Nuclear translocation of STAT3 and NF-κB are independent of each other but NF-κB supports expression and activation of STAT3 Antons Martincuks, Katarzyna Andryka, Andrea K¨uster, Hildegard SchmitzVan de Leur, Michal Komorowski, Gerhard M¨uller-Newen PII: DOI: Reference:
S0898-6568(17)30006-2 doi: 10.1016/j.cellsig.2017.01.006 CLS 8821
To appear in:
Cellular Signalling
Received date: Revised date: Accepted date:
13 May 2016 31 December 2016 4 January 2017
Please cite this article as: Antons Martincuks, Katarzyna Andryka, Andrea K¨ uster, Hildegard Schmitz-Van de Leur, Michal Komorowski, Gerhard M¨ uller-Newen, Nuclear translocation of STAT3 and NF-κB are independent of each other but NFκB supports expression and activation of STAT3, Cellular Signalling (2017), doi: 10.1016/j.cellsig.2017.01.006
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.
ACCEPTED MANUSCRIPT Nuclear translocation of STAT3 and NF-B are independent of each other but NF-B supports expression and activation of STAT3
SC
RI
PT
Antons Martincuks1, Katarzyna Andryka2, Andrea Küster1, Hildegard Schmitz-Van de Leur1, Michal Komorowski2, Gerhard Müller-Newen1*
1
NU
Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany 2
D
AC CE P
TE
* Corresponding author:
[email protected]
MA
Systems Biology of Biochemical Signalling, Laboratory of Modelling in Biology and Medicine, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawinskiego 5b, 02-106 Warszawa, Poland
1
ACCEPTED MANUSCRIPT Abstract NF-κB and STAT3 are essential transcription factors in immunity and act at the interface of the
PT
transition from chronic inflammation to cancer. Different functional crosstalks between NF-κB and STAT3 have been recently described arguing for a direct interaction of both proteins. During
RI
a systematic analysis of NF-κB/STAT3 crosstalk we observed that appearance of the subcellular distribution of NF-κB and STAT3 in immunofluorescence heavily depends on the fixation
SC
procedure. Therefore, we established an optimized fixation protocol for the reliable simultaneous analysis of the subcellular distributions of both transcription factors. Using this protocol we
NU
found that cytokine-induced nuclear accumulation of NF-κB or STAT3 did not alter the subcellular distribution of the other transcription factor. Both knockout and overexpression of
MA
STAT3 does not have any major effect on canonical TNFα-NF-κB signalling in MEF or HeLa cells. Similarly, knockout of p65 did not alter nuclear accumulation of STAT3 in response to IL6. However, p65 expression correlates with elevated total cellular levels of STAT3 and STAT1
D
and supports activation of these transcription factors. Our findings in MEF cells argue against a
Keywords
AC CE P
functional interactions.
TE
direct physical interaction of free cellular NF-κB and STAT3 but point to more intricate
STAT3, NF-κB, signal transduction, nuclear translocation, crosstalk
Abbreviations CFP, cyan fluorescent protein; IL, interleukin; JAK, Janus kinase; MEF, murine embryonic fibroblasts; NF, nuclear factor; NLS, nuclear localization signal; PBS, phosphate-buffered saline; R, receptor; s, soluble; STAT, signal transducer and activator of transcription; TNF, tumour necrosis factor; WT, wild-type; YFP, yellow fluorescent protein
2
ACCEPTED MANUSCRIPT 1 Introduction The signal transducer and activator of transcription 3 (STAT3) belongs to a family of
PT
transcription factors consisting of STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and
RI
STAT6 which are involved in the regulation of numerous cellular processes [1]. STAT3 is
SC
ubiquitously expressed and has the most pleiotropic functions among all STATs [2]. The family of IL-6-type cytokines signals through the cytokine receptor gp130 and these cytokines are
NU
among the most potent physiological activators of STAT3 [3]. Like other members of the STAT
MA
family, STAT3 signalling involves tyrosine phosphorylation by Janus kinases (JAKs) or other tyrosine kinases, dimerization and nuclear translocation. In the nucleus, STAT3 binds to DNA
D
and regulates target gene expression. Upon dephosphorylation [4], STAT3 dissociates from DNA
TE
and exits the nucleus via CRM1-mediated export into the cytoplasm, where it becomes available for reactivation [5-7]. Aberrant STAT3 signalling has been shown to be a key factor in
AC CE P
oncogenesis, chronic inflammation and autoimmune conditions, defining STAT3 as a promising therapeutic target [8, 9].
Nuclear factor kappa B (NF-κB) is a collective name for several dimeric transcription factors composed of members of the Rel family of DNA-binding proteins comprising p65 (RelA), RelB, c-Rel, NF-κB1 (p50 and its precursor p105) NF-κB2 (p52 and its precursor p100). A heterodimer of p50 and p65 subunits is the most abundant form and often simply referred to as NF-κB. In this form, NF-κB is a crucial regulator of many physiological and pathophysiological processes including control of innate and adaptive immune responses, inflammation and cancer progression [10]. Similar to STAT3, NF-κB relays signals from activated receptors in the plasma membrane
to the nucleus, where it regulates gene expression. However, in resting cells NF-κB is maintained in an inactive form by sequestration in the cytoplasm through interaction with inhibitors of kappa
3
ACCEPTED MANUSCRIPT B (IκB). Canonical NF-κB activation by TNFα involves phosphorylation and proteasomal elimination of IκBα uncovering a nuclear localization signal (NLS) of NF-κB. [11].
PT
Consequently, NF-κB accumulates in the nucleus and regulates the expression of target genes
RI
[12]. NF-κB-dependent re-synthesis of IκB proteins as part of a negative feedback loop and
SC
additional mechanisms then lead to NF-κB dimer dissociation and CRM1-dependent nuclear export [13, 14].
NU
In recent years, a functional crosstalk between STAT3- and NF-κB-mediated signal transduction pathways has been described as having a central role in cancer progression [15, 16] and acute
MA
phase response in the liver [17]. Numerous types of interactions between NF-κB and STAT3
D
have been observed, including an overlapping set of target genes, physical association,
TE
competition or cooperation at gene promoters and others [15, 18]. However, a coherent picture of how both factors can directly influence each other’s signalling has not emerged. On the one
AC CE P
hand, STAT3 was required for the RhoA-induced NF-κB gene transcription, while nuclear accumulation of p65 subunit was reported to be impaired in STAT3 deficient cells upon both Rho and TNFα stimulation [19, 20]. In contrast, an augmented nuclear p65 presence and increased expression of typical NF-κB target genes upon STAT3 depletion was observed in A549 lung cancer cells [21], glioblastoma cells [22] and primary mouse pneumocytes [23]. Little is known about the effect of p65 deletion or overexpression on STAT3 activity. To resolve these issues, we have investigated the effects of conditional knockouts or overexpression of STAT3 and NF-κB transcription factors on each other. For this purpose, we have chosen cultured mouse embryonic fibroblasts (MEF) as a well-established system for studying the impact of gene knockouts on basic cellular processes. Although total knockout of STAT3 led to early embryonic lethality in mice [24], conditional gene targeting has been
4
ACCEPTED MANUSCRIPT successfully used to study STAT3 functions in specific tissues and cells [25, 26] and STAT3 knockout MEF have been established [27]. Furthermore, p65 deficient MEF have also been
PT
reported to proliferate and thrive in culture [28]. In this report, we have focused on the
RI
involvement of NF-κB p65 in IL-6-induced STAT3 activation, nuclear accumulation and gene
SC
expression, as well as on the reciprocal role of STAT3 in canonical TNFα-induced NF-κB signalling using p65-deficient and STAT3-deficient MEF along with inducible expression of the
MA
NU
transcription factors in HeLa cells.
