IGF-1 induced HIF-1α-TLR9 cross talk regulates inflammatory responses in glioma

7
IGF-1 induced HIF-1α-TLR9 cross talk regulates inammatory responses in glioma Sanchari Sinha, Nitin Koul, Deobrat Dixit, Vivek Sharma, Ellora Sen National Brain Research Centre, Manesar, Haryana 122 050, India abstract article info Article history: Received 10 June 2011 Accepted 27 June 2011 Available online 3 July 2011 Keywords: IGF-1 HIF-1α TLR9 Glioblastoma The insulin-like growth factor (IGF-1) induces hypoxia inducible factor (HIF-1α) regulated genes in glioblastoma multiforme (GBM). As HIF-1α links inammatory and oncogenic pathways in GBM, we investigated whether IGF-1 affects HIF-1α to regulate inammatory response in glioma cells under normoxia. IGF-1 induced Ras and Calmodulin-dependent kinase II (CaMKII) regulated HIF-1α transcriptional activity in glioma cells. Increase in HIF-1α was concurrent with decreased Toll-like receptor (TLR9) and CXCR4 expression and elevated suppressor of cytokine signaling (SOCS3) levels. Interestingly, while synthetic CpG containing oligodeoxynucleotide TLR9 agonist (CpG DNA) decreased IGF-1 mediated increase in HIF-1α activity, siRNA mediated knockdown of HIF-1α decreased TLR9 levels. This suggested that IGF-1 induced HIF- 1α-TLR9 axis is regulated by both positive and negative feedback loops. Importantly, TLR9 agonist reversed the effect of IGF-1 on CXCR4 and SOCS3 expression. While knockdown of HIF-1α abrogated IGF-1 mediated increase in SOCS3 it elevated IGF-1 induced decrease in CXCR4 levels. Thus HIF-1α positively and negatively regulates SOCS3 and CXCR4 expression respectively, in glioma cells. Though TLR9 agonist had no additive effect on IGF-1 mediated increase in pro-inammatory cytokines IL-1β, IL-6 and IL-8, treatment with TLR9 agonist alone elevated expression of these pro-inammatory cytokines. Our studies indicate that a complex HIF-1α-TLR9 cross-talk sustains a self-regulating cycle of inammatory response through intrinsic negative and positive feedback mechanisms. © 2011 Elsevier Inc. All rights reserved. 1. Introduction Signaling pathways emanating from the insulin-like growth factor-I receptor (IGF-IR) a transmembrane receptor tyrosine kinase activated by IGF, is associated with the growth and prolifer- ation of several tumor types including glioblastoma [13]. Although HIF-1α is a key regulator of cellular response to hypoxia [4] it can also be activated under normoxia in response to IGF [5,6]. HIF-1α inuences tumor growth [7] by regulating expression of genes implicated in angiogenesis, metabolism and cell survival [8]. IGF-1 is known to induce HIF-1α accumulation in Kaposi Sarcoma [9] and neuroblastoma [10]. HIF-1α-regulated genes are induced by IGF-1 in glioblastoma [11]. Suppression of IGF-1R inhibits tumorigenic potential of rat glioblas- toma cells [12]. We have demonstrated IL-1β induced Ras/Akt/Erk mediated HIF-1α activation in glioma cells [13]. Activated Ras elevates IGF-I mediated induction of the HIF-1α target VEGF [14]. Oncogenic Ras activation occurs in GBMs [15] and inhibition of Ras down- regulates HIF-1α activity in GBM [16]. CaMKII which translates intracellular changes in calcium is associated with glioma migration [17], and regulates Ras mediated Erk activation [18]. CaMKII regulates HIF-1α transcriptional activity under intermittent hypoxia [19] as well as under inammatory conditions [20]. By demonstrating that Ras rewires HIF1α-driven IL-1β autocrine loop [13,21], we have emphasized the pivotal role of HIF-1α in linking inammation and tumorigenesis [22]. HIF-1α is involved in host immune response during bacterial infection [23], and LPS is a potent inducer of HIF-1α [24]. Also, HIF-1α accumulation and target gene expression are impaired upon induction of endotoxin tolerance [25]. Activation of toll like receptors (TLRs) which recognize pathogen- associated molecular patterns triggers signaling events that initiate innate immunity and inammatory response [26]. Interestingly, HIF-1α regulates hypoxia induced TLR4 expression in macrophages [27]. While TLR9 increases metastatic potential of cancer cells through CXCR4 expression [28], the later modulates TLR9 mediated signaling [29]. TLR activation is modulated by negative regulators such as suppressor of cytokine signaling (SOCS) that feed back upon and inhibit TLR activation [30]. Moreover, SOCS3 which is constitutively expressed in GBM is not only involved in inducing radioresistance [31], but it also regulates CXCR4 function [32]. Given that TLR9 is expressed in GBM [33], we investigated whether IGF-1 affect HIF-1α to modulate TLR9 mediated signaling in glioma. Our study to understand the involvement of IGF-1 in HIF-1α regulated inammatory responses in GBM indicates that IGF-1 induced Ras/CaMKII enhances HIF-1α activation, under normoxia. Increase in Cellular Signalling 23 (2011) 18691875 Corresponding author. Tel.: + 91 124 2338921x235; fax: + 91 124 2338910/28. E-mail addresses: [email protected] (S. Sinha), [email protected] (N. Koul), [email protected] (D. Dixit), [email protected] (V. Sharma), [email protected] (E. Sen). 0898-6568/$ see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.cellsig.2011.06.024 Contents lists available at ScienceDirect Cellular Signalling journal homepage: www.elsevier.com/locate/cellsig

