4,794
Views
60
CrossRef citations to date
0
Altmetric
Review

STAT3 inhibitors for cancer therapy

Have all roads been explored?

, , &
Article: e22882 | Published online: 01 Jan 2013

Abstract

The signal transducer and activator of transcription STAT3 is a transcription factor which plays a key role in normal cell growth and is constitutively activated in about 70% of solid and hematological cancers. Activated STAT3 is phosphorylated on tyrosine and forms a dimer through phosphotyrosine/src homology 2 (SH2) domain interaction. The dimer enters the nucleus via interaction with importins and binds target genes. Inhibition of STAT3 results in the death of tumor cells, this indicates that it is a valuable target for anticancer strategies; a view that is corroborated by recent findings of activating mutations within the gene. Yet, there is still only a small number of STAT3 direct inhibitors; in addition, the high similarity of STAT3 with STAT1, another STAT family member mostly oriented toward apoptosis, cell death and defense against pathogens, requires that STAT3-inhibitors have no effect on STAT1. Specific STAT3 direct inhibitors consist of SH2 ligands, including G quartet oligodeoxynucleotides (ODN) and small molecules, they induce cell death in tumor cells in which STAT3 is activated. STAT3 can also be inhibited by decoy ODNs (dODN), which bind STAT3 and induce cell death. A specific STAT3 dODN which does not interfere with STAT1-mediated interferon-induced cell death has been designed pointing to the STAT3 DBD as a target for specific inhibition. Comprehensive analysis of this region is in progress in the laboratory to design DBD-targeting STAT3 inhibitors with STAT3/STAT1 discriminating ability.

Central Role of STAT3 in Tumors

STAT3 belongs to a family of transcription factors (TFs) comprising STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and STAT6.Citation1 Like STAT5, STAT3 was found to play an important role in cell growth,Citation2 and its activation has been described in nearly 70% of solid and hematological tumors,Citation3,Citation4 giving good reason for a search for specific direct inhibitors,Citation5,Citation6 of which there are unfortunately only a few, and none in the clinic to this day. STAT3 comprises several distinct functional domains including: an N-terminal domain containing an oligomerization and a coiled-coil domain, a DNA binding domain (DBD), a linker domain, a Src homology 2 (SH2) domain involved in the interaction of two monomers via phosphotyrosine 705 resulting in dimerization and a C-terminal transactivation domain (see ). STAT3 activation occurs following cytokine- or growth factor-receptor activation; it involves phosphorylation within the cytoplasm, dimerization and nuclear transferCitation7 (). Nuclear transfer of STAT3 requires nuclear localization signals (NLS) which are in the coiled-coil domain (comprising arginines 214 and 215Citation8) and in the dimer-dependent DBD (comprising arginines 414 and 417Citation9). The NLSs interact with importin αs, yet which of the five importin αs (α1, α3, α4, α5 or α7) actually carries STAT3 is still debated,Citation9,Citation10 the complex interacts with importin β and is carried through the nuclear pore complex (NPC) (). While arginines 214 and 215 appear to be the major importin-binding site, arginines 414 and 417 are thought to be required for STAT3 to adopt the proper conformation for importin binding.Citation9 Several studies have shown that STAT3 cycling is probably somewhat more complicated. Unphosphorylated forms of STAT3 can enter the nucleus and stimulate transcription of a subset of gene targets, apparently via interaction with the TF NFκB.Citation11 However, whether unphosphorylated STAT3 interacts on its own with importins for nuclear entry is not entirely clear: tyrosine 705-mutated STAT3 can shuttle to the nucleusCitation12 and phosphotyrosine 705/SH2-independent STAT3 dimers were shown to enter the nucleus (but more slowly than phosphorylated STAT3 dimers)Citation13 (). Interestingly, in the case of STAT1, unphosphorylated monomers enter the nucleus through direct interaction with the NPC proteins nucleoporins, not with importinsCitation14 and unphosphorylated STAT1 dimers bind DNA with a 200-fold lower affinity than phosphorylated STAT1 dimers;Citation15 in fact, single-molecule imaging showed that interferon (IFN)-γ-activated STAT1 has a reduced mobility and resides longer in the nucleus.Citation16 In any case, the nucleo-cytoplasmic shuttling of STAT3 is a major step of the activation process leading to increased transcriptional activity, suggesting that nuclear transfer of STAT3 per se can be a target for inhibition.

Figure 1. STAT3 with DNA-consensus sequence. STAT3 monomer showing the N-terminal coiled-coil domain, the DBD (half site), the SH2 domain and the C-terminal domain. Basic residues are blue and acid ones are red. The STAT3 crystal coordinates were downloaded from the protein data bank (PDB, file: 1BG1) and analyzed using the chimera program.Citation73 Note that the model shown comprises residues 136 to 716;Citation74 hence, the protein’s N-terminal and C-terminal domains (comprising the transactivation domain) are missing; the coordinates corresponding to the cDNA strand were missing in the model and had to be reconstructed.Citation65

Figure 1. STAT3 with DNA-consensus sequence. STAT3 monomer showing the N-terminal coiled-coil domain, the DBD (half site), the SH2 domain and the C-terminal domain. Basic residues are blue and acid ones are red. The STAT3 crystal coordinates were downloaded from the protein data bank (PDB, file: 1BG1) and analyzed using the chimera program.Citation73 Note that the model shown comprises residues 136 to 716;Citation74 hence, the protein’s N-terminal and C-terminal domains (comprising the transactivation domain) are missing; the coordinates corresponding to the cDNA strand were missing in the model and had to be reconstructed.Citation65

Figure 2. STAT3 activation. The transcription factor STAT3 is present in a latent inactive non-phosphorylated form in the cytoplasm. Activated cytokine receptors activate the kinases JAK, which phosphorylate tyrosines located in the cytoplasmic portion of the cytokine receptors creating STAT-binding motifs. Once bound to these motifs, STAT3 becomes in turn phosphorylated by the JAKs. The phospho STAT3 dimers (colored in pink) enter the nuclei and bind STAT3 target genes; note that the DNA-bound dimer is drawn differently to indicate the STAT3 conformational change suggested by molecular dynamics simulations.Citation22 STAT3 can also be activated by tyrosine kinases of the Src family (SFK), the SFKs can themselves be activated by G protein-coupled receptors (GPCR) or growth factor receptors, the growth factor receptors (including EGF-receptors and VEGF-receptors) can also phosphorylate STAT3. The DBD is created by the phosphotyrosine 705/SH2-mediated dimer. There are also monomers, and dimers with non-phosphorylated STAT3, which can shuttle between the cytoplasm and the nucleus.