2 Materials and Methods
D
2.1 Cytokines, cytokine receptors, antibodies and chemicals.
TE
Recombinant human IL-6 and soluble IL-6 receptor alpha (sIL-6Rα) were prepared as described
AC CE P
previously [29, 30]. Recombinant human TNFα was obtained from Peprotech (#300-01A, USA). Anti-pY701-STAT1 (#7649), anti-pY705-STAT3 (#9131), anti-pS536-p65 (#3033), anti-pS32IκBα (#2859), anti-STAT3 (#9139), anti-p65 (#4764), anti-IκBα (#4814 Cell Signaling, USA), anti-lamin A/C (sc-6215), anti-STAT1 (sc-346) anti-GAPDH (sc-32233, Santa Cruz Biotechnology, USA) and anti-dsRed (#632496, Clontech Laboratories, Mountain View, USA) antibodies were used for immunodetection. Anti-rabbit, anti-goat and anti-mouse horseradish peroxidase conjugated antibodies (DAKO, Germany) were used as secondary antibodies. 2.2 Plasmid constructs The pMOWS expression vectors were kindly provided by Prof. Jürgen Scheller, Heinrich-Heine University, Germany, as described in [31]. The expression vector containing human p65 was kindly provided by Dr. Marcus W. Covert [32] and was used for cloning of pMOWS-p65–YFP
5
ACCEPTED MANUSCRIPT by PCR. S276A point mutant of p65 was generated with PCR based QuikChange® XL SiteDirected Mutagenesis Kit according to manufacturer's instructions (Agilent Technologies, USA)
RI
antisense 5'- GCTCCCGGTCGGCAGGCCGCCGCAGC-3'.
PT
using the following primers: sense 5'-GCTGCGGCGGCCTGCCGACCGGGAGC-3' and
SC
2.3 Cell culture and transfection
HeLa, wild-type murine embryonic fibroblasts (MEF), STAT3-deficient (STAT3-/-) (generous
NU
gift from Prof. Valeria Poli, University of Turin, Italy, described in [27]) and p65-deficient (p65-
MA
/-) (generous gift from Prof. Lienhard Schmitz, Justus-Liebig-University, Germany, described in [28]) MEF were cultivated in Dulbecco’s modified Eagle’s medium (DMEM)/GlutaMAX
D
(Gibco, Paisley, UK) supplemented with 10% FCS (Life Technologies GmbH, Germany) and
TE
1% penicillin/streptomycin (Sigma-Aldrich, USA). STAT3-deficient MEF stably transfected with (WT)STAT3-CFP-YFP have been described earlier [7] and were additionally supplemented
AC CE P
with 300 μg/ml hygromycin B (Invivogen, France). MEF p65 -/- cells were retrovirally transduced with p65-YFP or p65(S276A)-YFP fusion proteins using the pMOWS system and supplemented with 1 μg/ml puromycin (Invivogen, France). Inducible HeLa Flp-In T-REx cells were stably transfected for the inducible expression of p65, dsRed (monomer), p65-dsRed (monomer) and STAT3-eGFP proteins using the Flp-In system (Invitrogen, UK) and supplemented with 250 μg/ml hygromycin B and 15 μg/ml blasticidin (Invivogen, France) for selection and 100 μg/ml hygromycin B and 10 μg/ml blasticidin for cultivation. The expression of the encoded protein was induced with 10 ng/ml doxycycline (Sigma-Aldrich, USA) for 24 h. 2.4 Preparation of cell lysates, SDS-PAGE and immunoblotting MEF were cultured on six-well plates and stimulated as described in the figure legends. For total cell lysates cells were lysed on ice for 30 minutes in RIPA lysis buffer (50 mM Tris-HCl pH 7.4, 6
ACCEPTED MANUSCRIPT 150 mM NaCl, 1 mM EDTA, 0.5% Nonidet P-40, 1 mM NaF, 15% glycerol). For subcellular fractionation, cells were resuspended in hypotonic lysis buffer (10 mM Tris-HCl pH 7.5, 10 mM
PT
NaCl, 3 mM MgCl2) and centrifuged at 2,000 RPM for 10 min to collect the supernatant
RI
(cytosolic fraction). The resulting pellet was washed three times in nuclear isolation buffer (10 mM Tris-HCl pH 7.5, 10 mM NaCl, 3 mM MgCl2, 0.5% Nonidet P-40) to remove cytoplasmic
SC
waste, resuspended in nuclear lysis buffer (20 mM Tris-HCl pH 7.5, 280 mM NaCl, 10 mM NaF,
NU
2% Triton X-100) and centrifuged again at 13,000 RPM for 10 min to collect the supernatant representing the nuclear fraction. All lysis buffers were supplemented with phosphatase/protease
MA
inhibitors (1 mM sodium orthovanadate, 0.5 mM EDTA, 0.25 mM phenylmethylsulfonylfluoride (PMSF), 5 μg/ml aprotinin, 2.5 μg/ml leupeptin). Protein concentrations of the lysates were
TE
D
determined by the Bio-Rad protein assay (Bio-Rad, Germany). Protein lysates were separated by 10% SDS-PAGE and transferred to a PVDF membrane by semi-dry blotting with subsequent
AC CE P
immunodetection using the indicated antibodies. Bound secondary antibodies were detected by pCA-ECL chemiluminescence and signals analysed with Fujifilm MultiGauge v3.2 software. 2.5 Immunofluorescence
Cells were grown on 18 mm glass coverslips and stimulated as indicated. After washing with PBS++ (2.7 mM KCl pH 7.5, 137 mM NaCl, 8.1 mM Na2HPO4, 1.5 mM KH2PO4, 1 mM MgCl2, 0.1 mM CaCl2) and fixed either with 3.7% PFA or ice-cold 100% methanol at room temperature for 20 minutes for one-step fixation, or with 3.7% PFA at room temperature and subsequent icecold 100% methanol at –20°C for 10 minutes. Afterwards, the cells were permeabilized with PBS++ containing 0.1% Triton X-100 (PBST++) for 5 minutes, quenched with 50 mM NH4Cl in PBST++ and blocked with PBST++ containing 5% BSA (Serva, Germany). Cells were labeled with rabbit p65 antibody and mouse STAT3 antibody. The manufacturer Cell Signaling
7
ACCEPTED MANUSCRIPT recommends paraformaldehyde fixation for p65 antibody (#4764) and methanol fixation for STAT3 antibody (#9131) during sample preparation. Alexa Fluor® 555 anti-rabbit and Alexa
PT
Fluor® 488 anti-mouse antibodies (A-31572 and A-21202; Thermo Fischer Scientific, USA)
RI
were used as secondary antibodies. Nuclei were stained with DRAQ5™ (Biostatus, UK).
SC
2.6 Confocal microscopy
Confocal imaging was performed with a Zeiss LSM 710 confocal microscope (Zeiss, Germany)
NU
using a NA 40x/1.1 water immersion objective. Alexa Fluor® 555 fluorescence was excited
MA
using the 561 nm laser, emission was detected in the range of 562-650 nm. Alexa Fluor® 488 signal was excited using the argon 488 nm laser, emission was detected in the range of 493-550
D
nm. DRAQ5™ nuclear staining was detected using the 633 nm line of the Red Helium-Neon
TE
laser and a 661-759 nm detection range. Mean nuclear fluorescence of A555-stained p65 and
AC CE P
A488-stained STAT3 was calculated using Mathworks MATLAB®. 2.7 RNA preparation and quantitative RT-PCR MEF were stimulated as indicated and total RNA was extracted according to the manufacturer's instructions using RNeasy Mini Kit (Qiagen, Netherlands). Afterwards, 1 μg of total RNA was reverse transcribed using random hexamers (Sigma-Aldrich, USA) and Omniscript Kit (Qiagen, Netherlands). qPCR was performed with Sybr-Green reaction mix (#QT650-02; Bioline, USA) using Rotorgene (Qiagen, Netherlands). Expression was normalized to the housekeeper gene mGUSB (#QT00176715, Qiagen, Germany). Following primer sequences were used for amplification of cDNAs: 5`- CCG CGG GCA CCT TTC-3` forward and 5`- TTG ACG CTC AAC GTG AAG AAGT - 3` reverse for socs3, 5`- CTC CCC CTA CCA GCT TAC CT -3` forward and 5`- TAG GGC AGC TCA TCC TCT GT - 3` reverse for ikba.