Transcript of IGF-1 induced HIF-1α-TLR9 cross talk regulates inflammatory responses in glioma

Cellular Signalling 23 (2011) 1869–1875

Contents lists available at ScienceDirect

Cellular Signalling

j ourna l homepage: www.e lsev ie r.com/ locate /ce l l s ig

IGF-1 induced HIF-1α-TLR9 cross talk regulates inflammatory responses in glioma

Sanchari Sinha, Nitin Koul, Deobrat Dixit, Vivek Sharma, Ellora Sen ⁎National Brain Research Centre, Manesar, Haryana 122 050, India

⁎ Corresponding author. Tel.: +91 124 2338921x235E-mail addresses: [email protected] (S. Si

(N. Koul), [email protected] (D. Dixit), viveksharmabt@[email protected] (E. Sen).

0898-6568/$ – see front matter © 2011 Elsevier Inc. Aldoi:10.1016/j.cellsig.2011.06.024

a b s t r a c t

a r t i c l e i n f o

Article history:Received 10 June 2011Accepted 27 June 2011Available online 3 July 2011

Keywords:IGF-1HIF-1αTLR9Glioblastoma

The insulin-like growth factor (IGF-1) induces hypoxia inducible factor (HIF-1α) regulated genes inglioblastoma multiforme (GBM). As HIF-1α links inflammatory and oncogenic pathways in GBM, weinvestigated whether IGF-1 affects HIF-1α to regulate inflammatory response in glioma cells under normoxia.IGF-1 induced Ras and Calmodulin-dependent kinase II (CaMKII) regulated HIF-1α transcriptional activity inglioma cells. Increase in HIF-1α was concurrent with decreased Toll-like receptor (TLR9) and CXCR4expression and elevated suppressor of cytokine signaling (SOCS3) levels. Interestingly, while synthetic CpGcontaining oligodeoxynucleotide TLR9 agonist (CpG DNA) decreased IGF-1 mediated increase in HIF-1αactivity, siRNA mediated knockdown of HIF-1α decreased TLR9 levels. This suggested that IGF-1 induced HIF-1α-TLR9 axis is regulated by both positive and negative feedback loops. Importantly, TLR9 agonist reversedthe effect of IGF-1 on CXCR4 and SOCS3 expression. While knockdown of HIF-1α abrogated IGF-1 mediatedincrease in SOCS3 it elevated IGF-1 induced decrease in CXCR4 levels. Thus HIF-1α positively and negativelyregulates SOCS3 and CXCR4 expression respectively, in glioma cells. Though TLR9 agonist had no additiveeffect on IGF-1 mediated increase in pro-inflammatory cytokines IL-1β, IL-6 and IL-8, treatment with TLR9agonist alone elevated expression of these pro-inflammatory cytokines. Our studies indicate that a complexHIF-1α-TLR9 cross-talk sustains a self-regulating cycle of inflammatory response through intrinsic negativeand positive feedback mechanisms.

; fax: +91 124 2338910/28.nha), [email protected] (V. Sharma),

l rights reserved.

© 2011 Elsevier Inc. All rights reserved.

1. Introduction

Signaling pathways emanating from the insulin-like growthfactor-I receptor (IGF-IR) — a transmembrane receptor tyrosinekinase activated by IGF, is associated with the growth and prolifer-ation of several tumor types including glioblastoma [1–3]. AlthoughHIF-1α is a key regulator of cellular response to hypoxia [4] it can alsobe activated under normoxia in response to IGF [5,6]. HIF-1αinfluences tumor growth [7] by regulating expression of genesimplicated in angiogenesis, metabolism and cell survival [8]. IGF-1 isknown to induce HIF-1α accumulation in Kaposi Sarcoma [9] andneuroblastoma [10].

HIF-1α-regulated genes are induced by IGF-1 in glioblastoma [11].Suppression of IGF-1R inhibits tumorigenic potential of rat glioblas-toma cells [12]. We have demonstrated IL-1β induced Ras/Akt/Erkmediated HIF-1α activation in glioma cells [13]. Activated Ras elevatesIGF-I mediated induction of the HIF-1α target VEGF [14]. OncogenicRas activation occurs in GBMs [15] and inhibition of Ras down-regulates HIF-1α activity in GBM [16]. CaMKII which translatesintracellular changes in calcium is associated with glioma migration

[17], and regulates Ras mediated Erk activation [18]. CaMKII regulatesHIF-1α transcriptional activity under intermittent hypoxia [19] aswell as under inflammatory conditions [20].

By demonstrating that Ras rewires HIF1α-driven IL-1β autocrineloop [13,21], we have emphasized the pivotal role of HIF-1α in linkinginflammation and tumorigenesis [22]. HIF-1α is involved in hostimmune response during bacterial infection [23], and LPS is a potentinducer of HIF-1α [24]. Also, HIF-1α accumulation and target geneexpression are impaired upon induction of endotoxin tolerance [25].Activation of toll like receptors (TLRs) which recognize pathogen-associated molecular patterns triggers signaling events that initiateinnate immunity and inflammatory response [26]. Interestingly, HIF-1αregulates hypoxia induced TLR4 expression inmacrophages [27]. WhileTLR9 increases metastatic potential of cancer cells through CXCR4expression [28], the later modulates TLR9 mediated signaling [29]. TLRactivation is modulated by negative regulators such as suppressor ofcytokine signaling (SOCS) that feed backupon and inhibit TLR activation[30]. Moreover, SOCS3 which is constitutively expressed in GBM is notonly involved in inducing radioresistance [31], but it also regulatesCXCR4 function [32]. Given that TLR9 is expressed in GBM [33], weinvestigated whether IGF-1 affect HIF-1α to modulate TLR9 mediatedsignaling in glioma.