Figure 2. STAT3 activation. The transcription factor STAT3 is present in a latent inactive non-phosphorylated form in the cytoplasm. Activated cytokine receptors activate the kinases JAK, which phosphorylate tyrosines located in the cytoplasmic portion of the cytokine receptors creating STAT-binding motifs. Once bound to these motifs, STAT3 becomes in turn phosphorylated by the JAKs. The phospho STAT3 dimers (colored in pink) enter the nuclei and bind STAT3 target genes; note that the DNA-bound dimer is drawn differently to indicate the STAT3 conformational change suggested by molecular dynamics simulations.Citation22 STAT3 can also be activated by tyrosine kinases of the Src family (SFK), the SFKs can themselves be activated by G protein-coupled receptors (GPCR) or growth factor receptors, the growth factor receptors (including EGF-receptors and VEGF-receptors) can also phosphorylate STAT3. The DBD is created by the phosphotyrosine 705/SH2-mediated dimer. There are also monomers, and dimers with non-phosphorylated STAT3, which can shuttle between the cytoplasm and the nucleus.

Figure 3. STAT3 nuclear entry. The tyrosine phosphorylated STAT3 dimer interacts with the importins through its two NLSs [one within the coiled-coil (mL) domain: arginines 214 and 215; one within the DBD: arginines 414 and 417]. This complex is carried through the nuclear pore complex (NPC) by the Ran GDP which is formed in the cytoplasm by hydrolysis of Ran-bound GTP by GTPase activating protein (GAP). The NPC comprises nucleoporins (Nup). The importins release the dimer once it enters the nucleus. The high level of Ran-GTP in the nucleus is the result of a high GTP-exchange factor (GEF) activity in this compartment. The STAT3 dimer interacts with the STAT3 DNA consensus motif and then is released; once released from DNA the dimer is dephosphorylated by a nuclear phosphatase (PtPase). The dephosphorylated STAT3 is exported to the cytoplasm in combination with exportins and Ran-GTP.

Figure 3. STAT3 nuclear entry. The tyrosine phosphorylated STAT3 dimer interacts with the importins through its two NLSs [one within the coiled-coil (mL) domain: arginines 214 and 215; one within the DBD: arginines 414 and 417]. This complex is carried through the nuclear pore complex (NPC) by the Ran GDP which is formed in the cytoplasm by hydrolysis of Ran-bound GTP by GTPase activating protein (GAP). The NPC comprises nucleoporins (Nup). The importins release the dimer once it enters the nucleus. The high level of Ran-GTP in the nucleus is the result of a high GTP-exchange factor (GEF) activity in this compartment. The STAT3 dimer interacts with the STAT3 DNA consensus motif and then is released; once released from DNA the dimer is dephosphorylated by a nuclear phosphatase (PtPase). The dephosphorylated STAT3 is exported to the cytoplasm in combination with exportins and Ran-GTP.

Constitutive activation of STAT3 in tumors can result from upstream activated signaling components, including increased cytokines (IL-6 and IL-10) production, activated receptor (cytokine receptors, VEGFR and EGFR) and non-receptor tyrosine kinases (including JAKs, Src and Abl). Recently, mutated hyperactive forms of STAT3 have been detected in tumors, interestingly most of the described mutations are located within STAT3’s SH2Citation17,Citation18 (see ref. Citation19). Due to the involvement of tyrosine protein kinases in the STAT3 activating pathways, tyrosine kinase inhibitors indirectly inhibit STAT3 resulting in anti-tumor activity. But STAT3 can also be activated when suppressors of the STAT signaling pathway are inactivated such as the SOCS (suppressor of cytokine signaling), the PIAS (protein inhibitor of activated STATs) and protein tyrosine phosphatases;Citation20 furthermore, in cells in which proliferation results from the inactivation of negative regulators of signaling, such as the inhibitor of the PI3Kinase pathway PTEN, or the inhibitor of the proapoptotic TF p53, MDM2, the inhibition of upstream signaling pathways may have little effect. Thus, when upstream STAT3 activators are not identified or do not have any known inhibitor, or when STAT3 is itself activated it becomes a relevant target for anti-tumor treatments. This reasoning, reinforced by STAT3’s previously noted “signaling bottleneck” situation,Citation21 has been used by many authors searching for STAT3-specific direct inhibitors.

The Peculiar STAT3-STAT1 Connection

The high similarity between STAT3 and STAT1 is intriguing. Both TFs share a 50% amino-acid sequence homologyCitation22 and share similar activating stimuli (types I and II IFNs, cytokines and growth factors); yet, their functions differ: STAT1 is mostly involved in immunity, host defense against pathogens and cell death (see refs. Citation23 and Citation24), with the notable exception that in certain contexts STAT1 exerts proliferative potential,Citation25 while STAT3 is mostly involved in cell growth and proliferation (see ref. Citation3). Their gene targets are mostly distinct: STAT3 stimulates the transcription of cell growth-associated genes, including cyclin-D1, survivin, Vegf, C-myc, Bcl-xL, Mcl-1, vascular endothelial growth factor, IL-10, transforming growth factor β and Bcl2Citation3,Citation26 and STAT1 stimulates the transcription of pro-inflammatory and anti-proliferative genes, including caspases, iNOS, Mdm2, p21waf/cip1 and p27kip1Citation27,Citation28; but there is also an overlap of repertoires.Citation23 In reality, STAT3 and STAT1 recognize very similar DNA consensus sequences, based on a TTCNNN(T,G)AA motifCitation29 (see ). In this motif, a 3N spacing (N representing any base) is most frequently present for STAT1 and STAT3, as shown by electrophoretic migration shift assay (EMSA).Citation30 Natural binding sites share this consensus with minor variations such as a preference for T at position −5 for STAT1, but there is greater diversity outside this consensusCitation31 (see ), suggesting that target recognition in vivo requires additional co-factors. Thus, when attempting to inhibit STAT3 with the aim to kill tumor cell or block their growth, one must use substances that have no effect on STAT1, as has been pointed outCitation32,Citation33 and shown in tumor cell lines with siRNA-suppressed STAT1.Citation34,Citation35 Indeed, STAT1 is required for the antiproliferative effects of interferons α and γ,Citation36 apoptosis is defective in STAT1-null cellsCitation37 and STAT1 is the major effector of IFN-γ, a cytokine with antitumor and cancer immunosurveillance functions.Citation38,Citation39

Table 1. Comparison and alignment of the STAT3 and STAT1 specific DNA recognition sites and of the dODNs used to inhibit STAT3

STAT3 Direct Inhibitors

The recognition that activated STAT3 is widely present in tumors and that its inhibition is a valuable anti-cancer strategy led to a search for STAT3-targeting compounds. Among those the DNA-alkylating platinum complexes were found to induce the death of tumor cells with activated STAT3 and were thought to act directly on STAT3;Citation40 other compounds target the SH2 of STAT3, including G quartet oligodeoxynucleotides and small molecules, some of which are highly specific for STAT3. STAT3 inhibitors also comprise the decoy oligodeoxynucleotides which target the DBD.