8
ACCEPTED MANUSCRIPT 2.8 Statistical analysis The relative cDNA expression ratio was calculated by the ∆∆Ct method [33]. Quantification of
PT
immunofluorescence, imunoblotting and RT-PCR data was plotted using GraphPad Prism 6
RI
software and statistically evaluated by Student’s t-test. A P value of less than 0.05 was
NU
SC
considered as significant.
3 Results
MA
3.1 Canonical nuclear translocation of STAT3 and NF-κB are independent of each other. We previously showed IL-6-induced nuclear accumulation of endogenous STAT3 using
TE
D
methanol fixation and immunofluorescence staining of MEF [34]. However, paraformaldehyde fixation has been reported to be better suited for visualization of NF-κB p65 localization by
AC CE P
immunofluorescence [35]. The choice of the processing method is most important for appearance of the immunostaining and the interpretation of the results may vary depending on the protocol used [36]. In order to find optimal conditions to analyse both p65 and STAT3 subcellular localization in parallel, the two conditions of fixation were compared along with combined paraformaldehyde and methanol treatment (Fig. 1A). MEF were treated for 30 min with TNF or IL-6 to induce nuclear translocation of endogenous p65 or STAT3, respectively, or left untreated. TNFα-induced nuclear accumulation of p65 was clearly visible in paraformaldehyde fixed cells but hardly detectable upon methanol fixation (Suppl. Fig. 1A,B). However, paraformaldehyde fixation is not suited for the analysis of cytokine-induced nuclear accumulation of STAT3 because already in unstimulated cells the bulk of STAT3 appeared nuclear (Suppl. Fig. 1A). In order to resolve these problems, we applied a combined protocol of paraformaldehyde fixation
9
ACCEPTED MANUSCRIPT followed by methanol treatment and after immunofluorescence staining assessed the subcellular localization of both factors by confocal microscopy (Suppl. Fig. 1C). Under these conditions,
PT
STAT3 shows uniform distribution in resting state and nuclear accumulation upon IL-6, but not TNFα treatment, as expected. In turn, p65 is cytoplasmic in unstimulated cells and enters the
RI
nucleus after TNFα but not after IL-6 stimulation. These results correlate with subcellular
SC
fractionation analysis (Suppl. Fig. 1D) and demonstrate that a joint paraformaldehyde and
NU
methanol fixation protocol is useful for antibodies with different fixation preferences for indirect
MA
immunofluorescence.
Next, we determined whether cytokine-induced nuclear translocation of one factor affects subcellular distribution of the other one. Physical and functional interaction between p65 and
TE
D
STAT3 have been reported in cells after prolonged IL-6 or TNFα treatment [37]. For this reason, we stimulated MEF with TNFα and IL-6 for up to 8 hours and analysed activation and nuclear
AC CE P
presence of both factors by subcellular fractionation (Fig. 1B). Immunoblotting of cytoplasmic and nuclear extracts revealed that TNFα treatment resulted in IκBα phosphorylation and degradation as expected as well as p65 nuclear accumulation after 30 minutes with less pronounced nuclear presence after 4 and 8 hour stimulation time. However, STAT3 activation and subcellular localization was not affected. Similarly, IL-6 induced STAT3 phosphorylation and nuclear translocation without having any significant effect on p65 activation and nuclear presence. Furthermore, these observations were confirmed by simultaneous visualization and quantification of endogenous p65 and STAT3 subcellular distribution under the same conditions via indirect immunofluorescence (Fig. 1C). Therefore, it appears evident that nuclear accumulation of STAT3 or NF-κB does not affect the subcellular distribution of the other transcription factor in wild-type MEF, arguing against a physical association of the two proteins.
10
ACCEPTED MANUSCRIPT 3.2 Absence of STAT3 has no significant effect on canonical TNFα-induced NF-κB activation, nuclear translocation and target gene expression.
PT
In wild-type MEF, upon TNFα treatment serine 536 phosphorylation of p65 peaked after 5
RI
minutes of stimulation, while IκBα was degraded after 15 minutes and returned to normal levels
SC
at 60 minutes time point (Fig. 2A), representing canonical NF-κB signalling [38]. It has been reported that STAT3 ablation or blockade affects NF-κB signalling [21-23]. Using STAT3
NU
knockout MEF, we analysed the impact of STAT3 deletion on canonical TNFα-induced NF-κB
MA
activation. STAT3-/- cells were stimulated with TNFα for indicated time points and compared with similarly treated WT MEF. As seen in Fig. 2A, STAT3-deficient cells showed essentially the same pattern of p65 Ser-536 phosphorylation kinetics, IκBα Ser-32 phosphorylation and
TE
D
degradation in response to TNFα stimulation. Cells deficient in endogenous p65 served as a negative control and showed lower total IκBα and, interestingly, also lower STAT3 levels.
AC CE P
Analysis of GAPDH levels demonstrated equal loading of all lanes. Next, we assessed canonical NF-κB target gene ikba expression changes upon TNFα treatment. As shown by quantitative real time PCR, wild-type and STAT3 deficient fibroblasts have comparable TNFα-induced ikba mRNA fold change levels (Fig. 2B). Moreover, in p65 deficient MEF ikba mRNA upregulation was impaired and stable transfection of p65-YFP fusion protein in these cells reversed the effect, confirming p65 involvement in ikba expression. Finally, we compared p65 nuclear translocation in WT and STAT3-/- MEF using confocal microscopy and subcellular fractionation approaches. In addition to activation, p65 nuclear entry was unaltered after 30 minutes of TNFα stimulation in STAT3-deficient cells compared with WT MEF, as seen by immunofluorescence (Fig. 2C) and immunoblotting of cytoplasmic and nuclear
11
ACCEPTED MANUSCRIPT fractions (Fig. 2D). Taken together, our data show that STAT3 knockout has no major effect on canonical TNFα-stimulated NF-κB signalling in MEF.
PT
3.3 Absence of NF-κB p65 subunit has no influence on IL-6-induced STAT3 activation, but
RI
negatively affects total STAT3 levels.
SC
To analyse the influence of p65 on canonical STAT3 signalling, we stimulated wild-type and p65 deficient cells with IL-6 for 30 minutes and assessed STAT3 tyrosine 705 phosphorylation
NU
via immunoblotting. Both phosphorylation and total levels of endogenous STAT3 were
MA
decreased in p65 knockout cells compared to wild-type (Fig. 3A). Detection of GAPDH verified equal protein loading in both blots. Subsequently, we analysed expression of the STAT3 target
D
gene socs3 upon p65 deletion. As expected, IL-6 treatment led to approximately 10-fold increase
TE
in socs3 mRNA in wild-type MEF and this effect was absent in STAT3-/- cells. In turn, p65
AC CE P
knockout led to an almost 2-fold decrease in socs3 upregulation probably due to lower total STAT3 levels. Stable expression of STAT3-CFP-YFP in STAT3-/- cells complemented the lack of endogenous STAT3 as expected. However, in p65-/- MEF reconstituted with p65-YFP socs3 mRNA upregulation in response to IL-6 was significantly increased compared to wild-type cells (Fig. 3B). Lastly, to assess the influence of p65 knockout on STAT3 nuclear import, we stimulated cells with IL-6 for 30 minutes and detected STAT3 nuclear accumulation by confocal microscopy and immunoblotting of cytoplasmic and nuclear lysates. Despite lower total STAT3 fluorescence signal, p65-/- MEF showed prominent nuclear accumulation of endogenous STAT3 similar to wild-type cells (Fig. 3C). The same pattern was observed in nuclear extracts. (Fig. 3D). These findings show that upon p65 knockout IL-6-induced STAT3 activation is unaffected but total STAT3 levels are decreased resulting in diminished expression of STAT3 target genes upon IL-6 stimulation.