Our study to understand the involvement of IGF-1 in HIF-1αregulated inflammatory responses in GBM indicates that IGF-1 inducedRas/CaMKII enhances HIF-1α activation, under normoxia. Increase in

Fig. 1. IGF-1 induces HIF-1α activity in glioma cells in a Ras dependent manner (a)Western blot demonstrates a dose dependent increase in HIF-1α expression in glioma cells treatedwith IGF-1 for 24 h. The figure is a representative of three independent experiments. Blots were reprobed for C23 to establish equivalent loading. (b) Treatment with IGF-1 increasesHIF-1α transcriptional activity in glioma cells. Following transfection of glioma cells with HIF-1α reporter constructs, cells were treated with different concentration of IGF-1 for 24 hand reporter assay was performed to determine HIF-1α activity. The graph represents fold change in activity over control. (c) IGF-1 increases Ras activity in glioma cells. Ras activityin IGF-1 treated glioma cells was determined by the ability of Ras-GTP to bind to a specific protein domain of Raf in the form of a GST-fusion protein. An increase in Ras activity wasobserved in cells treated with IGF-1 for 30 mins. The figure is representative from three independent experiments with similar results. (d) DN-RasN17 inhibits the ability of IGF-1 toinduce HIF-1α activity. Cells co-transfected with RasN17 and HIF-1α luciferase reporter constructs were treated with IGF-1. After 24 h luciferase reporter assay was performed todetermine HIF-1α activity. The graph represents fold change in activity over control. Values in b and d represent the means±SEM from 3 independent experiments. * Significantincrease from control, # Significant decrease from IGF-1 treated cells (Pb0.05).

1870 S. Sinha et al. / Cellular Signalling 23 (2011) 1869–1875

IGF-1 induced HIF-1α is concurrent with decrease TLR9 and CXCR4levels and elevation in SOCS3 expression. Interestingly, IGF-1 inducesbothnegative andpositiveHIF-1α-TLR9 loops in glioma. IGF-1mediatedchanges in SOCS3 and CXCR4 is dependent on both HIF-1α and TLR9.This work forges the first link between IGF-1 induced HIF-1α inregulating TLR9 dependent inflammatory responses in glioma.

2. Materials and methods

2.1. Cell culture

Glioblastoma cell lines U87MG [13] and T98G [13] obtained fromAmerican Type Culture Collection (ATCC) were cultured in DMEMsupplemented with 10% FBS. On attaining semi-confluence, cells wereswitched to serum free media (SFM) and after 6 h, cells were treatedwith different concentration of IGF-1 in SFM. All reagents werepurchased from Sigma unless otherwise stated. The HIF-1α luciferasereporter was a gift by Chinmay Mukhopadhyay (JNU, India) and hasbeen described [34]. DN-RasN17 was purchased from Clontech.

2.2. Western blot analysis

Whole cell lysates and nuclear extract were prepared from cellstreated with or without IGF-1, in the presence or absence of TLR9agonist CpG ODN 2006 (Imgenex) or CaMKII inhibitor KN93 asdescribed previously [13], and protein concentration was determinedby the BCA method. Lysates were electrophoresed on 7% to 12%polyacrylamide gel and Western blot analysis was performed asdescribed [13] using the following antibodies — HIF-1α (ΒD bio-sciences), C23 (Santa Cruz), CaMKII, CXCR4, SOCS3 and TLR9(Imgenex). Antibodies were purchased from Cell Signaling unlessotherwise mentioned.

2.3. Measurement of Ras activity

The Ras activity was performed using a commercially available Rasactivation assay kit purchased fromUpstate Biotechnology, as describedpreviously [13].

2.4. Luciferase assay

Transfection and reporter assay was performed as described [13].Briefly, cells at ~60–70% confluence in 24-well plates were transientlytransfected with 0.3 μg of HIF-1α and 10 ng of the Renilla luciferaseexpression vector pRL-TK as a transfection control using Lipofecta-mine 2000 (Life Technologies-Invitrogen). Briefly, 18 h after trans-fection, cells were serum starved for 4 h followed by treatment with20 ng/ml IGF-1 for 24 h. Luciferase activity was measured using Dualluciferase assay kit according to the manufacturer's protocol (Pro-mega) with GloMax 96 microplate luminometer. The results areexpressed as fold change in activity over control. HIF-1α transcrip-tional activity in the presence of dominant negative Ras constructswere performed by co-transfecting cells with 0.3 μg of RasN17 andHIF-1α reporter constructs. For co-transfection experiments involvingDN construct, control transfection using the appropriate emptyvectors for each construct was used as described [13].

2.5. Small interfering RNA (siRNA) transfection

Eighteen hours prior to transfection, 3 x104 cells were seeded onto24-well plates in medium without antibiotics and transfection with50 nmol/l duplex HIF-1α siRNA (Thermo Fischer Scientific) was carriedusing Lipofectamine 2000 (Life Technologies-Invitrogen) as described[13]. Non-specific siRNA was purchased from Proligo (Singapore).

2.6. Cytokine bead array

Cytometric bead array kit (CBA kit; BD Biosciences) was used toquantitatively measure cytokine levels in the supernatant collectedfrom control and glioma cells treated with different combinations ofIGF-1 and TLR9 agonist CpG ODN 2006 (Imgenex). The assay wasperformed and analyzed on FACS Calibur (Becton Dickinson) asdescribed previously [35].