Small molecules targeting the SH2 domain and inhibiting STAT3 dimerization

The dimerization of STAT3 through reciprocal phosphotyrosine 705/SH2 interaction can be impaired by a phosphopeptide with the sequence PpYLKTK. This phosphopeptide inhibits STAT3 activity in tumor cell lines, induces cell deathCitation41 and has a high affinity and specificity for STAT3: in particular it had no effect on the SH2-containing tyrosine kinase p56lck, and little effect on STAT1 as determined by EMSA. Despite their efficacy and specificity, the inefficient cell penetration of phosphopeptides led to a search for smaller equivalents using computational docking studies exploring the phosphotyrosine 705/SH2 interaction area (see ). These studies yielded several small molecules with high affinity and high specificity for STAT3, including STA-21,Citation42 Stattic,Citation43 S31-201Citation44 and recently BP-1-102, a compound with improved bioavailability and anti-cancer properties.Citation45 The mechanism of action of these compounds is thought to be based on their interaction with STAT3 SH2 (), an area where the other monomer’s phosphotyrosine 705 docks, thereby impairing the formation of the active dimer, as shown inCitation44 (see , borrowed from ref. Citation44; the phosphotyrosine peptide is represented together with the inhibitor S31-201, both form H-bonds with the same residues, including lysine 591, serine 611, serine 613 and arginine 609). Interestingly, STAT1-specific inhibitors have also been obtained using this peptidomimetic strategy.Citation46 STAT3 SH2-targeting compounds are efficacious STAT3 inhibitors, they inhibit STAT3 DNA binding, reduce STAT3-dependent cell proliferation and expression of biological targets. Yet, experimental demonstration of their interaction with STAT3 is missing, the actual data are based on computational studies, not on actual interaction measurements, as noted earlier.Citation47 This implies that a compound claimed to interact with SH2 might actually interfere with the binding of STAT3 to the receptor’s phosphotyrosine site leading to biological effects similar to those of JAK-inhibitors. The compound might also interact with the DBD with just the same effect in the cell (reduced DNA binding, cell death). Besides, compounds usually undergo modifications in a biological environment (cells or body fluids), these modifications can occur before the compounds reach their target. For instance, Stattic’s inhibitory activity of STAT3 increases with time and is temperature- and dithiothreitol-sensitive, this suggests that it interacts with a cysteine, such as cysteine 687 which is next to the phosphopeptide motif of STAT3 and faces the phosphopeptide-binding area of SH2.Citation47 While this point does not invalidate Stattic’s specificity for STAT3, it suggests possible limitations for its in vivo utilization. Despite considerable progress and clear anti-cancer efficacy, this family of compounds has not reached the clinic.

Figure 4. A detailed view of STAT3 SH2 and DBD regions. (A) STAT3 surface obtained as in . The areas in squares are the SH2 region where small molecules interact (B and C) and the SH2/DBD region where G quartets interact (D and E). (B) Close up view of the region of the SH2 of STAT3 that interacts with the small molecule inhibitors: the key aminoacids involved in interaction are labeled; the inhibitors interact particularly in the groove located between arginine 595 and lysine 591. (C) The same area as in (B) is shown but STAT3 and STAT1 are superimposed; this shows the overall great similarity between these two regions, yet some differences are present, accounting for the capacity of the inhibitors to discriminate between STAT3 and STAT1 (arrows); STAT1 crystal coordinatesCitation66 used in (C) were from PDB file 1BF5. (D) Modeled interaction of inhibitor S31-201 with STAT3 SH2 domain, note the important role of lysine 591, serine 611 and arginine 609 in the interaction. (E) Same as (D), with added phosphotyrosine-peptide, showing its overlap with the STAT3 SH2/small molecule inhibitors binding area. (F) Detailed view of the region of STAT3 interacting with G quartets, this region includes the phosphotyrosine 705-interacting region of SH2 and a neighboring region including part of the DBD, including glutamic acid 638, glutamine 644, aspartic acid 647, glutamine 643 and asparagine 646. (G) Same region as in F is shown with the superimposition of STAT1, the major differences are indicated by arrows. [Panels (D and E) are reprinted from ref. Citation44 with permission; © 2007 National Academy of Sciences USA.]

Figure 4. A detailed view of STAT3 SH2 and DBD regions. (A) STAT3 surface obtained as in Figure 1. The areas in squares are the SH2 region where small molecules interact (B and C) and the SH2/DBD region where G quartets interact (D and E). (B) Close up view of the region of the SH2 of STAT3 that interacts with the small molecule inhibitors: the key aminoacids involved in interaction are labeled; the inhibitors interact particularly in the groove located between arginine 595 and lysine 591. (C) The same area as in (B) is shown but STAT3 and STAT1 are superimposed; this shows the overall great similarity between these two regions, yet some differences are present, accounting for the capacity of the inhibitors to discriminate between STAT3 and STAT1 (arrows); STAT1 crystal coordinatesCitation66 used in (C) were from PDB file 1BF5. (D) Modeled interaction of inhibitor S31-201 with STAT3 SH2 domain, note the important role of lysine 591, serine 611 and arginine 609 in the interaction. (E) Same as (D), with added phosphotyrosine-peptide, showing its overlap with the STAT3 SH2/small molecule inhibitors binding area. (F) Detailed view of the region of STAT3 interacting with G quartets, this region includes the phosphotyrosine 705-interacting region of SH2 and a neighboring region including part of the DBD, including glutamic acid 638, glutamine 644, aspartic acid 647, glutamine 643 and asparagine 646. (G) Same region as in F is shown with the superimposition of STAT1, the major differences are indicated by arrows. [Panels (D and E) are reprinted from ref. Citation44 with permission; © 2007 National Academy of Sciences USA.]

G quartet oligodeoxynucleotides

G quartets are G-rich oligodeoxynucleotides that form potassium-dependent four-stranded intramolecular G-quartet structures.Citation48 They inhibit STAT3 at micromolar concentrations and induce the death of several tumor cell lines, including head and neck cancer lines, they also arrest the development of breast tumor, prostate tumor or non-small cell lung cancer xenografts in nude mice.Citation48,Citation49 These reagents are interesting in that their specificity for STAT3, demonstrated by EMSA showing high affinity binding to STAT3 and much lower affinity for STAT1,Citation50 is somewhat unexpected. A computational study of the interaction of the G quartet with STAT3 and STAT1 showed the following SH2 domain amino-acids to be involved in the binding: glutamic acid 638, glutamine 644, asparagine 647, glutamine 643 and asparagine 646Citation51 (). Using a 3D analysis program (Accelrys) we compared STAT3 and STAT1 surfaces in the area where the G quartet interacts and found that the surfaces differ significantly (). However, the use of G quartets is problematic due to their large size and potassium dependence, which limit cellular delivery.Citation52,Citation53