12
ACCEPTED MANUSCRIPT 3.4 NF-B stimulates expression and activation of STAT1 and STAT3, but not STAT5 To further explore the observation of lower total STAT3 levels in p65-/- MEF (Fig. 2A, 3A), we
PT
analysed total protein levels of STAT3, as well as STAT1 and STAT5. In p65-deficient cells
RI
total expression of STAT1 and STAT3, but not STAT5, was downregulated (Fig. 4A). To
SC
examine whether lower levels of STAT3 and STAT1 were not an off-target effect of genetic manipulations in p65-/- cells, we stably transfected these cells with constructs expressing p65-
NU
YFP and the transcriptionally defective p65(S276A)-YFP mutant [39]. Upon expression of p65-
MA
YFP total amounts of STAT1 and STAT3 recovered (Fig. 4A and B) and were comparable to expression levels in wild-type MEF, confirming the involvement of p65 in the regulation of
D
STAT1 and STAT3 expression (Fig. 3B). Moreover, expression of p65(S276A)-YFP did not
TE
restore STAT1 and STAT3 levels, suggesting the requirement of p65 transcriptional activity in regulation of STAT1/3 expression. Interestingly, total STAT5 levels were not affected by these
AC CE P
manipulations (Fig. 4A and B).
Since we could rescue total STAT3 and STAT1 levels upon p65-YFP reconstitution in p65-/cells, which also resulted in an increase in socs3 mRNA upon IL-6 treatment compared to wildtype MEF (Fig. 3B), we studied the effect of p65 overexpression on expression and activation of endogenous STAT3. For this purpose a HeLa Flp-In T-REx system was utilized, which allows for conditional expression of the gene of interest upon doxycycline induction. The cells were stably transfected with a plasmid encoding p65-dsRed. Doxycycline induced and non-induced cells were either stimulated with IL-6 or left unstimulated and cytosolic and nuclear fractions were prepared. Immunoblotting revealed a considerable increase in tyrosine phosphorylation and nuclear accumulation of STAT3 in cells overexpressing p65-dsRed upon IL-6 stimulation (Fig. 4C). Moreover, endogenous STAT1 shows an atypical activation and nuclear presence upon IL-6
13
ACCEPTED MANUSCRIPT treatment when p65 is overexpressed, confirming the influence of p65 levels on both STAT3 and STAT1 signaling. However, in contrast to MEF, p65-dsRed overexpression does not
PT
substantially alter total STAT3 and STAT1 protein amounts in HeLa cells, as judged by
RI
immunoblotting of cytoplasmic fractions (Fig. 4C). In order to prove, that the p65-dsRed overexpression effect on STAT3 overactivation is p65 dependent, we generated two additional
SC
inducible HeLa Flp-In T-REx cell lines stably transfected with p65 or dsRed. These cells were
NU
stimulated with IL-6 or left unstimulated with or without doxycycline induction and total cell lysates were analysed by immunoblotting. Only cells expressing p65 STAT3 showed increased
MA
IL-6-induced phosphorylation upon doxycycline induction (Suppl. Fig. 2A), confirming that the effect observed in Fig. 4B is not due to dsRed induction or doxycycline treatment itself. Taken
TE
D
together, inducible p65 overexpression in HeLa cells leads to an increased STAT3 activation and uncharacteristic STAT1 nuclear presence upon IL-6 stimulation. Of note, upon STAT3
AC CE P
overexpression in HeLa cells IκBα phosphorylation and degradation, as well as p65 nuclear translocation were not affected (Suppl. Fig. 2B). 4 Discussion
STAT3 and NF-κB are pleiotropic transcription factors that are involved in various physiological processes and both are constitutively activated in numerous cancers most often due to activation of upstream signalling pathways in response to autocrine and paracrine signals present within the tumour microenvironment [16]. In recent years, several forms of crosstalk between NF-κB and STAT3 pathways have been described [15, 18]. Here we show that STAT3 has no direct influence on canonical TNFα-induced NF-κB signalling, while NF-κB p65 subunit positively affects STAT3 and STAT1 signalling.
14
ACCEPTED MANUSCRIPT We first defined optimal sample preparation conditions for simultaneous visualization of both p65
and
STAT3
in
cultured
cells
via
immunofluorescence
using
combined
PT
paraformaldehyde/methanol treatment. The choice of the processing method is most important
RI
for appearance of the immunostaining and the interpretation of the results may vary depending on the protocol used [36]. In our hands, NF-κB p65 subunit nuclear accumulation upon TNFα
SC
treatment is clearly detectable after paraformaldehyde fixation but hardly visible after methanol
NU
fixation (Fig. 1A, Suppl. Fig.1A and B). Our findings are in agreement with previously published data that methanol treatment results in a perinuclear localization of p65 in human dendritic cells
MA
upon stimulation [35]. Paraformaldehyde preparation also preserved the intracellular and
TE
FasL proteins in liver cells [40].
D
membrane structures better than methanol and resulted in a more accurate localization of Fas and
In turn, several reports describe the visualization of STAT3 nuclear translocation by microscopy
AC CE P
have using different methods of fixation [34, 41, 42]. Our data demonstrates that paraformaldehyde fixation leads to a predominant nuclear localization of endogenous STAT3 both in resting and IL-6-stimulated cells, while methanol fixation and combined protocols lead to a more uniform distribution of STAT3 in unstimulated cells and a significant increase in nuclear STAT3 after cytokine treatment (Fig. 1A, Suppl. Fig.1A). These observations could explain previously reported prominent nuclear localization of endogenous STAT3 in both resting and activated states in different cell lines [42], which is contradictory to live-cell imaging of ectopically expressed STAT3 fluorescent fusion proteins showing predominant cytoplasmic localization of STAT3 in resting cells and rapid nuclear accumulation upon stimulation [34, 43]. Paraformaldehyde fixation has been shown previously to cause misinterpretation of the subcellular localization of plant proteins causing dissociation of these proteins from metaphase
15
ACCEPTED MANUSCRIPT chromosomes [44]. While paraformaldehyde preserves structural elements at the expense of reduced antigen accessibility, fixation with methanol can disturb structural integrity of the cells
PT
[45]. We found a combined paraformaldehyde/methanol fixation protocol to be optimal for
RI
parallel p65 and STAT3 visualization using antibodies with different fixation preferences. In line with our observations, combined treatment has been reported previously to be more useful for
SC
proper visualization of transmembrane and soluble cytoplasmic proteins than paraformaldehyde
NU
or methanol fixation alone [46, 47]. Using combined fixation and subcellular fractionation approaches, we demonstrate that canonical activation and nuclear entry of STAT3 and NF-κB
MA
are independent of each other (Fig. 1B and D). Our observations from MEF do not reproduce previously observed interactions between the two factors [37], indicating that previously
TE
D
observed different crosstalk forms of STAT3 and NF-κB p65 subunit upon TNFα or IL-6
dependent.