2.7. Statistical analysis

All comparisons between groups were performed using two-tailedPaired student's t-test. A value of Pb0.05 was considered significant.

1871S. Sinha et al. / Cellular Signalling 23 (2011) 1869–1875

3. Results

3.1. IGF-1 induces HIF-1α expression and transcriptional activity inglioma cells

As IGF-1 induces HIF-1α-regulated genes in glioblastoma [11], wedetermined whether IGF-1 effects HIF-1α expression in glioma. IGF-1elevated HIF-1α expression in glioma cells in a dose dependentmanner as compared to control (Fig. 1a). While treatment with 5 and10 ng/ml of IGF-1 slightly elevated HIF-1α expression, maximalincrease was observed upon treatment with 20 ng/ml of IGF-1 for24 h (Fig. 1a). A significant ~2.3-fold increase in HIF-1α transcrip-tional activity over control was observed in glioma cells treated with10 and 20 ng/ml of IGF-1 for 24 h (Fig. 1b). As increase in HIF-1αactivation was maximal in cells treated with 20 ng/ml of IGF-1 allsubsequent experiments were performed with this dose of IGF-1.

3.2. IGF-1 mediated increase in Ras activity regulates HIF-1α activity

IGF signaling promotes cellular proliferation by stimulatingRas/ERK pathway necessary for tumor growth [6]. Since Ras activationoccurs in GBM [15], we determined whether IGF-1 regulates Ras

Fig. 2. CaMKII regulate IGF-1 mediated HIF-1α activity in a Ras independent manner.(a) IGF-1 increases CaMKII phosphorylation in glioma cells as determined by Westernblot analysis. The figure is a representative from three independent experiments withsimilar results. (b) IGF-1 mediated increase in CaMKII expression is unaltered in cellstransfected with DN-RasN17. Mock and DN-RasN17 transfected cells were treated withIGF-1 for 24 h and CaMKII expression was determined by Western blotting.Representative blot is shown from three independent experiments with identicalresults. Blots were reprobed for β-actin to establish equivalent loading. (c) IGF-1induced HIF-1α activity is dependent on CaMKII activation. Cells transfected with HIF-1α reporter construct were treated with IGF-1 in the presence or absence of 10 μM ofCaMKII inhibitor KN93. After 24 h luciferase reporter assay was performed to determineHIF-1α activity. The graph represents fold change in activity over control. Valuesrepresent the means±SEM from 3 independent experiments. * Significant increasefrom control, # Significant decrease from IGF-1 treated cells (Pb0.05).

activity in glioma cells. IGF-1 elevated Ras activity in glioma cells(Fig. 1c). We have demonstrated the existence of an IL-1β-Ras- HIF-1αaxis in GBM [13]. To investigate whether elevated Ras plays a rolein IGF-1 induced HIF-1α activation, we determined HIF-1α transcrip-tional activity in cells transfected with DN-RasN17 in the presenceand absence of IGF-1. Increase in HIF-1α activation induced by IGF-1was abrogated in cells transfected with DN-RasN17 (Fig. 1d). Thissuggested the involvement of IGF-1 induced Ras in HIF-1α activation.

3.3. IGF-1 induced CaMKII regulates HIF-1α activity independent of Ras

CaMKII regulates HIF-1α transcriptional activity [19] and plays arole in glioma cell migration [17]. As IGF-1 regulates calcium channelsthrough CaMKII signaling [36], we investigated whether CaMKIIregulates HIF-1α transcriptional activity in IGF-1 treated cells. Anincrease in CaMKII phosphorylation was observed upon IGF-1treatment (Fig. 2a). As relation between Ras and CaMKII has beenreported [18] we investigated whether Ras contributes to increasedCaMKII expression in IGF-1 treated cells. The increase in CaMKII is Rasindependent as its expression was not affected in cells transfectedwith DN-Ras (N17) (Fig. 2b). Importantly, IGF-1 induced HIF-1αtranscriptional activity was abrogated in cells treated with CaMKIIinhibitor KN93 (Fig. 2c). Thus, IGF-1 induced Ras and CaMKIIindependently regulate HIF-1α activation.

Fig. 3. Existence of TLR9-CaMKII axis in IGF-1 treated glioma cells. (a) IGF-1 decreasesTLR9 expression in glioma cells. Western blot demonstrates TLR9 expression in gliomacells treated with 20 ng/ml of IGF-1 for 24 h. (b) CaMKII has no effect on TLR9expression in IGF-1 treated glioma cells. Cells were treated with IGF-1 in the presenceand absence of CaMKII inhibitor KN93 andWestern blot was performed to demonstrateTLR9 levels. (c) TLR9 agonist regulates CaMKII expression in glioma cells. Cells weretreated with IGF-1 in the presence and absence of TLR9 agonist CpG ODN 2006(5 μg/ml) and Western blot was performed to demonstrate CaMKII levels. Panels (a, b,c) are representative blots from at least three independent experiments with similarresults. Blots were reprobed for β-actin to establish equivalent loading.

1872 S. Sinha et al. / Cellular Signalling 23 (2011) 1869–1875

3.4. Decreased TLR9 levels in IGF-1 treated cells regulates CaMKII

As TLR9 signaling activates CaMKII [37] and TLR9 is expressed inGBM [33], we determined TLR9 levels in IGF-1 treated cells withelevated CaMKII levels. A decrease in TLR9 expressionwas observed inIGF-1 treated cells (Fig. 3a). As inhibition of CaMKII significantlysuppresses TLR9 induced inflammatory responses [37], we nextdetermined the co-relation between CaMKII and TLR9 in IGF-1 treatedcells. Inhibition of CaMKII has no effect on TLR9 expression, as it levelin control and IGF-1 treated cells were comparable (Fig. 3b).Treatment with TLR9 agonist inhibited IGF-1 mediated increase inCaMKII expression (Fig. 3c).