Decoy oligodeoxynucleotides targeting the DNA binding domain

Decoy oligodeoxynucleotides (dODNs) are short stretches of double stranded DNA containing a TF’s consensus binding sequence. Once in the cells, dODNs inhibit the corresponding TF as shown for NFκB in animal models (cardiovascular diseaseCitation54 and ischemia-reperfusion injuryCitation55) and in cancer cells.Citation56 This property has been used for STAT3: in cells in which STAT3 is constitutively activated, STAT3 dODNs () induce cell death.Citation35,Citation57,Citation58 While some studies suggested that dODNs must enter nuclei to exert their inhibitory action,Citation59,Citation60 our studies of the subcellular location of STAT3 in dODN-treated cells have shown that the dODNs prevent nuclear translocation of STAT3.Citation35,Citation58 The dODNs are thought to interact with STAT3′s DBD within the cytoplasm and to prevent interaction with importinsCitation35 which carry the STAT3 dimer within the nucleus.Citation7 Inhibition of importin binding by the dODN is thought to result from the masking of the DBD-located NLS (arginines 414 and 417) (see and ) indirectly confirming its involvement in nuclear entry.Citation9 In fact, competition between DNA binding and importin binding was observed in vitro with STAT1Citation61 and within cells with STAT3.Citation35 Thus, despite the uncertainty regarding the two STAT3 NLSs’ relative requirement for nuclear transfer, it seems that the DBD-located NLS plays an important role for nuclear translocation since its masking is sufficient to impair it, confirming that it may be a good target for inhibition, as suggested by others.Citation7,Citation53,Citation62

Figure 5. Detailed view of the area of STAT3 interacting with DNA and importins. (A) Detail of the area of STAT3 interacting with the DNA consensus sequence. Arginines 414 and 417 play a key role in the interaction of STAT3 with importins and nuclear translocation, arginine 423 is involved in the interaction of DNA with STAT3.Citation65 (B) The same area as in (A) is shown, the corresponding area of STAT1 has been superimposed, showing the high level of homology between STAT3 and STAT1, except for glutamic acid 421 (in italic) of STAT1 whose interaction with DNA is very different from that of arginine 423 of STAT3 (see ref. Citation65), and other differences indicated by arrows.

Figure 5. Detailed view of the area of STAT3 interacting with DNA and importins. (A) Detail of the area of STAT3 interacting with the DNA consensus sequence. Arginines 414 and 417 play a key role in the interaction of STAT3 with importins and nuclear translocation, arginine 423 is involved in the interaction of DNA with STAT3.Citation65 (B) The same area as in (A) is shown, the corresponding area of STAT1 has been superimposed, showing the high level of homology between STAT3 and STAT1, except for glutamic acid 421 (in italic) of STAT1 whose interaction with DNA is very different from that of arginine 423 of STAT3 (see ref. Citation65), and other differences indicated by arrows.

As discussed above, the opposed cellular functions of STAT1 and STAT3, in spite of their similarity, is puzzling. STAT1 and STAT3 can form heterodimers, whose function is not elucidated to this day, they recognize very similar DNA motifs (table), and they have targets in common and regulate one another. Indeed, in STAT3-deficient cells, STAT3-activators such as IL-6 trigger an IFN-γ-like response through STAT1 activationCitation63,Citation64; and in STAT1-deficient cells, IFN-γ and IFN-α trigger STAT3-dependent proliferative responses.Citation23 Thus the STAT1/STAT3 cross-regulation suggests that STAT3 inhibitors may work best in STAT1-expressing cells.Citation28,Citation35 It is therefore essential to inhibit STAT3 without inhibiting STAT1 to keep cell death processes operational. In this regard, it should be noted that STAT3-dODNs inhibit STAT3Citation57,Citation58 but also inhibit activated STAT1, thereby abolishing IFN-γ-induced cell deathCitation35,Citation58 and reducing the dODNs anti-STAT3 efficacy. Computer analysis of the dODN’s interaction with STAT1 and STAT3 DBDs showed that within the highly similar DNA sequences, there were subtle differences including a T at positions −7 and −5, a dC at position 0, a dA at position +5 (see , dODN #22),Citation65 which had been previously noted by computer analysisCitation66 and DNA-binding studies.Citation30,Citation31,Citation67,Citation68 This allowed designing a dODN that could inhibit STAT3 without inhibiting STAT1, demonstrating that at this level specific interaction can be achieved.Citation65 Therapeutic use of the dODNs not only requires specific target recognition, but also stability in biological fluids. Modifications, including phosphothioate end modification and hairpin structuresCitation35,Citation58,Citation69,Citation70 considerably increased intracellular stability and efficacy. A recent improvement consisting of a cyclic STAT3 dODNs comprising a hairpin at both ends was designed and found to reduce xenograft tumors following intravenous administration.Citation71 Furthermore, a STAT3 dODN has been found to have few side effects when administered to primates,Citation72 suggesting interesting therapeutic perspectives.

Another possibility is to design smaller molecules mimicking the dODN’s interaction with STAT3, similarly to what was achieved with the SH2 domain. The STAT3 area that interacts with the DNA target and the dODN () is of particular interest because it also interacts with importins. This area comprises the DBD-located NLS including arginines 414 and 417 that are located closely to arginine 423, involved in interaction with the dODN; these three arginines surround an opening in the surface in which small molecules could interact (). Furthermore, superimposition of STAT3 and STAT1 showed that in spite of their striking overall similarity, these areas comprise in the same location glutamic acid 421 in STAT1 and arginine 423 in STAT3Citation65 (), a difference that might be exploited for the design of a STAT3 small molecule inhibitor with STAT3/STAT1 discriminating capacity; studies are underway in the laboratory to try and design such a reagent.

Conclusion

Because STAT3 is constitutively activated in nearly 70% of tumors, it provides a valuable target for anti-cancer therapy. The recent findings that tumors harbor activating mutations of STAT3 underlines the need for additional STAT3 inhibitors, and inhibitors that target other regions of STAT3 than its SH2, especially as most identified mutations are within this region. In addition, studies on cancer cell lines indicate that even when STAT3 direct targeting is insufficient to induce cell death; it can diminish the resistance to other anti-cancer compounds such as doxorubicin or EGFR inhibitors. Finally, STAT3 direct inhibitors are also probably less toxic than many others because inhibition of STAT3 in mature cells has minimal effects.

