Neovascularization is a limiting factor in tumor growth and progression. It is well known that changes in the tumor microenvironment, such as hypoxia and glucose deprivation (GD), can induce VEGF production. However, the mechanism linking GD to tumor growth and angiogenesis is unclear. We hypothesize that GD induces the angiogenic switch in tumors through activation of the unfolded protein response (UPR). We report that UPR activation in human tumors results in elevated expression of proangiogenic mediators and a concomitant decrease in angiogenesis inhibitors. cDNA microarray results showed that GD-induced UPR activation promoted upregulation of a number of proangiogenic mediators (VEGF, FGF-2, IL-6, etc.) and downregulation of several angiogenic inhibitors (THBS1, CXCL14, and CXCL10). In vitro studies revealed that partially blocking UPR signaling by silencing protein kinase RNA–like ER kinase (PERK) or activating transcription factor 4 (ATF4) significantly reduced the production of angiogenesis mediators induced by GD. However, suppressing the alpha subunit of hypoxia-inducible factors had no effect on this process. Chromatin immunoprecipitation (ChIP) confirmed binding of ATF4 to a regulatory site in the VEGF gene. In vivo results confirmed that knockdown of PERK in tumor cells slows down tumor growth and decreases tumor blood vessel density. Collectively, these results show that the PERK/ATF4 arm of UPR mediates the angiogenic switch and is a potential target for antiangiogenic cancer therapy. Cancer Res; 72(20); 5396–406. ©2012 AACR.

Dividing tumors can rapidly outgrow their blood supply. This results in a toxic tumor microenvironment (TME) characterized with hypoxia, acidic pH, glucose deprivation (GD), and amino acid deficiency. Increasing evidence suggests that the TME contains stressors that promote accumulation of misfolded proteins in the lumen of the endoplasmic reticulum (ER). This in turn activates intracellular signaling pathways termed as the unfolded protein response (UPR; refs. 1–3). Initially, the UPR is cytoprotective aimed at restoring normal ER function (4). However, in the presence of severe or prolonged ER stress, cell death programs are activated (3, 4).

Mammalian UPR is controlled by 3 ER-resident transmembrane proteins that serve as proximal sensors of ER stress and activate downstream signaling effectors, such as protein kinase RNA–like ER kinase (PERK), inositol requiring 1 (IRE1), and activating transcription factor 6 (ATF6; ref. 5). These effectors are maintained in an inactive state through association with the molecular chaperone, glucose-regulated protein 78 kDa (Grp78; ref. 2). Upon ER stress Grp78 dissociates from these sensors, which activates UPR signaling (6). After dimerization and transautophosphorylation, activated PERK relays signal by phosphorylating the alpha subunit of eukaryotic initiation factor-2 (eIF-2α), which in turn inhibits general translation initiation and selectively translates several mRNAs including ATF4 (7). ATF4 transactivates expression of several genes, such as C/EBP homologous protein (CHOP), a transcription factor implicated in apoptosis (8, 9), and Grp78 (10). Upon UPR activation, IRE1 splices an unconventional intron from the X-box-binding protein 1 (XBP1) mRNA, producing an active transcription factor XBP1-s (11, 12), which translocates into the nucleus and modulates the expression of several proteins involved in folding or clearance of aberrant proteins (2, 13). Finally, during ER stress, ATF6 is processed into an active transcription factor, moves into the nucleus, and upregulates ER chaperones and folding enzymes (2, 14).

Studies have shown that GD induces the expression of VEGF in different tumor cell lines (15–17), suggesting that besides hypoxia (18, 19), low concentration of nutrients play a role in triggering angiogenesis (20). It is also known that the pathologic stimulus GD causes ER stress and alters gene expression through UPR signaling (20). However, mechanistic studies of UPR-mediated angiogenesis have been conducted for the most part using nonphysiologic agents, such as thapsigargin (TG), focusing mainly on VEGF expression (21, 22). The mechanism underlying GD-induced UPR in tumor angiogenesis has not been fully elucidated.

In this study, we report that activation of the UPR by GD plays a pivotal role in tumor angiogenesis by activating the angiogenic switch and that the PERK/ATF4 pathway of the UPR is involved in this process. These results suggest that targeting the proangiogenic arm of the UPR may reveal new strategies for cancer treatment.