3.5. Existence of negative and positive TLR9-HIF-1α feed-back loop inglioma cells

As decrease in TLR9 expression was concurrent with increasedHIF-1α activation, we investigated whether TLR9 activation couldaffect HIF-1α in IGF-1 treated cells. IGF-1 induced HIF-1α wasabrogated to control levels in the presence of TLR9 agonist (Fig. 4a).Treatment with TLR9 agonist alone also resulted in a significantdecrease in HIF-1α activation (Fig. 4a). As siRNA mediated knock-down of HIF-1α inhibits hypoxia-induced TLR4 expression inmacrophages [27], we next investigated the role of HIF-1α in TLR9induction. Contrary to our expectation, knockdown of HIF-1αabrogated TLR9 level in both control and IGF-1 treated cells

Fig. 4. Complex cross-talk between HIF-1α and TLR9 in glioma cells. (a) TLR9 activationdecreases IGF-1 induced HIF-1α activation in glioma cells. Cells transfected with HIF-1αreporter construct were treated with IGF-1 in the presence or absence of 5 μg/ml ofTLR9 agonist CpG ODN 2006. After 24 h luciferase reporter assay was performed todetermine HIF-1α activity. The graph represents fold change in activity over control.Values represent the means±SEM from 3 independent experiments. * Significantincrease from control, # Significant decrease from IGF-1 treated cells (Pb0.05). (b)siRNA mediated knock-down of HIF-1α reduces TLR9 levels in both control and IGF-1treated cells. Mock and HIF-1α siRNA transfected cells were treated with IGF-1 for 24 hand TLR9 expression was determined by Western blotting. The figure is representativeblot from at least three independent experiments with similar results. Blots werereprobed for β-actin to establish equivalent loading. Inset shows HIF-1α levels in cellstransfected with HIF-1α siRNA.

(Fig. 4b). This finding highlights the complexity of TLR9 signaling inHIF-1α regulation and vice versa, as both negative and positive TLR9-HIF-1α feedback loops exists in glioma cells.

3.6. IGF-1 mediated decrease in CXCR4 is HIF-1α and TLR9 dependent

HIF-1α regulates CXCR4 expression in GBM [38] and CXCR4 isimportant for TLR9 agonist to enhance metastasis [39]. We thereforedetermined CXCR4 expression in IGF-1 treated cells with elevatedHIF-1α and decreased TLR9 levels. A decrease in CXCR4 expressionwas observed upon IGF-1 treatment (Fig. 5a). As hypoxia has beenshown to increase CXCR4 in glioma cells [38], we determined thewhether HIF-1α regulates CXCR4 expression in these cells. Knock-down of HIF-1α elevated IGF-1 mediated decrease in CXCR4 levels(Fig. 5b). Since decrease in CXCR4 was concurrent with TLR9expression, correlation between the two was also investigated. Thedecrease in CXCR4 levels observed upon IGF-1 treatmentwas revertedto levels comparable to control in the presence of TLR9 agonist(Fig. 5c). Thus, HIF-1α inhibition and TLR9 activation prevents IGF-1mediated decrease in CXCR4 levels.

3.7. Increase in SOCS3 expression in IGF-1 treated cells is HIF-1α andTLR9 dependent

Functional inactivation of CXCR4 mediated response involvesupregulation of SOCS3 [32]. As CXCR4 levels were down-regulated inIGF-1 treated cells, we determined SOCS3 expression in these cells.Treatment with IGF-1 elevated SOCS-3 expression (Fig. 6a). Thoughno direct link between SOCS3 and HIF-1α is known, SOCS3 is inducedby hypoxia in pulmonary arterial smooth muscle cell [40]. Wetherefore investigated the relationship between SOCS3 and HIF-1α bydetermining SOCS3 expression in glioma cells transfected with HIF-

Fig. 5. IGF-1mediated decrease in CXCR4 levels is HIF-1α and TLR9 dependent. (a) IGF-1decrease CXCR4 levels in glioma cells. Western blot demonstrates decrease in CXCR4levels in glioma cells treated with IGF-1 for 24 h. (b) siRNA mediated knock-down ofHIF-1α reverses IGF-1 mediated decrease in CXCR4 expression. Cells transfected withHIF-1α siRNA were treated with IGF-1 for 24 h and CXCR4 expression was determinedby Western blotting. (c) TLR9 activation increases IGF-1 induced decrease in CXCR4expression in glioma cells. Cells were treated with IGF-1 in the presence and absence ofTLR9 agonist for 24 h, andWestern blot was performed to determine CXCR4 expression.The panels (a–c) are representative of three independent experiments. Blots werereprobed with β-actin to establish equivalent loading.