AC CE P
stimulation are not universally applicable, but rather cell-type and extracellular environment
To assess the potential role of STAT3 in canonical NF-κB signalling, we compared TNFαinduced pathway activation in wild-type and STAT3-/- cells. Our results demonstrate that the absence of STAT3 has no significant effect on total p65 levels, p65 phosphorylation on serine 536, IκBα phosphorylation and degradation, as well as subsequent p65 nuclear translocation and ikba gene upregulation (Fig. 2A-D). At least five kinases converge on p65 phosphorylation at serine 536 [48], which is one of the most extensively studied p65 posttranslational modifications [49], suggesting that all molecular steps upstream of p65 phosphorylation remained unaffected
upon STAT3 deletion. In contrast to previously published data [20], the absence of STAT3 had no detrimental effects on TNFα-induced nuclear translocation of NF-κB p65 and target gene expression, as demonstrated by optimized immunostaining protocol and subcellular fractionation
16
ACCEPTED MANUSCRIPT experiments (Fig. 2C-D). IκBα is a classical NF-κB target gene, which has is linked with p65 in a regulatory feedback loop [50]. Deletion of p65, but not STAT3, had detrimental effects on ikba
PT
expression and this effect can be rescued by p65 reconstitution, confirming the dependence of
RI
IκBα expression on NF-κB p65 subunit (Fig. 2B). Since the regulation of several other target genes has been reported to require both p65 and STAT3 DNA association [51-53], genes other
SC
than ikba may have been influenced by STAT3 deletion. However, proper ikba upregulation
NU
indicates that canonical NF-κB signalling is intact in STAT3-/- cells. Furthermore, overexpression of STAT3 also showed no effect on p65 activation and nuclear translocation in
MA
HeLa cells (Suppl. Fig. 2B). These data indicate that STAT3 has no direct influence on canonical NF-κB signalling upon TNFα stimulation, confirming further that previously published
TE
D
observations of STAT3 silencing being beneficial for NF-κB activation in cancer or primary cells [21-23] are not universal, but rather cell-type or stimulus specific, along with the impact of
AC CE P
oncogenesis or the extracellular microenvironment. Moreover, separate or combined activation of p65 and STAT3 in MEF did not result in any detectable physical association of the two factors in coimmunoprecipitation experiments (data not shown), in contrast to previously published observations in coronary artery smooth muscle cells [54]. Knockout of endogenous p65 in MEF led to downregulation of total STAT3 and STAT1 levels, but not STAT5, and this effect was rescued in p65-YFP-complemented p65-/- cells (Fig. 4A and B) confirming the involvement of p65 in controlling total STAT3 levels [55, 56]. Serine phosphorylation of the p65 subunit affects the activity of NF-κB and its interaction with coregulators [57]. Phosphorylation of p65 at serine 276 regulates a subset of NF-κB target genes with specific cis-acting elements in their promoters, such as those encoding IL-6, IL-8, Gro-β, and ICAM-1 [57], as well as allowing for binding of CREB-binding protein (CBP)/p300 [58].
17
ACCEPTED MANUSCRIPT Our data revealed, that serine 276 substitution to alanine in p65 led to a similar decrease in STAT1 and partially STAT3 amounts as observed in p65-deficient cells, indicating that
PT
transcriptional activity of p65 is involved in regulation of STAT1 and STAT3 expression.
RI
Despite lower total amounts of protein, cytokine-induced nuclear import of STAT3 remained
SC
intact in p65-/- cells (Fig. 3B,C) suggesting that IL-6 induced signalling itself was less affected. Interestingly, reconstitution of p65-/- cells with p65-YFP led to an increased socs3 upregulation
NU
in response to IL-6 (Fig. 3D). Several reports suggest a positive association between NF-κB transcriptional activity and STAT3 activation. In the absence of tumour suppressor p53, both
MA
NF-κB and STAT3 were constitutively activated and depleting the expression of p65 led to
D
suppression of STAT3 constitutive activation [55]. Similarly, suppression of NF-κB activity by
TE
p65R siRNA in Sirt1-null MEF reduced mitochondrial function and the expression of STAT3 [56]. In turn, in human hypopharyngeal epithelium cells acidic-bile salts induced substantial
AC CE P
increase in total p65 amounts, which was subsequently correlated with STAT3 upregulation [59]. Moreover, in our hands inducible p65-dsRed overexpression in HeLa cells led to an increased STAT3 activation and nuclear accumulation along with abnormal STAT1 phosphorylation and nuclear presence (Fig. 4C). Under normal conditions in HeLa, as well as other cancer-derived cell lines, IL-6-type cytokines do not induce a significant nuclear accumulation of STAT1 [60]. Yet, in our hands inducible increase of total p65 amount led to a substantial nuclear presence of both activated STAT3 and STAT1 upon IL-6 treatment. Therefore we conclude that p65 supports expression and activation of STAT3 and STAT1, which requires further analysis. STAT3 and STAT1 have been demonstrated to separately associate with NF-κB in the cytoplasm with different effects [37, 61] and recent report demonstrated that higher expression of NFκB in rheumatoid arthritis patients significantly correlated with increased STAT1 and STAT3 amounts
18
ACCEPTED MANUSCRIPT in peripheral leukocytes [62]. While both NF-κB and STAT3 signalling pathways, but not MAPKs, were dysregulated by interferons in STAT1-null macrophages, overactivation of
PT
STAT3 was the key player in downregulation of NF-κB signalling [63]. Additionally, deficiency
RI
of one of the major NF-κB activating receptors TLR4 led to enhanced STAT1-driven Th1 differentiation and suppressed STAT3-driven Th17 expansion [64], whereas IL-27, a crucial
SC
cytokine for bridging innate and adaptive immunity, has been shown to rely on all three
NU
transcription factors for its proper signalling [65]. Taking all this into account, a triple collaboration of STAT1, STAT3 and NF-κB in immunological and pathological processes is
MA
worth exploring further. Taken together, our results suggest that TNFα-induced canonical NF-κB signalling does not rely on STAT3, while expression of the NF-κB p65 subunit positively
AC CE P
Conclusions
TE
D
correlate with total STAT3 and STAT1 levels and augments STAT3/STAT1 activation.
Taken together, our results suggest that STAT3 does not interfere directly with canonical TNFαinduced NF-κB signalling. However, expression of NF-κB positively correlates with total STAT3 and STAT1, but not STAT5, levels and supports STAT3 and STAT1 activation. Furthermore, our study demonstrates that the fixation procedure has a strong influence on the appearance of the subcellular distribution of STAT3 and NF-κB having important implications for the interpretation of published data. An improved immunostaining protocol for reliable staining of both NF-κB and STAT3 with antibodies preferring different fixation protocols and visualization by fluorescence microscopy is provided.
19
ACCEPTED MANUSCRIPT Conflict of interest
PT
The authors disclose no potential conflicts of interest. All authors approved the final article. Acknowledgments
RI
This work was supported by the Studienstiftung des deutschen Volkes (to AM) and the
SC
‘Immunohistochemistry and Confocal Microscopy Facility’, a core facility of the
NU
Interdisciplinary Center for Clinical Research (IZKF) Aachen within the Faculty of Medicine at RWTH Aachen University. MK and KA were funded by the Polish Ministry of Science and
MA
Higher Education under IUVENTUS PLUS grant IP2012016572. KA was supported by the Erasmus+ programme. The authors would like to thank Natalie Rinis for generation of the p65-
D
dsRed HeLa Flp-In T-REx stable cell line, Thomas Wilhelm for help with qRT-PCR and Oxana
AC CE P
TE
Khamidova for help with immunofluorescence image quantification.
20
ACCEPTED MANUSCRIPT References 1.
Levy, DE, and Darnell, JE, Jr. (2002). Stats: transcriptional control and biological impact. Nat Rev
PT
Mol Cell Biol 3: 651-662. Levy, DE, and Lee, CK (2002). What does Stat3 do? J Clin Invest 109: 1143-1148.
3.
Heinrich, PC, Behrmann, I, Haan, S, Hermanns, HM, Müller-Newen, G, and Schaper, F (2003).
RI
2.
Haspel, RL, Salditt-Georgieff, M, and Darnell, JE, Jr. (1996). The rapid inactivation of nuclear
NU
4.
SC
Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J 374: 1-20.
tyrosine phosphorylated Stat1 depends upon a protein tyrosine phosphatase. EMBO J 15: 6262-
5.
MA
6268.
Bhattacharya, S, and Schindler, C (2003). Regulation of Stat3 nuclear export. J Clin Invest 111:
Pranada, AL, Metz, S, Herrmann, A, Heinrich, PC, and Müller-Newen, G (2004). Real time
TE
6.
D
553-559.
7.
AC CE P
analysis of STAT3 nucleocytoplasmic shuttling. J Biol Chem 279: 15114-15123. Herrmann, A, Vogt, M, Mönnigmann, M, Clahsen, T, Sommer, U, Haan, S, et al. (2007). Nucleocytoplasmic shuttling of persistently activated STAT3. J Cell Sci 120: 3249-3261. 8.