Cells

The head and neck squamous cell carcinoma (HNSCC) cell line UM-SCC-81B (from Dr. Thomas E. Carey, Departments of Otorhinolaryngology and Pharmacology, University of Michigan Medical School, Ann Arbor, MI), the breast cancer cell line MCF7 (American Type Culture Collection), the glioma cell line U87 (from Dr. Yi Sun, Department of Radiation Oncology, University of Michigan, Ann Arbor, MI), and mouse embryonic fibroblast (MEF) cell lines (MEF–PERK+/+ and MEF–PERK−/−, from Dr. Andrew Fribley, Department of Pediatrics, Division of Hematology/Oncology, Wayne State University, Detroit, MI) were maintained in Dulbecco's Modified Eagle's Medium (DMEM) with high glucose (Invitrogen) with 10% FBS. All tumor cell lines were authenticated recently by DNA fingerprinting with short tandem repeat profiling. Primary human dermal microvascular endothelial cells (HDMEC; Cambrex) were cultured in endothelial cell growth medium-2 (EGM2; Cambrex). TG and tunicamycin (TM) were purchased from Sigma.

Immunofluorescence and immunohistochemical analysis

Immunofluorescence and immunohistochemistry (IHC) were conducted as described previously (23) with primary antibodies against Grp78, CHOP (Santa Cruz), Ki67, and CD31 (BD Pharmingen). Goat antirabbit Alexa Flour 594 (Molecular Probes) and 4′,6-diamidino-2-phenylindole (DAPI) counterstaining were used for immunofluorescence staining. The Polink-2 horseradish peroxidase broad kit with 3,3′-diaminobenzidine (DAB) chromogen was used for IHC staining. Normal human oral mucosa (NHM, from Dr. Hector Rios, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI) was used as control. All imaging was done using a Leica DM5000 microscope.

Laser capture microdissection

Laser capture microdissection (LCM) was conducted as previously described (24). Approximately 500,000 epithelial cells from either HNSCC or NHM were collected using a pulsed 337-nm UV laser. RNA from at least 15 independent tumors and 10 NHM tissues were pooled respectively and analyzed using real-time PCR (qPCR).

Microarray analysis

Total RNA was isolated from UM-SCC-81B cells treated with or without GD (glucose, 0.1 mmol/L) using the RNeasy Plus Mini Kit (Qiagen). A Human gene chip U133 Plus 2.0 (Affymetrix) was used to analyze gene expression. Details and results can be accessed in Gene Expression Omnibus (GEO) repository (GSE38583).

Immunoblotting

Whole-cell lysates were resolved by SDS–PAGE and transferred onto polyvinylidene difluoride membrane (Pierce) and probed with the antibodies: Grp78, CHOP, ATF4, β-actin (Santa Cruz), HIF1α (BD Pharmingen), PERK (Cell Signaling), and spliced XBP1 (Biolegend). Horse radish peroxidase-conjugated secondary antibodies were from Santa Cruz. SuperSignal West Pico Chemiluminescent Substrate (Pierce) was used to visualize immunoreactive bands.

ELISA

ELISA was conducted according to the manufacturer's instructions. Briefly, cell culture supernatants were diluted 1:10, applied to each well (100 μL), incubated at room temperature for 2 hours, and washed 3 times. The secondary antibody reaction was conducted at room temperature (1 hour). Stabilized chromogen was used for colorimetric reactions. Optical density was measured at 450 nm using a plate reader (Spectra Max M2).

Lentivirus infection

GFP-expressing lentiviral constructs expressing short hairpin RNA (shRNA) against PERK, ATF4, and HIF1α were from Open Biosystems. For infection, 1 × 105 cells were plated in 6-cm plates, infected with the lentivirus and sorted with flow cytometry to ensure 100% positivity. Established stable cell lines were cultured with 2 μg/mL puromycin.

Chromatin immunoprecipitation

Chromatin immunoprecipitation (ChIP) analysis was conducted using an Agarose-Chip Kit (Pierce) according to the manufacturer's instruction. Cells were treated for 18 hours with complete DMEM containing normal glucose (25 mmol/L) or low glucose (2 mmol/L), respectively. The chromatin solution was incubated overnight with ATF4 antibody (sc-200, Santa Cruz), nonimmune rabbit IgG, and anti-RNA polymerase II antibody. Purified complexes and input DNAs were analyzed by PCR. The ATF4-binding sites of VEGF gene (AsnSyn site) and asparagine synthetase gene (NSRE-1, nutrient-sensing response element, used as a positive control) have been described previously (25, 26). Amplicons were resolved on 1% agarose gels and visualized by SYBR Safe DNA gel stain (Invitrogen).