Fig. 6. IGF-1 increases expression of SOCS3 and pro-inflammatory cytokines. (a) IGF-1 increases SOCS3 expression in glioma cells. Cells were treated with IGF-1 for 24 h, andWesternblot was performed to determine SOCS3 expression. (b) IGF-1 increases SOCS3 expression in a HIF-1α dependent manner. siRNA mediated knock-down of HIF-1α attenuates IGF-1induced increase in SOCS3 expression. Mock and HIF-1α siRNA transfected cells were treated with IGF-1 for 24 h and SOCS3 expression was determined by Western blotting. (c)TLR9 activation abrogates IGF-1 induced increase in SOCS3 expression in glioma cells. Cells were treated with IGF-1 in the presence and absence of TLR9 agonist for 24 h, andWesternblot was performed to determine SOCS3 expression. Panels (a, b and c) are representative blots from at least three independent experiments with similar results. Blots were probedwith β-actin to establish equivalent loading. (d) TLR9 activation induces expression of pro-inflammatory cytokines in glioma cells both in the presence and absence of IGF-1. Cellswere treated with IGF-1 in the presence and absence of TLR9 agonist for 24 h, and CBA was performed to determine levels of pro-inflammatory cytokines. Values represent themeans±SEM from 3 individual experiments. * Significant change from control Pb0.05. (e) Proposed model for IGF-1 triggered HIF-1α dependent regulation of inflammatoryresponse in glioma cells.

1873S. Sinha et al. / Cellular Signalling 23 (2011) 1869–1875

1α siRNA. Interestingly, siRNA mediated knock-down of HIF-1αabrogated IGF-1 induced increase in SOCS3 expression (Fig. 6b).Moreover, treatment with TLR9 agonist decreased IGF-1 inducedincrease in SOCS3 expression (Fig. 6c).

3.8. IGF-1 regulates pro-inflammatory response in glioma cells

Sincewehave reported that an IL-1β-HIF-1α autocrine loop sustainsinflammatory response in glioma cells [13], and SOCS3 is known toprotect pancreatic β-cells from IL-1β mediated cytotoxicity [41], weinvestigated the status of IL-1β in IGF-1 treated cells with elevated HIF-1α and SOCS3 expression. A significant increase in IL-1β levels wasobserved upon IGF-1 treatment (Fig. 6d). Treatment with TLR9 agonisteither alone or in presence of IGF-1 elevated IL-1β to levels similar tothat observed upon IGF-1 treatment. Presence of TLR9 agonist had noeffect on the ability of IGF-1 to maintain elevated IL-1β levels (Fig. 6d).Similar trend in the expression of other pro-inflammatory cytokinessuch as IL-6 and IL-8 was observed in glioma cells treated with IGF-1 inthe presence and absence of TLR9 agonist (Fig. 6d). Taken together our

findings indicate that a complex cross-talk between TLR9 and HIF-1αregulates inflammatory responses in IGF-1 treated glioma cells (Fig.6e).

4. Discussion

IGF signaling plays an important role in malignant transformationand protection from apoptosis in a broad range of human cancer[3,42]. Glioma produces IGFs and express elevated levels of IGFreceptors compared with normal brain tissue [43,44]. Besides,inhibition of IGF receptor disrupts a pro-survival and pro-angiogenicIGF-HIF pathway in glioblastoma cells [45]. Our finding suggests thatIGF-1 induced Ras and CaMKII act independently to elevate HIF-1αactivation in glioma cells.

IGF-1 induced increase in HIF-1α activation is concomitant withdecreased TLR9 and CXCR4 levels and elevated SOCS3 expression. Themost important finding of this studywas the identification of a complexcrosstalk between TLR9 and HIF-1α in response to IGF-1, undernormoxia. TLR9 and HIF-1α seem to regulate each other through twoopposite feedback loops.While knockdownofHIF-1αdecreases TLR9bybehaving asmutual regulator of each other in a positive regulatory loop,

1874 S. Sinha et al. / Cellular Signalling 23 (2011) 1869–1875

activation of TLR9 signaling abrogates HIF-1α activation through anegative regulatory loop. This finding highlights the complexity of TLR9signaling in HIF-1α activation and vice versa in glioma cell, as bothnegative and positive TLR9-HIF-1α feedback loops act in tandem toregulate inflammatory response. It is possible that thenegative feedbackoccurs beneath a given threshold of the two proteins and that thepositive loop starts when this threshold is crossed.

As TLR9 signaling regulates both CaMKII and HIF-1α in IGF-1treated cells and since CaMKII triggered by TLR ligands promoteinflammatory responses [37], it is likely that TLR9 act as a sensor tomaintain the inflammatory millieu through regulation of CaMKII andHIF-1α. We have reported that HIF-1α maintains persistently highlevel of IL-1β through an HIF-1α-IL1β autocrine loop in glioma cells[13]. Pro-inflammatory cytokines in the tumor microenvironmentplays a major factor in tumorigenesis [46]. The simultaneous increasein SOCS3 and decrease in TLR9 was concurrent with heightened levelof IL-1β and pro-inflammatory cytokines. It is known that TLR9promotes inflammatory response [37] and SOCS3 prevents IL-1βmediated cytotoxicity [41]. As TLR9 activation decreases IGF-1mediated increase in SOCS3 expression, it is tempting to speculatethat low TLR9 levels maintains elevated SOCS3 expression in IGF-1treated cells to prevent pro-inflammatory cytokines from crossing thethreshold beyond levels required to sustain a TLR9-HIF-1α axis. It ispossible that negative TLR9-HIF-1α axis is initiated to maintainelevated HIF-1α levels in presence of IGF-1. Another importantfinding of this study is the elucidation of the involvement of HIF-1α inthe regulation of SOCS3.

