.

Keywords: Signaling pathway; Glioblastoma cancer stem cells; STAT-3; Therapeutic target

Introduction

The cancer stem cells are defined as a cell population residing in tumors, having self -renewal capacity and dividing to give rise to the variety of tumor cells [1,2]. Those cells can reconstitute both the tumor cell hierarchy and the clinical disease state in vivo after xenotransplantation [3]. Although the existence of cancer stem cells in human leukemia is established [1,3], those cells were later discovered to exist in various solid tumors including breast [4], colon [5], lung [6], liver cancers [7], and glioblastoma [8].

Glioblastoma is the most common and lethal brain tumor that shows aggressive natures, with a median overall survival (OS) of less than15 months after diagnosis. The standard first-line treatment includes resection as much as possible, followed by concurrent radioand chemotherapy with temozolomide (TMZ), and then 6–12 months of chemotherapy with TMZ. Despite the treatment, recurrence is universal. Glioblastoma is thought to contain a population of selfrenewing glioblastoma cancer stem cells (GCSCs) that contributes to treatment resistance [9]. GCSCs are functionally defined with selfrenewal measured by serial neurosphere assay in vitro and tumor forming capacity through serial transplantation. GCSCs have been shown to differentiate into astrocytes, oligodendrocytes and neurons [10]. Commonly used GCSC markers are as follows: cell surface: CD133, CD15, CD44, CXCR4; integrin alpha–6; protein (cytoplasmic & nuclear): netin, Musashi-1, Bmi-1; transcriptional factor: Sox2, enzyme, ALDH1 [9]. Recently, researches on glioblastoma have focused on the elucidation of various signaling pathways in GCSCs, particularly aberrant activation. Here, we describe the signaling pathway in GCSCs, particularly focusing on the signal transducer and activator of transcription-3 (STAT-3) as a therapeutic target [10].

Up and downstream signaling pathways of STAT-3 in GSCSs

The activation of several signaling pathways including receptor tyrosine kinase, Akt, MARK, Wnt, Notch, Hedgehog, and JAK/STAT pathways is involved in the progression and proliferation in GCSCs [11]. Among those various signaling pathways in GCSCs, activator of JAK/STAT signal transduction pathway is aberrantly activated in glioblastoma likewise in other solid tumors including breast, lung, ovarian, pancreatic, skin, prostate cancers [12]. STAT-3 contributes to the tumor progression by promoting cell proliferation, cell cycle progression, the inhibition of apoptosis, and tissue invasion [13]. In addition, STAT-3 plays an important role in wound healing, T-cell development, and immune evasion [13,14]. The activation of STAT-3 is implicated in not only controlling critical cellular events involved in tumorigenesis, cell cycle progression, angiogenesis [15], and immune evasion, but also maintaining “stemness” such as self-renewal capacity, ability of differentiation to various tumor cells, and reconstitution of the clinical state in vivo after xeno-transplantation [13,16]. Many tumor-derived cell lines require STAT proteins, particularly STAT-3, to maintain a transformed phenotype [17], and a constitutively active STAT-3 mutant, STAT-3C, was sufficient to transform benign tumor cells to malignant tumor cells, and these transformed cells could form tumors in nude mice [18]. Furthermore, a dominant-negative mutant of STAT-3 blocked transformation by v-src [19]. STAT-3 is activated through phosphorylation of tyrosine 705 that initiates to form a complex composed of two phosphorylated STAT-3 monomers (pSTAT-3). pSTAT-3 homodimers translocate to the nucleus and bind DNA. STAT3 is activated through tyrosine phosphorylation by various cytokines such as interleukin-6 (IL-6) cytokine families (IL-6, oncostatin M, and leukemia inhibitory factor), IL-4, IL-10, IL- 11, and IL-23 and growth factors including platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF) and epidermal growth factor (EGF) [20]. In addition, STAT-3 is tyrosine phosphorylated by three types of kinases such as receptor tyrosine kinases including EGFR, FGFR, and PDGFR, Janus kinase (JAK) family members, and oncogenic kinases including Src and Bcl-Alb [21]. Active STAT-3 dimers bind to consequences in the promoters of genes such as Bcl-2, Bcl-xL, Mcl-1, p21WAF1/CIP1, and cyclin D1 [22]. As STAT-3 affects transcription of genes involved in cell cycle and antiapoptosis, regulation of STAT-3 activity is required to prevent malignant transformation of cells. After induction of target gene expression, multiple STAT-3-endogenous negative regulators attenuate STAT-3 signaling. Suppressors of cytokine signaling (SOCS)-3 and von Hipple-Lindau (VHL) proteins down-regulate the upstream kinase activity responsible for STAT-3 phosphorylation [23,24], while the protein inhibitors of activated STAT (PIAS)3 inhibits STAT-3 directly [25]. SOCS-3 and VHL proteins belong to BC-box protein families that contain a BC-box motif corresponding to the binding site of elongin BC [26], while PIAS3 belongs to PIAS families that contain a zinc ring finger domain, an NH2-terminal LXXLL motif, a COOH-terminal acidic domain, a serine/threoninerich domain and PINIT motif involved in the nuclear retention [27]. SOCS-3 and VHL inhibit JAK activation and subsequently attenuate STAT-3 signal transduction in a classic negative feedback loop in the cytoplasm [28], whereas PIAS3 inhibits STAT-3 DNA binding the nucleus and exhibit E3-SUMO (small ubiquitin-like modifier) ligase activity and SUMOylate a variety of transcription factors including p53, c-Jun, and c-Myb [29].