Abbreviations:
IFNs=

interferons

GAS=

gamma-interferon-activated sequence

EMSA=

electrophoretic migration shift assay

pY=

phosphotyrosine

Acknowledgments

This work was supported in part by a grant (BQR) from University Paris 13.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Schindler C, Levy DE, Decker T. JAK-STAT signaling: from interferons to cytokines. J Biol Chem 2007; 282:20059 - 63; http://dx.doi.org/10.1074/jbc.R700016200; PMID: 17502367
  • Frank DA. STAT signaling in the pathogenesis and treatment of cancer. Mol Med 1999; 5:432 - 56; PMID: 10449805
  • Frank DA. STAT3 as a central mediator of neoplastic cellular transformation. Cancer Lett 2007; 251:199 - 210; http://dx.doi.org/10.1016/j.canlet.2006.10.017; PMID: 17129668
  • Bar-Natan M, Nelson EA, Xiang M, Frank DA. STAT signaling in the pathogenesis and treatment of myeloid malignancies. JAK-STAT 2012; 1:55 - 64; http://dx.doi.org/10.4161/jkst.20006
  • Yue P, Turkson J. Targeting STAT3 in cancer: how successful are we?. Expert Opin Investig Drugs 2009; 18:45 - 56; http://dx.doi.org/10.1517/13543780802565791; PMID: 19053881
  • Mankan AK, Greten FR. Inhibiting signal transducer and activator of transcription 3: rationality and rationale design of inhibitors. Expert Opin Investig Drugs 2011; 20:1263 - 75; http://dx.doi.org/10.1517/13543784.2011.601739; PMID: 21751940
  • Reich NC, Liu L. Tracking STAT nuclear traffic. Nat Rev Immunol 2006; 6:602 - 12; http://dx.doi.org/10.1038/nri1885; PMID: 16868551
  • Ma J, Zhang T, Novotny-Diermayr V, Tan AL, Cao X. A novel sequence in the coiled-coil domain of Stat3 essential for its nuclear translocation. J Biol Chem 2003; 278:29252 - 60; http://dx.doi.org/10.1074/jbc.M304196200; PMID: 12746441
  • Ma J, Cao X. Regulation of Stat3 nuclear import by importin alpha5 and importin alpha7 via two different functional sequence elements. Cell Signal 2006; 18:1117 - 26; http://dx.doi.org/10.1016/j.cellsig.2005.06.016; PMID: 16298512
  • Liu L, McBride KM, Reich NC. STAT3 nuclear import is independent of tyrosine phosphorylation and mediated by importin-alpha3. Proc Natl Acad Sci U S A 2005; 102:8150 - 5; http://dx.doi.org/10.1073/pnas.0501643102; PMID: 15919823
  • Yang J, Liao X, Agarwal MK, Barnes L, Auron PE, Stark GR. Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes Dev 2007; 21:1396 - 408; http://dx.doi.org/10.1101/gad.1553707; PMID: 17510282
  • Pranada AL, Metz S, Herrmann A, Heinrich PC, Müller-Newen G. Real time analysis of STAT3 nucleocytoplasmic shuttling. J Biol Chem 2004; 279:15114 - 23; http://dx.doi.org/10.1074/jbc.M312530200; PMID: 14701810
  • Herrmann A, Vogt M, Mönnigmann M, Clahsen T, Sommer U, Haan S, et al. Nucleocytoplasmic shuttling of persistently activated STAT3. J Cell Sci 2007; 120:3249 - 61; http://dx.doi.org/10.1242/jcs.03482; PMID: 17726064
  • Marg A, Shan Y, Meyer T, Meissner T, Brandenburg M, Vinkemeier U. Nucleocytoplasmic shuttling by nucleoporins Nup153 and Nup214 and CRM1-dependent nuclear export control the subcellular distribution of latent Stat1. J Cell Biol 2004; 165:823 - 33; http://dx.doi.org/10.1083/jcb.200403057; PMID: 15210729
  • Wenta N, Strauss H, Meyer S, Vinkemeier U. Tyrosine phosphorylation regulates the partitioning of STAT1 between different dimer conformations. Proc Natl Acad Sci U S A 2008; 105:9238 - 43; http://dx.doi.org/10.1073/pnas.0802130105; PMID: 18591661
  • Speil J, Baumgart E, Siebrasse JP, Veith R, Vinkemeier U, Kubitscheck U. Activated STAT1 transcription factors conduct distinct saltatory movements in the cell nucleus. Biophys J 2011; 101:2592 - 600; http://dx.doi.org/10.1016/j.bpj.2011.10.006; PMID: 22261046
  • Pilati C, Amessou M, Bihl MP, Balabaud C, Nhieu JT, Paradis V, et al. Somatic mutations activating STAT3 in human inflammatory hepatocellular adenomas. J Exp Med 2011; 208:1359 - 66; http://dx.doi.org/10.1084/jem.20110283; PMID: 21690253
  • Koskela HL, Eldfors S, Ellonen P, van Adrichem AJ, Kuusanmäki H, Andersson EI, et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N Engl J Med 2012; 366:1905 - 13; http://dx.doi.org/10.1056/NEJMoa1114885; PMID: 22591296
  • Groner B. Determinants of the extent and duration of STAT3 signaling. JAK-STAT 2012; 1:211 - 5; http://dx.doi.org/10.4161/jkst.21469
  • Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol 2007; 7:41 - 51; http://dx.doi.org/10.1038/nri1995; PMID: 17186030
  • Darnell JE Jr.. Transcription factors as targets for cancer therapy. Nat Rev Cancer 2002; 2:740 - 9; http://dx.doi.org/10.1038/nrc906; PMID: 12360277
  • Lin J, Buettner R, Yuan YC, Yip R, Horne D, Jove R, et al. Molecular dynamics simulations of the conformational changes in signal transducers and activators of transcription, Stat1 and Stat3. J Mol Graph Model 2009; 28:347 - 56; http://dx.doi.org/10.1016/j.jmgm.2009.08.013; PMID: 19781967
  • Regis G, Pensa S, Boselli D, Novelli F, Poli V. Ups and downs: the STAT1:STAT3 seesaw of Interferon and gp130 receptor signalling. Semin Cell Dev Biol 2008; 19:351 - 9; http://dx.doi.org/10.1016/j.semcdb.2008.06.004; PMID: 18620071
  • Najjar I, Fagard R. STAT1 and pathogens, not a friendly relationship. Biochimie 2010; 92:425 - 44; http://dx.doi.org/10.1016/j.biochi.2010.02.009; PMID: 20159032
  • Kovacic B, Stoiber D, Moriggl R, Weisz E, Ott RG, Kreibich R, et al. STAT1 acts as a tumor promoter for leukemia development. Cancer Cell 2006; 10:77 - 87; http://dx.doi.org/10.1016/j.ccr.2006.05.025; PMID: 16843267
  • Jarnicki A, Putoczki T, Ernst M. Stat3: linking inflammation to epithelial cancer - more than a “gut” feeling?. Cell Div 2010; 5:14; http://dx.doi.org/10.1186/1747-1028-5-14; PMID: 20478049