Naugler, WE, and Karin, M (2008). The wolf in sheep's clothing: the role of interleukin-6 in immunity, inflammation and cancer. Trends Mol Med 14: 109-119.
9.
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.
10. Vallabhapurapu, S, and Karin, M (2009). Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol 27: 693-733. 11. Hayden, MS, and Ghosh, S (2008). Shared principles in NF-kappaB signaling. Cell 132: 344-362. 12. Natoli, G, and De Santa, F (2006). Shaping alternative NF-kappaB-dependent gene expression programs: new clues to specificity. Cell Death Differ 13: 693-696.
21
ACCEPTED MANUSCRIPT 13. Arenzana-Seisdedos, F, Turpin, P, Rodriguez, M, Thomas, D, Hay, RT, Virelizier, JL, et al. (1997). Nuclear localization of I kappa B alpha promotes active transport of NF-kappa B from the
PT
nucleus to the cytoplasm. J Cell Sci 110 ( Pt 3): 369-378. 14. Chen, L, Fischle, W, Verdin, E, and Greene, WC (2001). Duration of nuclear NF-kappaB action
RI
regulated by reversible acetylation. Science 293: 1653-1657.
SC
15. Grivennikov, SI, and Karin, M (2010). Dangerous liaisons: STAT3 and NF-kappaB collaboration
NU
and crosstalk in cancer. Cytokine Growth Factor Rev 21: 11-19. 16. Grivennikov, SI, and Karin, M (2011). Inflammatory cytokines in cancer: tumour necrosis factor
MA
and interleukin 6 take the stage. Ann Rheum Dis 70 Suppl 1: i104-108. 17. Bode, JG, Albrecht, U, Haussinger, D, Heinrich, PC, and Schaper, F (2012). Hepatic acute phase
D
proteins--regulation by IL-6- and IL-1-type cytokines involving STAT3 and its crosstalk with NF-
TE
kappaB-dependent signaling. Eur J Cell Biol 91: 496-505.
AC CE P
18. Bollrath, J, and Greten, FR (2009). IKK/NF-kappaB and STAT3 pathways: central signalling hubs in inflammation-mediated tumour promotion and metastasis. EMBO Rep 10: 1314-1319. 19. Debidda, M, Wang, L, Zang, H, Poli, V, and Zheng, Y (2005). A role of STAT3 in Rho GTPaseregulated cell migration and proliferation. J Biol Chem 280: 17275-17285. 20. Zouein, FA, Duhe, RJ, Arany, I, Shirey, K, Hosler, JP, Liu, H, et al. (2014). Loss of STAT3 in mouse embryonic fibroblasts reveals its Janus-like actions on mitochondrial function and cell viability. Cytokine 66: 7-16. 21. Kulesza, DW, Carre, T, Chouaib, S, and Kaminska, B (2013). Silencing of the transcription factor STAT3 sensitizes lung cancer cells to DNA damaging drugs, but not to TNFalpha- and NK cytotoxicity. Exp Cell Res 319: 506-516.
22
ACCEPTED MANUSCRIPT 22. McFarland, BC, Gray, GK, Nozell, SE, Hong, SW, and Benveniste, EN (2013). Activation of the NFkappaB pathway by the STAT3 inhibitor JSI-124 in human glioblastoma cells. Mol Cancer Res 11:
PT
494-505. 23. Grabner, B, Schramek, D, Mueller, KM, Moll, HP, Svinka, J, Hoffmann, T, et al. (2015). Disruption
RI
of STAT3 signalling promotes KRAS-induced lung tumorigenesis. Nature communications 6:
SC
6285.
NU
24. Takeda, K, Noguchi, K, Shi, W, Tanaka, T, Matsumoto, M, Yoshida, N, et al. (1997). Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc Natl Acad Sci USA
MA
94: 3801-3804.
25. Takeda, K, Kaisho, T, Yoshida, N, Takeda, J, Kishimoto, T, and Akira, S (1998). Stat3 activation is
D
responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation
TE
and characterization of T cell-specific Stat3-deficient mice. J Immunol 161: 4652-4660.
AC CE P
26. Alonzi, T, Maritano, D, Gorgoni, B, Rizzuto, G, Libert, C, and Poli, V (2001). Essential role of STAT3 in the control of the acute-phase response as revealed by inducible gene inactivation [correction of activation] in the liver. Mol Cell Biol 21: 1621-1632. 27. Costa-Pereira, AP, Tininini, S, Strobl, B, Alonzi, T, Schlaak, JF, Is'harc, H, et al. (2002). Mutational switch of an IL-6 response to an interferon-gamma-like response. Proc Natl Acad Sci USA 99: 8043-8047. 28. Moreno, R, Sobotzik, JM, Schultz, C, and Schmitz, ML (2010). Specification of the NF-kappaB transcriptional response by p65 phosphorylation and TNF-induced nuclear translocation of IKK epsilon. Nucleic Acids Res 38: 6029-6044. 29. Arcone, R, Pucci, P, Zappacosta, F, Fontaine, V, Malorni, A, Marino, G, et al. (1991). Single-step purification and structural characterization of human interleukin-6 produced in Escherichia coli from a T7 RNA polymerase expression vector. Eur J Biochem 198: 541-547.
23
ACCEPTED MANUSCRIPT 30. Weiergräber, O, Hemmann, U, Küster, A, Müller-Newen, G, Schneider, J, Rose-John, S, et al. (1995). Soluble human interleukin-6 receptor: expression in insect cells, purification and
PT
characterization. Eur J Biochem 234: 661-669. 31. Suthaus, J, Tillmann, A, Lorenzen, I, Bulanova, E, Rose-John, S, and Scheller, J (2010). Forced
RI
homo- and heterodimerization of all gp130-type receptor complexes leads to constitutive
SC
ligand-independent signaling and cytokine-independent growth. Mol Biol Cell 21: 2797-2807.
NU
32. Tay S, Hughey JJ, Lee TK, Lipniacki T, Quake SR, Covert MW. Single-cell NF-kappaB dynamics reveal digital activation and analogue information processing. Nature. 2010, 466(7303):267-71.
MA
33. Livak, KJ, and Schmittgen, TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402-408.
D
34. Vogt, M, Domoszlai, T, Kleshchanok, D, Lehmann, S, Schmitt, A, Poli, V, et al. (2011). The role of
AC CE P
Sci 124: 900-909.
TE
the N-terminal domain in dimerization and nucleocytoplasmic shuttling of latent STAT3. J Cell
35. Blaecke, A, Delneste, Y, Herbault, N, Jeannin, P, Bonnefoy, JY, Beck, A, et al. (2002). Measurement of nuclear factor-kappa B translocation on lipopolysaccharide-activated human dendritic cells by confocal microscopy and flow cytometry. Cytometry 48: 71-79. 36. Schnell, U, Dijk, F, Sjollema, KA, and Giepmans, BN (2012). Immunolabeling artifacts and the need for live-cell imaging. Nat Methods 9: 152-158. 37. Yang, J, Liao, X, Agarwal, MK, Barnes, L, Auron, PE, and Stark, GR (2007). Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes Dev 21: 1396-1408. 38. Adli, M, and Baldwin, AS (2006). IKK-i/IKKepsilon controls constitutive, cancer cell-associated NF-kappaB activity via regulation of Ser-536 p65/RelA phosphorylation. J Biol Chem 281: 2697626984.
24
ACCEPTED MANUSCRIPT 39. Dong J, Jimi E, Zhong H, Hayden MS, Ghosh S. Repression of gene expression by unphosphorylated NF-kappaB p65 through epigenetic mechanisms. Genes Dev. 2008,
PT
22(9):1159-73. 40. Vekemans, K, Rosseel, L, Wisse, E, and Braet, F (2004). Immuno-localization of Fas and FasL in
RI
rat hepatic endothelial cells: influence of different fixation protocols. Micron 35: 303-306.