Real-time PCR and conventional reverse transcription PCR

Total mRNA was extracted from cultured cells with RNeasy plus minikit (Qiagen) following manufacturer's instructions. cDNA was synthesized using the Verso cDNA Kit (Thermo Scientific). Real-time PCR was conducted in 384-well plate with the ABI PRISM 7900HT Sequence Detection System. Primers used for qPCR (Grp78, CHOP, 18S, ATF4, VEGF, IL-6, FGF-2, CXCL10, and CXCL14) were from Applied Biosystems. RT-PCR was also used to check gene expression (IL-6, VEGFA, FGF-2, CTGF, Grp78, and 18s; details in Supplementary Data).

Sprout formation assay

HDMECs (1.5 × 105 cells) were seeded in 6-well plates containing 1.5 mL layer of gelled type I collagen (Cohesion) for 24 hours, then cultured in EBM2 (2% FBS) containing concentrated GDCM (conditioned medium collected from cells cultured with GD) or NGCM (conditioned medium collected from cells cultured with regular medium) for 3 days. Number of sprouts in 12 random microscope fields per well was counted on day 3 in triplicate per condition. Images were taken with a Leica DMI 3000B.

Tumor growth and angiogenesis in vivo

Tumor cells (UM-SCC-81B-scshRNA and UM-SCC-81B-shPERK, 5 × 105) were injected subcutaneously in the flanks of severe combined immunodeficient mice (SCID) mice (Harlan Laboratories). Tumor volume was measured every 3 days from day 17 postinjection with a digital caliper. Tumor volumes were calculated using the formula volume (mm3) = length × width2/2. At the endpoint, tumors were surgically removed and analyzed for tumor angiogenesis.

Statistical analysis

Data are expressed as mean ± SD and analyzed using unpaired 2-tailed Student t test. A value of P < 0.05 was considered to be significant.

UPR activation in human tumors coincides with upregulation of proangiogenic mediators and downregulation of angiogenic inhibitors

To investigate the role of UPR in HNSCC, we examined expression of UPR markers Grp78 and CHOP in patients with HNSCC (10 patients) and NHM (5 controls). Strong expression of both Grp78 (85%) and CHOP (88%) was detected in HNSCC as compared with NHM (11% and 30%, respectively; Fig. 1A–C and Supplementary Fig. S1), suggesting that the UPR was activated in tumor samples. With LCM, epithelial cells from both HNSCC (15 patients) and NHM (10 controls; Fig. 1D) were collected, and qPCR was carried out to determine relative expression levels of target genes. Significant Grp78 increase was observed in HNSCC (Fig. 1D), confirming activation of the UPR in cancer cells. Simultaneously, mRNA levels of IL-6 and VEGF showed a 2.9- and 3.5-fold increase, respectively, and the expression level of the antiangiogenic chemokine CXCL14 showed significant decrease (Fig. 1D). These results suggest that in human tumor samples, UPR is associated with a shift in the balance between pro- and antiangiogenic mediators in favor of angiogenesis.

Figure 1.

UPR activation in human tumor tissues coincides with upregulation of proangiogenic factors and downregulation of antiangiogenic factors. A, immunofluorescence staining of Grp78. B, IHC staining of CHOP. C, quantification showing percentage of Grp78 and CHOP positive cells. D, epithelial cells were collected using LCM from NHM (10 samples, pooled) and HNSCC (15 samples, pooled). qPCR was used to analyze the expression of Grp78 and angiogenic mediators. Gene expression levels in tumor tissues were normalized to their expression in normal mucosa (defined as 1). All the pictures were taken at ×100 magnification. Scale bar, 50 μm; *, P < 0.05.

Figure 1.

UPR activation in human tumor tissues coincides with upregulation of proangiogenic factors and downregulation of antiangiogenic factors. A, immunofluorescence staining of Grp78. B, IHC staining of CHOP. C, quantification showing percentage of Grp78 and CHOP positive cells. D, epithelial cells were collected using LCM from NHM (10 samples, pooled) and HNSCC (15 samples, pooled). qPCR was used to analyze the expression of Grp78 and angiogenic mediators. Gene expression levels in tumor tissues were normalized to their expression in normal mucosa (defined as 1). All the pictures were taken at ×100 magnification. Scale bar, 50 μm; *, P < 0.05.