  • Chin YE, Kitagawa M, Kuida K, Flavell RA, Fu XY. Activation of the STAT signaling pathway can cause expression of caspase 1 and apoptosis. Mol Cell Biol 1997; 17:5328 - 37; PMID: 9271410
  • Avalle L, Pensa S, Regis G, Novelli F, Poli V. STAT1 and STAT3 in tumorigenesis a matter of balance. JAK-STAT 2012; 1:65 - 72; http://dx.doi.org/10.4161/jkst.20045
  • Darnell JE Jr., Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994; 264:1415 - 21; http://dx.doi.org/10.1126/science.8197455; PMID: 8197455
  • Seidel HM, Milocco LH, Lamb P, Darnell JE Jr., Stein RB, Rosen J. Spacing of palindromic half sites as a determinant of selective STAT (signal transducers and activators of transcription) DNA binding and transcriptional activity. Proc Natl Acad Sci U S A 1995; 92:3041 - 5; http://dx.doi.org/10.1073/pnas.92.7.3041; PMID: 7708771
  • Ehret GB, Reichenbach P, Schindler U, Horvath CM, Fritz S, Nabholz M, et al. DNA binding specificity of different STAT proteins. Comparison of in vitro specificity with natural target sites. J Biol Chem 2001; 276:6675 - 88; http://dx.doi.org/10.1074/jbc.M001748200; PMID: 11053426
  • Schindler C. STATs as activators of apoptosis. Trends Cell Biol 1998; 8:97 - 8; http://dx.doi.org/10.1016/S0962-8924(98)01233-1; PMID: 9695817
  • Lee CK, Smith E, Gimeno R, Gertner R, Levy DE. STAT1 affects lymphocyte survival and proliferation partially independent of its role downstream of IFN-gamma. J Immunol 2000; 164:1286 - 92; PMID: 10640742
  • Shim SH, Sung MW, Park SW, Heo DS. Absence of STAT1 disturbs the anticancer effect induced by STAT3 inhibition in head and neck carcinoma cell lines. Int J Mol Med 2009; 23:805 - 10; PMID: 19424608
  • Souissi I, Najjar I, Ah-Koon L, Schischmanoff PO, Lesage D, Le Coquil S, et al. A STAT3-decoy oligonucleotide induces cell death in a human colorectal carcinoma cell line by blocking nuclear transfer of STAT3 and STAT3-bound NF-κB. BMC Cell Biol 2011; 12:14; http://dx.doi.org/10.1186/1471-2121-12-14; PMID: 21486470
  • Bromberg JF, Horvath CM, Wen Z, Schreiber RD, Darnell JE Jr.. Transcriptionally active Stat1 is required for the antiproliferative effects of both interferon alpha and interferon gamma. Proc Natl Acad Sci U S A 1996; 93:7673 - 8; http://dx.doi.org/10.1073/pnas.93.15.7673; PMID: 8755534
  • Kumar A, Commane M, Flickinger TW, Horvath CM, Stark GR. Defective TNF-alpha-induced apoptosis in STAT1-null cells due to low constitutive levels of caspases. Science 1997; 278:1630 - 2; http://dx.doi.org/10.1126/science.278.5343.1630; PMID: 9374464
  • Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001; 410:1107 - 11; http://dx.doi.org/10.1038/35074122; PMID: 11323675
  • Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol 2006; 6:836 - 48; http://dx.doi.org/10.1038/nri1961; PMID: 17063185
  • Turkson J, Zhang S, Palmer J, Kay H, Stanko J, Mora LB, et al. Inhibition of constitutive signal transducer and activator of transcription 3 activation by novel platinum complexes with potent antitumor activity. Mol Cancer Ther 2004; 3:1533 - 42; PMID: 15634646
  • Turkson J, Ryan D, Kim JS, Zhang Y, Chen Z, Haura E, et al. Phosphotyrosyl peptides block Stat3-mediated DNA binding activity, gene regulation, and cell transformation. J Biol Chem 2001; 276:45443 - 55; http://dx.doi.org/10.1074/jbc.M107527200; PMID: 11579100
  • Song H, Wang R, Wang S, Lin J. A low-molecular-weight compound discovered through virtual database screening inhibits Stat3 function in breast cancer cells. Proc Natl Acad Sci U S A 2005; 102:4700 - 5; http://dx.doi.org/10.1073/pnas.0409894102; PMID: 15781862
  • Schust J, Sperl B, Hollis A, Mayer TU, Berg T. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization. Chem Biol 2006; 13:1235 - 42; http://dx.doi.org/10.1016/j.chembiol.2006.09.018; PMID: 17114005
  • Siddiquee K, Zhang S, Guida WC, Blaskovich MA, Greedy B, Lawrence HR, et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc Natl Acad Sci U S A 2007; 104:7391 - 6; http://dx.doi.org/10.1073/pnas.0609757104; PMID: 17463090
  • Zhang X, Yue P, Page BD, Li T, Zhao W, Namanja AT, et al. Orally bioavailable small-molecule inhibitor of transcription factor Stat3 regresses human breast and lung cancer xenografts. Proc Natl Acad Sci U S A 2012; 109:9623 - 8; http://dx.doi.org/10.1073/pnas.1121606109; PMID: 22623533
  • Gunning PT, Katt WP, Glenn M, Siddiquee K, Kim JS, Jove R, et al. Isoform selective inhibition of STAT1 or STAT3 homo-dimerization via peptidomimetic probes: structural recognition of STAT SH2 domains. Bioorg Med Chem Lett 2007; 17:1875 - 8; http://dx.doi.org/10.1016/j.bmcl.2007.01.077; PMID: 17336521
  • McMurray JS. A new small-molecule Stat3 inhibitor. Chem Biol 2006; 13:1123 - 4; http://dx.doi.org/10.1016/j.chembiol.2006.11.001; PMID: 17113993
  • Jing N, Li Y, Xiong W, Sha W, Jing L, Tweardy DJ. G-quartet oligonucleotides: a new class of signal transducer and activator of transcription 3 inhibitors that suppresses growth of prostate and breast tumors through induction of apoptosis. Cancer Res 2004; 64:6603 - 9; http://dx.doi.org/10.1158/0008-5472.CAN-03-4041; PMID: 15374974
  • Weerasinghe P, Garcia GE, Zhu Q, Yuan P, Feng L, Mao L, et al. Inhibition of Stat3 activation and tumor growth suppression of non-small cell lung cancer by G-quartet oligonucleotides. Int J Oncol 2007; 31:129 - 36; PMID: 17549413
  • Jing N, Zhu Q, Yuan P, Li Y, Mao L, Tweardy DJ. Targeting signal transducer and activator of transcription 3 with G-quartet oligonucleotides: a potential novel therapy for head and neck cancer. Mol Cancer Ther 2006; 5:279 - 86; http://dx.doi.org/10.1158/1535-7163.MCT-05-0302; PMID: 16505101