SC
41. Ray, S, Boldogh, I, and Brasier, AR (2005). STAT3 NH2-terminal acetylation is activated by the
NU
hepatic acute-phase response and required for IL-6 induction of angiotensinogen. Gastroenterology 129: 1616-1632.
MA
42. Liu, L, McBride, KM, and Reich, NC (2005). STAT3 nuclear import is independent of tyrosine phosphorylation and mediated by importin-alpha3. Proc Natl Acad Sci USA 102: 8150-8155.
D
43. Martincuks, A, Fahrenkamp, D, Haan, S, Herrmann, A, Kuster, A, and Muller-Newen, G (2016).
AC CE P
trafficking. Cell Signal.
TE
Dissecting functions of the N-terminal domain and GAS-site recognition in STAT3 nuclear
44. Li, MW, Zhou, L, and Lam, HM (2015). Paraformaldehyde Fixation May Lead to Misinterpretation of the Subcellular Localization of Plant High Mobility Group Box Proteins. PLoS One 10: e0135033.
45. Smith-Clerc, J, and Hinz, B (2010). Immunofluorescence detection of the cytoskeleton and extracellular matrix in tissue and cultured cells. Methods Mol Biol 611: 43-57. 46. Pollice, AA, McCoy, JP, Jr., Shackney, SE, Smith, CA, Agarwal, J, Burholt, DR, et al. (1992). Sequential paraformaldehyde and methanol fixation for simultaneous flow cytometric analysis of DNA, cell surface proteins, and intracellular proteins. Cytometry 13: 432-444. 47. Brock, R, Hamelers, IH, and Jovin, TM (1999). Comparison of fixation protocols for adherent cultured cells applied to a GFP fusion protein of the epidermal growth factor receptor. Cytometry 35: 353-362.
25
ACCEPTED MANUSCRIPT 48. Buss, H, Dorrie, A, Schmitz, ML, Hoffmann, E, Resch, K, and Kracht, M (2004). Constitutive and interleukin-1-inducible phosphorylation of p65 NF-{kappa}B at serine 536 is mediated by
PT
multiple protein kinases including I{kappa}B kinase (IKK)-{alpha}, IKK[60], IKK{epsilon}, TRAF family member-associated (TANK)-binding kinase 1 (TBK1), and an unknown kinase and couples
RI
p65 to TATA-binding protein-associated factor II31-mediated interleukin-8 transcription. J Biol
SC
Chem 279: 55633-55643.
NU
49. Schmitz, ML, Mattioli, I, Buss, H, and Kracht, M (2004). NF-kappaB: a multifaceted transcription factor regulated at several levels. Chembiochem 5: 1348-1358.
MA
50. Scott, ML, Fujita, T, Liou, HC, Nolan, GP, and Baltimore, D (1993). The p65 subunit of NF-kappa B regulates I kappa B by two distinct mechanisms. Genes Dev 7: 1266-1276.
D
51. Yu, Z, Zhang, W, and Kone, BC (2002). Signal transducers and activators of transcription 3
TE
(STAT3) inhibits transcription of the inducible nitric oxide synthase gene by interacting with
AC CE P
nuclear factor kappaB. Biochem J 367: 97-105. 52. Agrawal, A, Cha-Molstad, H, Samols, D, and Kushner, I (2003). Overexpressed nuclear factorkappaB can participate in endogenous C-reactive protein induction, and enhances the effects of C/EBPbeta and signal transducer and activator of transcription-3. Immunology 108: 539-547. 53. Hagihara, K, Nishikawa, T, Sugamata, Y, Song, J, Isobe, T, Taga, T, et al. (2005). Essential role of STAT3 in cytokine-driven NF-kappaB-mediated serum amyloid A gene expression. Genes Cells 10: 1051-1063. 54. Dhar, K, Rakesh, K, Pankajakshan, D, and Agrawal, DK (2013). SOCS3 promotor hypermethylation and STAT3-NF-kappaB interaction downregulate SOCS3 expression in human coronary artery smooth muscle cells. Am J Physiol Heart Circ Physiol 304: H776-785. 55. Guo, AK, Hou, YY, Hirata, H, Yamauchi, S, Yip, AK, Chiam, KH, et al. (2014). Loss of p53 enhances NF-kappaB-dependent lamellipodia formation. J Cell Physiol 229: 696-704.
26
ACCEPTED MANUSCRIPT 56. Bernier, M, Paul, RK, Martin-Montalvo, A, Scheibye-Knudsen, M, Song, S, He, HJ, et al. (2011). Negative regulation of STAT3 protein-mediated cellular respiration by SIRT1 protein. J Biol
PT
Chem 286: 19270-19279. 57. Anrather J, Racchumi G, Iadecola C. cis-acting, element-specific transcriptional activity of
RI
differentially phosphorylated nuclear factor-kappa B. J Biol Chem. 2005, 280(1):244-52
SC
58. Zhong H, Voll RE, and Ghosh S. Phosphorylation of NF-kappa B p65 by PKA stimulates
CBP/p300. Mol. Cell, 1998, 1, 661-71.
NU
transcriptional activity by promoting a novel bivalent interaction with the coactivator
MA
59. Sasaki, CT, Issaeva, N, and Vageli, DP (2015). In vitro model for gastroduodenal reflux-induced nuclear factor-kappaB activation and its role in hypopharyngeal carcinogenesis. Head Neck.
D
60. Haan S, Keller JF, Behrmann I, Heinrich PC, Haan C. Multiple reasons for an inefficient STAT1
TE
response upon IL-6-type cytokine stimulation. Cell Signal. 2005, 17(12):1542-50.
AC CE P
61. Kramer, OH, Baus, D, Knauer, SK, Stein, S, Jager, E, Stauber, RH, et al. (2006). Acetylation of Stat1 modulates NF-kappaB activity. Genes Dev 20: 473-485. 62. Świerkot J, Nowak B, Czarny A, Zaczyńska E, Sokolik R, Madej M, Korman L, Sebastian A, Wojtala P, Lubiński Ł, Wiland P. The Activity of JAK/STAT and NF-κB in Patients with Rheumatoid Arthritis. Adv Clin Exp Med. 2016, 25(4):709-17. 63. Kim, HS, Kim, DC, Kim, HM, Kwon, HJ, Kwon, SJ, Kang, SJ, et al. (2015). STAT1 deficiency redirects IFN signalling toward suppression of TLR response through a feedback activation of STAT3. Sci Rep 5: 13414. 64. Xu, QQ, Zhou, Q, Xu, LL, Lin, H, Wang, XJ, Ma, WL, et al. (2015). Toll-like receptor 4 signaling inhibits malignant pleural effusion by altering Th1/Th17 responses. Cell Biol Int 39: 1120-1130.
27
ACCEPTED MANUSCRIPT 65. Guzzo, C, Che Mat, NF, and Gee, K (2010). Interleukin-27 induces a STAT1/3- and NF-kappaBdependent proinflammatory cytokine profile in human monocytes. J Biol Chem 285: 24404-
AC CE P
TE
D
MA
NU
SC
RI
PT
24411.