The Notch signaling pathway is involved in cell fate decisions during normal development and in the genesis of glioblastoma [30]. The downstream effects of Notch signaling are highly tissue and time dependent, and Notch has been implicated both in the maintenance of neural progenitors and in the generation of glia during development of the brain [31]. It has been reported previously that in the developing central nervous system, there is cross-talk between the Notch and STAT3 pathways because STAT-3 binds to adjacent site in the Notch1 promoter [32]. Notch pathway genes were up-regulated in GCSCs by constitutive activation of STAT-3 [33], but in contrast the activation and phosphorylation of STAT3 is mediated by the direct binding of several Notch effectors to STAT3 [34].

Inhibiting STAT-3 activity in tumor cells with either dominantnegative or STAT-3 inhibition by STAT-3 siRNA resulted in increased expression of pro-inflammatory mediators including IFN-β, TNF-α, and IL-6 [35]. STAT-3 signaling is dampened by protein tyrosine phosphatases, such as the SH2-domain containing tyrosine phosphatase family (SHP-1, SHP-2), which downregulate STAT-3 activation directly by phosphorylating active STAT-3 complexes [36] (Figure 1).

Inhibitory regulators of STAT-3

STAT-3 is a promising target for GCSCs, not only because it is a convergence point in several important signaling pathways that promote proliferation [15], invasion [37], immune evasion [14,38,39], anti-apoptosis [40], anti-autophagy [41], formation of peritumoral edema [42] and maintenance of “stemness” [15] but also because aberrant STAT-3 activation results from upstream dysregulation [43] (Table 1). There are two approaches to inhibit STAT-3: (1) through exogenous regulators such as RNA interference and chemical inhibitors, and (2) through endogenous regulators such as PIAS3, SOCS-3, and VHL [13] (Table 2). Direct STAT-3 inhibition has been achieved with dominant negative constructs, oligonucleotides, or, phosphopeptidic agents that mimic the native tyrosine 705 containing binding sequence or non-native STAT3-binding sequences [10]. Some platinum compounds interfere with STAT-3 activation and abrogate signaling mediated by constitutively active STAT-3 [44]. Direct inhibition of STAT-3 activity using RNA interference induced apoptosis and inhibited survival [17]. Among decoy oligonucleotides of STAT-3 binding site, G-quartets, having competitive inhibitory structures comprised of guanine-rich oligonucleotides, inhibited STAT-3 binding to endogenous gene promoters and subsequently attenuated STAT-3-induced gene expression by competitively binding activated STAT-3 [45]. STX-0119 (a N-[2-(1,3,4-oxadiazolyl)]-4- quinolinecarboxamide derivative) inhibited STAT3 dimerization and suppressed the growth of transplanted tumors of GCSCs. STX-0119 inhibited proliferation and sphere formation in GCSCs by downregulating the gene expression of STAT3-target genes including cyclin D1, survivin, Bcl-xL, c-myc, MMP2, VEGF and HIF-1α [46]. Similarly, S31-201 inhibited STAT-3 homodimer formation by inhibiting STAT-3 DNA binding, and showed the antitumor activity by inhibiting STAT-3 induced gene expression [47]. In addition, small molecule, nonphosphorylated STAT-3 inhibitor, 31 (SH-4-54) that strongly binds to STAT3 protein. SG-4-54 effectively suppresses STAT3 phosphorylation and its downstream transcriptional target genes and potently kills GCSCs. Moreover, in vivo, SH-4-54 inhibited pSTAT-3 in vivo and potently controlled glioblastoma tumor growth with exhibition of blood-brain barrier permeability [10].