  • Zhu Q, Jing N. Computational study on mechanism of G-quartet oligonucleotide T40214 selectively targeting Stat3. J Comput Aided Mol Des 2007; 21:641 - 8; http://dx.doi.org/10.1007/s10822-007-9147-6; PMID: 18034310
  • Jing N, Tweardy DJ. Targeting Stat3 in cancer therapy. Anticancer Drugs 2005; 16:601 - 7; http://dx.doi.org/10.1097/00001813-200507000-00002; PMID: 15930886
  • Johnston PA, Grandis JR. STAT3 signaling: anticancer strategies and challenges. Mol Interv 2011; 11:18 - 26; http://dx.doi.org/10.1124/mi.11.1.4; PMID: 21441118
  • Morishita R, Nakagami H, Taniyama Y, Matsushita H, Yamamoto K, Tomita N, et al. Oligonucleotide-based gene therapy for cardiovascular disease. Clin Chem Lab Med 1998; 36:529 - 34; http://dx.doi.org/10.1515/CCLM.1998.090; PMID: 9806454
  • Aoki M, Morishita R, Higaki J, Moriguchi A, Hayashi S, Matsushita H, et al. Survival of grafts of genetically modified cardiac myocytes transfected with FITC-labeled oligodeoxynucleotides and the beta-galactosidase gene in the noninfarcted area, but not the myocardial infarcted area. Gene Ther 1997; 4:120 - 7; http://dx.doi.org/10.1038/sj.gt.3300361; PMID: 9081702
  • Lin MT, Chang CC, Chen ST, Chang HL, Su JL, Chau YP, et al. Cyr61 expression confers resistance to apoptosis in breast cancer MCF-7 cells by a mechanism of NF-kappaB-dependent XIAP up-regulation. J Biol Chem 2004; 279:24015 - 23; http://dx.doi.org/10.1074/jbc.M402305200; PMID: 15044484
  • Leong PL, Andrews GA, Johnson DE, Dyer KF, Xi S, Mai JC, et al. Targeted inhibition of Stat3 with a decoy oligonucleotide abrogates head and neck cancer cell growth. Proc Natl Acad Sci U S A 2003; 100:4138 - 43; http://dx.doi.org/10.1073/pnas.0534764100; PMID: 12640143
  • Tadlaoui Hbibi A, Laguillier C, Souissi I, Lesage D, Le Coquil S, Cao A, et al. Efficient killing of SW480 colon carcinoma cells by a signal transducer and activator of transcription (STAT) 3 hairpin decoy oligodeoxynucleotide--interference with interferon-gamma-STAT1-mediated killing. FEBS J 2009; 276:2505 - 15; http://dx.doi.org/10.1111/j.1742-4658.2009.06975.x; PMID: 19476491
  • Park YG, Nesterova M, Agrawal S, Cho-Chung YS. Dual blockade of cyclic AMP response element- (CRE) and AP-1-directed transcription by CRE-transcription factor decoy oligonucleotide. gene-specific inhibition of tumor growth. J Biol Chem 1999; 274:1573 - 80; http://dx.doi.org/10.1074/jbc.274.3.1573; PMID: 9880535
  • Bene A, Kurten RC, Chambers TC. Subcellular localization as a limiting factor for utilization of decoy oligonucleotides. Nucleic Acids Res 2004; 32:e142; http://dx.doi.org/10.1093/nar/gnh139; PMID: 15498923
  • Nardozzi J, Wenta N, Yasuhara N, Vinkemeier U, Cingolani G. Molecular basis for the recognition of phosphorylated STAT1 by importin alpha5. J Mol Biol 2010; 402:83 - 100; http://dx.doi.org/10.1016/j.jmb.2010.07.013; PMID: 20643137
  • Meyer T, Vinkemeier U. STAT nuclear translocation: potential for pharmacological intervention. Expert Opin Ther Targets 2007; 11:1355 - 65; http://dx.doi.org/10.1517/14728222.11.10.1355; PMID: 17907964
  • Costa-Pereira AP, Tininini S, Strobl B, Alonzi T, Schlaak JF, Is’harc H, et al. Mutational switch of an IL-6 response to an interferon-gamma-like response. Proc Natl Acad Sci U S A 2002; 99:8043 - 7; http://dx.doi.org/10.1073/pnas.122236099; PMID: 12060750
  • Schiavone D, Avalle L, Dewilde S, Poli V. The immediate early genes Fos and Egr1 become STAT1 transcriptional targets in the absence of STAT3. FEBS Lett 2011; 585:2455 - 60; http://dx.doi.org/10.1016/j.febslet.2011.06.020; PMID: 21723864
  • Souissi I, Ladam P, Cognet JA, Le Coquil S, Varin-Blank N, Baran-Marszak F, et al. A STAT3-inhibitory hairpin decoy oligodeoxynucleotide discriminates between STAT1 and STAT3 and induces death in a human colon carcinoma cell line. Mol Cancer 2012; 11:12; http://dx.doi.org/10.1186/1476-4598-11-12; PMID: 22423663
  • Chen X, Vinkemeier U, Zhao Y, Jeruzalmi D, Darnell JE Jr., Kuriyan J. Crystal structure of a tyrosine phosphorylated STAT-1 dimer bound to DNA. Cell 1998; 93:827 - 39; http://dx.doi.org/10.1016/S0092-8674(00)81443-9; PMID: 9630226
  • Horvath CM, Wen Z, Darnell JE Jr.. A STAT protein domain that determines DNA sequence recognition suggests a novel DNA-binding domain. Genes Dev 1995; 9:984 - 94; http://dx.doi.org/10.1101/gad.9.8.984; PMID: 7774815
  • Lamb P, Seidel HM, Haslam J, Milocco L, Kessler LV, Stein RB, et al. STAT protein complexes activated by interferon-gamma and gp130 signaling molecules differ in their sequence preferences and transcriptional induction properties. Nucleic Acids Res 1995; 23:3283 - 9; http://dx.doi.org/10.1093/nar/23.16.3283; PMID: 7667105
  • Dolinnaya N, Metelev V, Oretskaya T, Tabatadze D, Shabarova Z. Hairpin-shaped DNA duplexes with disulfide bonds in sugar-phosphate backbone as potential DNA reagents for crosslinking with proteins. FEBS Lett 1999; 444:285 - 90; http://dx.doi.org/10.1016/S0014-5793(99)00059-9; PMID: 10050776
  • Laguillier C, Hbibi AT, Baran-Marszak F, Metelev V, Cao A, Cymbalista F, et al. Cell death in NF-kappaB-dependent tumour cell lines as a result of NF-kappaB trapping by linker-modified hairpin decoy oligonucleotide. FEBS Lett 2007; 581:1143 - 50; http://dx.doi.org/10.1016/j.febslet.2007.02.024; PMID: 17328896
  • Sen M, Thomas SM, Kim S, Yeh JI, Ferris RL, Johnson JT, et al. First-in-human trial of a STAT3 decoy oligonucleotide in head and neck tumors: implications for cancer therapy. Cancer Discov 2012; 2:694 - 705; http://dx.doi.org/10.1158/2159-8290.CD-12-0191; PMID: 22719020