While CXCR4 modulates TLR9 mediated signaling [29], TLR9increasesmetastatic potential of cancer cells through CXCR4 expression[28]. Although TLR9 agonist has no contribution towards sustaining theelevated pro-inflammatory cytokines levels triggered by IGF-1, treat-ment with TLR9 agonist alone elevates pro-inflammatory cytokines tolevels similar to IGF-1 treated cells. TLR9 agonists stimulate innate andadaptive anti-tumor immune responses and have demonstratedpotential for the treatment of cancer [47,48]. This along with the abilityof TLR9 agonist to elevate CXCR4 levelswarrants investigation regardingthe use of TLR9 agonist as an effective anti-glioma target. Thoughpharmacologic inhibition of HIF-1α or the SDF-1/CXCR4 interactionabrogates regrowth of GBM [49], inhibition of HIF-1α reversed IGF-1mediated decrease in CXCR4 expression. These findings suggested thatHIF-1α regulates CXCR4 in different ways depending on the context ofmicroenvironment.

5. Conclusion

Our findings have not only highlighted (i) a previously unrecog-nized function of HIF-1α as an important regulator of SOCS3 and TLR9expression, but has also (ii) established HIF-1α as a link between twoapparently unrelated but crucial component of glioma tumormicroenvironment- growth factor and inflammation. Taken together,our data underline the complexity of HIF-1α in its crosstalk with TLR9under normoxia. This study prompts further investigation intomechanisms governing the dialog between HIF-1α and TLR9 to revealnew molecular components that participates in regulating inflamma-tory responses in glioma.

Conflict of interest

There are no competing financial interests in relation to the workand we have nothing to disclose.

Acknowledgments

The work was supported by a research grant from the Department ofBiotechnology (DBT)Government of India (BT/PR12924/Med/30/235/09)

and partly by the Innovative Young Biotechnologist Award (IYBA) by DBTto ES.

References

[1] H.N. Antoniades, T. Galanopoulos, J. Neville-Golden, M. Maxwell, Int. J. Cancer50 (2) (1992) 215–222.

[2] D. LeRoith, C.T. Roberts Jr., Cancer Lett. 195 (2) (2003) 127–137.[3] M.N. Pollak, E.S. Schernhammer, S.E. Hankinson, Nat. Rev. Cancer 4 (7) (2004)

505–518.[4] N.V. Iyer, L.E. Kotch, F. Agani, S.W. Leung, E. Laughner, R.H. Wenger, M. Gassmann,

J.D. Gearhart, A.M. Lawler, A.Y. Yu, G.L. Semenza, Genes Dev. 12 (2) (1998)149–162.

[5] R. Fukuda, K. Hirota, F. Fan, Y.D. Jung, L.M. Ellis, G.L. Semenza, J. Biol. Chem.277 (41) (2002) 38205–38211.

[6] K.M. Sutton, S. Hayat, N.M. Chau, S. Cook, J. Pouyssegur, A. Ahmed, N. Perusinghe,R. Le Floch, J. Yang, M. Ashcroft, Oncogene 26 (27) (2007) 3920–3929.

[7] P.H. Maxwell, G.U. Dachs, J.M. Gleadle, L.G. Nicholls, A.L. Harris, I.J. Stratford, O.Hankinson, C.W. Pugh, P.J. Ratcliffe, Proc. Natl. Acad. Sci. U S A 94 (15) (1997)8104–8109.

[8] G.L. Semenza, Trends Mol. Med. 8 (4 Suppl.) (2002) S62–S67.[9] S.B. Catrina, I.R. Botusan, A. Rantanen, A.I. Catrina, P. Pyakurel, O. Savu, M. Axelson,

P. Biberfeld, L. Poellinger, K. Brismar, Clin. Cancer Res. 12 (15) (2006) 4506–4514.[10] K. Beppu, K. Nakamura, W.M. Linehan, A. Rapisarda, C.J. Thiele, Cancer Res. 65 (11)

(2005) 4775–4781.[11] W. Zundel, C. Schindler, D. Haas-Kogan, A. Koong, F. Kaper, E. Chen, A.R.

Gottschalk, H.E. Ryan, R.S. Johnson, A.B. Jefferson, D. Stokoe, A.J. Giaccia, GenesDev. 14 (4) (2000) 391–396.

[12] F. Rininsland, T.R. Johnson, C.L. Chernicky, E. Schulze, P. Burfeind, J. Ilan, Proc. Natl.Acad. Sci. U S A 94 (11) (1997) 5854–5859.

[13] V. Sharma, D. Dixit, N. Koul, V.S. Mehta, Sen. E. J. Mol. Med. 89 (2) (2011) 123–136.[14] M. Stearns, J. Tran, M.K. Francis, H. Zhang, C. Sell, Cancer Res. 65 (6) (2005)

2085–2088.[15] A. Guha, N. Lau, I. Huvar, D. Gutmann, J. Provias, T. Pawson, G. Boss, Oncogene

12 (3) (1996) 507–513.[16] R. Blum, J. Jacob-Hirsch, N. Amariglio, G. Rechavi, Y. Kloog, Cancer Res. 65 (3)

(2005) 999–1006.[17] V.A. Cuddapah, H. Sontheimer, J. Biol. Chem. 285 (15) (2010) 11188–11196.[18] M. Illario, A.L. Cavallo, K.U. Bayer, T. Di Matola, G. Fenzi, G. Rossi, M. Vitale, J. Biol.

Chem. 278 (46) (2003) 45101–45108.[19] G. Yuan, J. Nanduri, C.R. Bhasker, G.L. Semenza, N.R. Prabhakar, J. Biol. Chem.

280 (6) (2005) 4321–4328.[20] J. Westra, E. Brouwer, I.A. van Roosmalen, B. Doornbos-van der Meer, M.A. van

Leeuwen,M.D. Posthumus, C.G. Kallenberg, BMCMusculoskeletDisord11 (2010) 61.[21] S. Kaluz, E.G. Van Meir, J. Mol. Med. 89 (2) (2011) 91–94.[22] Y.J. Jung, J.S. Isaacs, S. Lee, J. Trepel, L. Neckers, FASEB J. 17 (14) (2003) 2115–2117.[23] T. Cramer, Y. Yamanishi, B.E. Clausen, I. Forster, R. Pawlinski, N. Mackman, V.H.