28
ACCEPTED MANUSCRIPT Figure legends Figure 1: Analysis of STAT3 and NF-κB canonical activation. (A) Quantification of nuclear
PT
p65 and STAT3 via indirect immunofluorescence following 3.7% paraformaldehyde (PFA), methanol (MetOH) or combined (PFA+MetOH) fixation (means ± SD of app. n=100 cells per
RI
sample). MEF were stimulated with 10 ng/ml TNFα or 20 ng/ml IL-6 with 500 ng/ml sIL-6Rα for 30 minutes, fixed either with 3.7% PFA, methanol or combined 3.7% PFA and methanol
SC
treatment, permeabilized and stained using specific p65 and STAT3 antibodies, followed by incubation with secondary Alexa Fluor-555 and Alexa Fluor-488 conjugated antibodies
NU
respectively. ***p < 0.0005. *p < 0.05. n.s. = not significant. (B) Analysis of cytokine-induced STAT3 and p65 nuclear accumulation by cell fractionation. Wild-type MEF were stimulated
MA
with 20 ng/ml TNFα or 10 ng/ml IL-6 with 500 ng/ml sIL-6Rα for 30, 240 and 480 minutes or left untreated. Afterwards, cytoplasmic and nuclear extracts were prepared and immunoblotted with indicated antibodies. GAPDH served as cytoplasmic marker, lamin A/C served as nuclear
D
fraction loading control. Upper band represents lamin A, lower band represents lamin C. (C)
TE
Simultaneous visualization of p65 and STAT3 subcellular localization following TNFα treatment. Left: Cells were stimulated with 20 ng/ml TNFα for indicated time points or left
AC CE P
untreated, incubated with primary p65 and STAT3 antibodies, followed by immunostaining with Alexa Fluor-555 and Alexa Fluor-488 antibodies, respectively, as well as DRAQ5 nuclear marker. Right: Nuclear presence of p65 and STAT3 was quantified using MATLAB software (means ± SD of app. n=100 cells per sample). ***p < 0.0005. **p < 0.005. *p < 0.05. (D) Simultaneous visualization of p65 and STAT3 subcellular localization following IL-6 treatment. Cells were stimulated with 10 ng/ml IL-6 with 500 ng/ml sIL-6Rα for indicated time points and samples were prepared, visualized (left) and quantified (right) similarly to Fig.1C. Scale bars represent 10 μm. Figure 2: NF-κB activation, target gene expression and nuclear translocation in wild-type, STAT3-/- and p65-/- MEF upon TNFα stimulation. (A) Analysis of p65 serine 536 phosphorylation and IκBα phosphorylation and degradation. Top: cell lines were stimulated with 20 ng/ml TNFα for 5, 15 and 60 minutes or left untreated. Total cell lysates were prepared and immunoblotted with the indicated antibodies. GAPDH immunodetection served as a loading control. Bottom: Phospho(S536)-p65 and total IκBα signals from three independent
29
ACCEPTED MANUSCRIPT immunoblotting experiments were quantified using Fujifilm MultiGauge v3.2 software. ***p < 0.0005. **p < 0.005. *p < 0.05. n.s. = not significant. (B) IκBα mRNA expression upon TNFα treatment. Wild-type MEF, STAT3-/-, p65-/- cells, as well as p65-/- cells stably transfected with
PT
a fluorescent p65-YFP construct were treated with 10 ng/ml TNFα for 45 minutes. Whole cellular mRNA was isolated, and the expression of ikba was measured by RT-qPCR. Expression
RI
was normalized to the housekeeper protein mGUSB. Data are representative of three
SC
independent experiments. ***p < 0.0005. n.s. = not significant. (C) Subcellular localization of p65 and STAT3 following 30 minutes of 20 ng/ml TNFα stimulation. Left: Indicated cell lines
NU
were fixed with combined PFA/methanol treatment, incubated with primary p65 and STAT3 antibodies, followed by immunostaining with Alexa Fluor-555 and Alexa Fluor-488 antibodies,
MA
respectively, as well as DRAQ5 nuclear marker. Scale bars represent 10 μm. Right: nuclear presence of p65 was quantified using MATLAB software (means ± SD of app. n=100 cells per sample). ***p < 0.0005. n.s. = not significant. (D) Analysis of p65 nuclear accumulation by cell
D
fractionation. Wild-type and STAT3-/- MEF were stimulated with 20 ng/ml TNFα for 30
TE
minutes, followed by cytoplasmic and nuclear extract preparation and immunoblotting with p65 antibody. Lamin A/C served as nuclear fraction loading control, GAPDH served as cytoplasmic
AC CE P
marker.
Figure 3: STAT3 activation, nuclear translocation and target gene expression in wild-type, p65-/- and STAT3-/- cells upon IL-6 stimulation. (A) Analysis of STAT3 tyrosine 705 phosphorylation. Cell lines were stimulated with 10 ng/ml IL-6 and 500 ng/ml soluble IL-6R (sIL-6Rα) for 30 minutes or left untreated. Total cell lysates were prepared and immunoblotted with the indicated antibodies. GAPDH immunodetection served as a loading control. (B) Increase in SOCS3 mRNA expression upon IL-6 treatment. Wild-type MEF, STAT3-/-, p65-/cells, as well as STAT3-/- MEF stably transfected with STAT3-CFP-YFP and p65-/- cells stably transfected with fluorescent p65-YFP construct were treated with 20 ng/ml IL-6 and 500 ng/ml soluble IL-6R (sIL-6Rα) for 30 minutes. Whole cellular mRNA was isolated, and the expression of socs3 was measured by RT-qPCR. Expression was normalized to the housekeeper protein mGUSB. Data are representative of three independent experiments. *p < 0.0005. ***p < 0.0005. (C) Subcellular localization of p65 and STAT3 following 30 minutes of 10 ng/ml IL-6 and 500 ng/ml soluble IL-6R (sIL-6Rα) stimulation. Left: indicated cell lines were fixed with combined PFA/methanol treatment, incubated with primary p65 and STAT3 antibodies, followed by
30
ACCEPTED MANUSCRIPT immunostaining with Alexa Fluor-555 and Alexa Fluor-488 antibodies, respectively, as well as DRAQ5 nuclear marker. Scale bars represent 10 μm. Right: nuclear presence of STAT3 was quantified using MATLAB software (means ± SD of app. n=100 cells per sample). ***p <
PT
0.0005. n.s. = not significant. (D) Analysis of STAT3 nuclear accumulation by cell fractionation. Wild-type and p65-/- MEF were stimulated with 10 ng/ml IL-6 and 500 ng/ml soluble IL-6R
RI
(sIL-6Rα) for 30 minutes, followed by cytoplasmic and nuclear extract preparation and
SC
immunoblotting with STAT3 antibodies. Lamin A/C served as nuclear fraction loading control, GAPDH served as cytoplasmic marker.
NU
Figure 4: Effect of p65 presence on STAT1, STAT3 and STAT5 expression. (A) Analysis of total STAT1, STAT3 and STAT5 levels upon p65-YFP reconstitution. Total cell lysates of WT,
MA
p65 -/- and p65 -/- MEF stably transfected with p65-YFP or p65(S276A)-YFP were prepared and immunoblotted with the indicated antibodies. (B) Quantification of total STAT1, STAT3 and STAT5 levels from cell lines analyzed in Fig.4A. Immunoblots from three independent
D
experiments were quantified using Fujifilm MultiGauge v3.2 software. *p < 0.05. **p < 0.005.
TE
***p < 0.0005. n.s. = not significant. (C) STAT3 activation and nuclear translocation of STAT1 and STAT3 after p65-dsRed overexpression. HeLa Flp-In T-REx cells stably transfected with
AC CE P
p65-dsRed fluorescent fusion protein were left untreated or induced with 10 ng/ml doxycycline for 24 h to induce the expression of the transgene. Subsequently, cells were stimulated with 20 ng/ml IL-6 and 500 ng/ml soluble IL-6R (sIL-6Rα) for 30 minutes and cytosolic and nuclear extracts were prepared and analysed by immunoblotting with the indicated antibodies. Antibodies against GAPDH and lamin A/C were used as cytoplasmic and nuclear loading controls respectively.
31
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
32
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
33
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
34
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
35
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
36
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
37
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
38
AC CE P
TE
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
39
ACCEPTED MANUSCRIPT Highlights Reliable parallel detection of p65 and STAT3 subcellular distribution
Subcellular distribution of NF-κB and STAT3 is independent of each other
STAT3 deletion or overexpression does not affect canonical TNFα-NF-κB signalling
Deletion of p65 reduces expression of STAT3 and STAT1
Overexpression of p65 increases STAT3 and STAT1 activation in response to IL-6
AC CE P
TE
D
MA
NU
SC
RI
PT
40