  • Sen M, Tosca PJ, Zwayer C, Ryan MJ, Johnson JD, Knostman KA, et al. Lack of toxicity of a STAT3 decoy oligonucleotide. Cancer Chemother Pharmacol 2009; 63:983 - 95; http://dx.doi.org/10.1007/s00280-008-0823-6; PMID: 18766340
  • Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, et al. UCSF Chimera--a visualization system for exploratory research and analysis. J Comput Chem 2004; 25:1605 - 12; http://dx.doi.org/10.1002/jcc.20084; PMID: 15264254
  • Becker S, Groner B, Müller CW. Three-dimensional structure of the Stat3beta homodimer bound to DNA. Nature 1998; 394:145 - 51; http://dx.doi.org/10.1038/28101; PMID: 9671298
  • Valente AJ, Xie JF, Abramova MA, Wenzel UO, Abboud HE, Graves DT. A complex element regulates IFN-gamma-stimulated monocyte chemoattractant protein-1 gene transcription. J Immunol 1998; 161:3719 - 28; PMID: 9759897
  • Wang YF, Yu-Lee LY. Multiple stat complexes interact at the interferon regulatory factor-1 interferon-gamma activation sequence in prolactin-stimulated Nb2 T cells. Mol Cell Endocrinol 1996; 121:19 - 28; http://dx.doi.org/10.1016/0303-7207(96)03840-3; PMID: 8865162
  • Rouyez MC, Lestingi M, Charon M, Fichelson S, Buzyn A, Dusanter-Fourt I. IFN regulatory factor-2 cooperates with STAT1 to regulate transporter associated with antigen processing-1 promoter activity. J Immunol 2005; 174:3948 - 58; PMID: 15778351
  • Pine R, Canova A, Schindler C. Tyrosine phosphorylated p91 binds to a single element in the ISGF2/IRF-1 promoter to mediate induction by IFN alpha and IFN gamma, and is likely to autoregulate the p91 gene. EMBO J 1994; 13:158 - 67; PMID: 8306959
  • Caldenhoven E, Coffer P, Yuan J, Van de Stolpe A, Horn F, Kruijer W, et al. Stimulation of the human intercellular adhesion molecule-1 promoter by interleukin-6 and interferon-gamma involves binding of distinct factors to a palindromic response element. J Biol Chem 1994; 269:21146 - 54; PMID: 7914891
  • Nguyen VT, Benveniste EN. Involvement of STAT-1 and ets family members in interferon-gamma induction of CD40 transcription in microglia/macrophages. J Biol Chem 2000; 275:23674 - 84; http://dx.doi.org/10.1074/jbc.M002482200; PMID: 10823830
  • Fujio Y, Kunisada K, Hirota H, Yamauchi-Takihara K, Kishimoto T. Signals through gp130 upregulate bcl-x gene expression via STAT1-binding cis-element in cardiac myocytes. J Clin Invest 1997; 99:2898 - 905; http://dx.doi.org/10.1172/JCI119484; PMID: 9185513
  • Dong Y, Rohn WM, Benveniste EN. IFN-gamma regulation of the type IV class II transactivator promoter in astrocytes. J Immunol 1999; 162:4731 - 9; PMID: 10202014
  • Gustafson KS, Ginder GD. Interferon-gamma induction of the human leukocyte antigen-E gene is mediated through binding of a complex containing STAT1alpha to a distinct interferon-gamma-responsive element. J Biol Chem 1996; 271:20035 - 46; http://dx.doi.org/10.1074/jbc.271.33.20035; PMID: 8702722
  • Stojanovic T, Wagner AH, Wang S, Kiss E, Rockstroh N, Bedke J, et al. STAT-1 decoy oligodeoxynucleotide inhibition of acute rejection in mouse heart transplants. Basic Res Cardiol 2009; 104:719 - 29; http://dx.doi.org/10.1007/s00395-009-0028-0; PMID: 19352584
  • Xu W, Comhair SA, Zheng S, Chu SC, Marks-Konczalik J, Moss J, et al. STAT-1 and c-Fos interaction in nitric oxide synthase-2 gene activation. Am J Physiol Lung Cell Mol Physiol 2003; 285:L137 - 48; PMID: 12788789
  • Marrero MB, Bencherif M. Convergence of alpha 7 nicotinic acetylcholine receptor-activated pathways for anti-apoptosis and anti-inflammation: central role for JAK2 activation of STAT3 and NF-kappaB. Brain Res 2009; 1256:1 - 7; http://dx.doi.org/10.1016/j.brainres.2008.11.053; PMID: 19063868
  • Hückel M, Schurigt U, Wagner AH, Stöckigt R, Petrow PK, Thoss K, et al. Attenuation of murine antigen-induced arthritis by treatment with a decoy oligodeoxynucleotide inhibiting signal transducer and activator of transcription-1 (STAT-1). Arthritis Res Ther 2006; 8:R17; http://dx.doi.org/10.1186/ar1869; PMID: 16507120
  • Magné S, Caron S, Charon M, Rouyez MC, Dusanter-Fourt I. STAT5 and Oct-1 form a stable complex that modulates cyclin D1 expression. Mol Cell Biol 2003; 23:8934 - 45; http://dx.doi.org/10.1128/MCB.23.24.8934-8945.2003; PMID: 14645506
  • Wagner BJ, Hayes TE, Hoban CJ, Cochran BH. The SIF binding element confers sis/PDGF inducibility onto the c-fos promoter. EMBO J 1990; 9:4477 - 84; PMID: 2176154
  • Bellido T, O’Brien CA, Roberson PK, Manolagas SC. Transcriptional activation of the p21(WAF1,CIP1,SDI1) gene by interleukin-6 type cytokines. A prerequisite for their pro-differentiating and anti-apoptotic effects on human osteoblastic cells. J Biol Chem 1998; 273:21137 - 44; http://dx.doi.org/10.1074/jbc.273.33.21137; PMID: 9694869
  • Auernhammer CJ, Bousquet C, Melmed S. Autoregulation of pituitary corticotroph SOCS-3 expression: characterization of the murine SOCS-3 promoter. Proc Natl Acad Sci U S A 1999; 96:6964 - 9; http://dx.doi.org/10.1073/pnas.96.12.6964; PMID: 10359822
  • Kiuchi N, Nakajima K, Ichiba M, Fukada T, Narimatsu M, Mizuno K, et al. STAT3 is required for the gp130-mediated full activation of the c-myc gene. J Exp Med 1999; 189:63 - 73; http://dx.doi.org/10.1084/jem.189.1.63; PMID: 9874564
  • Pallard C, Gouilleux F, Bénit L, Cocault L, Souyri M, Levy D, et al. Thrombopoietin activates a STAT5-like factor in hematopoietic cells. EMBO J 1995; 14:2847 - 56; PMID: 7796811
  • Campbell GS, Meyer DJ, Raz R, Levy DE, Schwartz J, Carter-Su C. Activation of acute phase response factor (APRF)/Stat3 transcription factor by growth hormone. J Biol Chem 1995; 270:3974 - 9; http://dx.doi.org/10.1074/jbc.270.8.3974; PMID: 7876144