Haase, R. Jaenisch, M. Corr, V. Nizet, G.S. Firestein, H.P. Gerber, N. Ferrara, R.S.Johnson, Cell 112 (5) (2003) 645–657.

[24] S. Frede, C. Stockmann, P. Freitag, J. Fandrey, Biochem. J. 396 (3) (2006) 517–527.[25] S. Frede, C. Stockmann, S. Winning, P. Freitag, J. Fandrey, J. Immunol. 182 (10)

(2009) 6470–6476.[26] S. Akira, K. Takeda, Nat. Rev. Immunol. 4 (7) (2004) 499–511.[27] S.Y. Kim, Y.J. Choi, S.M. Joung, B.H. Lee, Y.S. Jung, J.Y. Lee, Immunology 129 (4)

(2010) 516–524.[28] T. Ren, Z.K.Wen, Z.M. Liu, Y.J. Liang, Z.L. Guo, L. Xu, Cancer Biol. Ther. 6 (11) (2007)

1704–1709.[29] M. Ishii, C.M. Hogaboam, A. Joshi, T. Ito, D.J. Fong, S.L. Kunkel, Eur. J. Immunol.

38 (8) (2008) 2290–2302.[30] F.Y. Liew, D. Xu, E.K. Brint, L.A. O'Neill, Nat. Rev. Immunol. 5 (6) (2005) 446–458.[31] H. Zhou, R. Miki, M. Eeva, F.M. Fike, D. Seligson, L. Yang, A. Yoshimura, M.A. Teitell,

C.A. Jamieson, N.A. Cacalano, Clin. Cancer Res. 13 (8) (2007) 2344–2353.[32] S.F. Soriano, P. Hernanz-Falcon, J.M. Rodriguez-Frade, A.M. De Ana, R. Garzon, C.

Carvalho-Pinto, A.J. Vila-Coro, A. Zaballos, D. Balomenos, A.C. Martinez, M.Mellado, J. Exp. Med. 196 (3) (2002) 311–321.

[33] Y. Meng, M. Kujas, Y. Marie, S. Paris, J. Thillet, J.Y. Delattre, A.F. Carpentier, J.Neurooncol. 88 (1) (2008) 19–25.

[34] S. Biswas, M.K. Gupta, D. Chattopadhyay, C.K. Mukhopadhyay, Am. J. Physiol. HeartCirc. Physiol. 292 (2) (2007) H758–766.

[35] V. Sharma, C. Joseph, S. Ghosh, A. Agarwal, M.K. Mishra, E. Sen, Mol. Cancer Ther.6 (9) (2007) 2544–2553.

[36] L. Gao, L.A. Blair, G.D. Salinas, L.A. Needleman, J. Marshall, J. Neurosci. 26 (23)(2006) 6259–6268.

[37] X. Liu, M. Yao, N. Li, C. Wang, Y. Zheng, X. Cao, Blood 112 (13) (2008) 4961–4970.[38] D. Zagzag, Y. Lukyanov, L. Lan, M.A. Ali, M. Esencay, O. Mendez, H. Yee, E.B. Voura,

E.W. Newcomb, Lab. Invest. 86 (12) (2006) 1221–1232.[39] L. Xu, Y. Zhou, Q. Liu, J.M. Luo, M. Qing, X.Y. Tang, X.S. Yao, C.H. Wang, Z.K. Wen,

Biochem. Biophys. Res. Commun. 382 (3) (2009) 571–576.[40] L. Bai, Z. Yu, G. Qian, P. Qian, J. Jiang, G. Wang, C. Bai, Respir. Physiol. Neurobiol.

152 (1) (2006) 83–91.[41] A.E. Karlsen, S.G. Ronn, K. Lindberg, J. Johannesen, E.D. Galsgaard, F. Pociot, J.H.

Nielsen, T. Mandrup-Poulsen, J. Nerup, N. Billestrup, Proc. Natl. Acad. Sci. U S A98 (21) (2001) 12191–12196.

[42] E. Foulstone, S. Prince, O. Zaccheo, J.L. Burns, J. Harper, C. Jacobs, D. Church, A.B.Hassan, J. Pathol. 205 (2) (2005) 145–153.

1875S. Sinha et al. / Cellular Signalling 23 (2011) 1869–1875

[43] R.P. Glick, T.G. Unterman, M. Van der Woude, L.Z. Blaydes, J. Neurosurg. 77 (3)(1992) 445–450.

[44] A.C. Sandberg-Nordqvist, P.A. Stahlbom, M. Reinecke, V.P. Collins, H. von Holst, V.Sara, Cancer Res. 53 (11) (1993) 2475–2478.

[45] M.B. Gariboldi, R. Ravizza, E. Monti, Biochem. Pharmacol. 80 (4) (2010) 455–462.

[46] L.M. Coussens, Z. Werb, Nature 420 (6917) (2002) 860–867.[47] A.M. Krieg, J. Clin. Invest. 117 (5) (2007) 1184–1194.[48] A.M. Krieg, Oncogene 27 (2) (2008) 161–167.[49] M. Kioi, H. Vogel, G. Schultz, R.M. Hoffman, G.R. Harsh, J.M. Brown, J. Clin. Invest.

120 (3) (2010) 694–705.