Close modal

GD can effectively induce the UPR and regulate angiogenic mediator production

To explain results obtained from human tumors, we investigated the role of GD-induced UPR in modulating angiogenesis related gene expression in vitro. Upon GD treatment (2–25 mmol/L), UPR was activated as shown by phosphorylation of PERK (upward shift in the bands) and increased expression of ATF4, Grp78, and spliced XBP1 (Fig. 2A). The strength of UPR (shown by dose-dependent increase of Grp78) correlates with upregulation of VEGF at both protein and mRNA levels (Fig. 2A and Supplementary Fig. S2A and S2B). Furthermore, mRNA levels of FGF-2 and IL-6 displayed the same trend as VEGF in response to gradient glucose treatment (Supplementary Fig. S2B–S2D). Results of RT-PCR analysis also showed that transcription of the UPR marker Grp78 as well as the proangiogenic factors VEGF, IL-6, CTGF, and FGF-2 were increased with GD treatment (Fig. 2B). Increased secretion of IL-6 and FGF-2 in response to GD was also observed (Fig. 2C).

Figure 2.

GD promotes expression of proangiogenic mediators in tumor cells. A, UM-SCC-81B cells were treated with glucose gradient (2–25 mmol/L) for 18 hours. PERK, XBP1s, ATF4, and Grp78 were used as indicators of UPR activation. VEGF secretion was quantified with ELISA. B, expression of IL-6, VEGF, FGF-2, CTGF, and Grp78 in response to GD (0.55 mmol/L and 2 mmol/L) was assessed with RT-PCR, and 18s was used as an internal control. C, expression of UPR markers and angiogenic factors in UM-SCC-81B cells treated with GD (2 mmol/L, 24 hours) were determined using qPCR. Cytokine secretion was evaluated with ELISA. D, UM-SCC-81B cells were treated with GD (2 mmol/L), TM (1 μg/mL), and TG (1 μmol/L). CXCL10 expression was quantified with qPCR and normalized to control (percentage). CXCL10 secretion was quantified with ELISA. *, P < 0.05.

Figure 2.

GD promotes expression of proangiogenic mediators in tumor cells. A, UM-SCC-81B cells were treated with glucose gradient (2–25 mmol/L) for 18 hours. PERK, XBP1s, ATF4, and Grp78 were used as indicators of UPR activation. VEGF secretion was quantified with ELISA. B, expression of IL-6, VEGF, FGF-2, CTGF, and Grp78 in response to GD (0.55 mmol/L and 2 mmol/L) was assessed with RT-PCR, and 18s was used as an internal control. C, expression of UPR markers and angiogenic factors in UM-SCC-81B cells treated with GD (2 mmol/L, 24 hours) were determined using qPCR. Cytokine secretion was evaluated with ELISA. D, UM-SCC-81B cells were treated with GD (2 mmol/L), TM (1 μg/mL), and TG (1 μmol/L). CXCL10 expression was quantified with qPCR and normalized to control (percentage). CXCL10 secretion was quantified with ELISA. *, P < 0.05.

Close modal

Interestingly, the expression of CXCL10, an antiangiogenic factor was reduced with GD treatment at both mRNA and protein levels (supernatant; 5- and 2-fold, respectively; Fig. 2D). The other 2 chemical UPR inducers, TM and TG also potently suppressed its expression at mRNA levels (Fig. 2D).

To further understand how angiogenesis related genes respond to GD, UM-SCC-81B cells were subjected to GD for 4 hours and 24 hours, and genes of interest were analyzed by cDNA microarray. A panel of proangiogenic mediators, such as VEGF, IL-6, FGF-2, TGFB2, NRG1, and NGF, were upregulated and several antiangiogenic mediators, such as CXCL10, CXCL14, and THBS1 were inhibited (Table 1). Collectively, these data suggest that UPR modulates the angiogenic switch by adjusting the balance between pro- and antiangiogenic mediators in favor of angiogenesis.

Table 1.

Glucose deprivation increases angiogenesis mediators expression

Fold changea
Gene symbolGene titleAngiogenic effect4 h24 h
CTGF Connective tissue growth factor Positive 3.17 3.42 
CXCL10 Chemokine (C-X-C motif) ligand 10 Negative −7.8 
CXCL14 Chemokine (C-X-C motif) ligand 14 Negative −2.1 −28.2 
CXCL3 Chemokine (C-X-C motif) ligand 3 Positive 7.14 2.05 
FGF2 Fibroblast growth factor 2 (basic) Positive 2.6 
HYOU1 Hypoxia upregulated 1 Positive 6.34 
IL-6 Interleukin 6 (interferon, β-2) Positive 2.12 2.59 
IL-8 Interleukin 8 Positive 3.5 −3 
MANF Pmesencephalic astrocyte-derived neurotrophic factor Positive 2.71 6.34 
NGF Nerve growth factor (β-polypeptide) Positive 2.71 
NRG1 Neuregulin 1 Positive 2.3 
PDGFA Platelet-derived growth factor α polypeptide Positive 
TGFB2 Transforming growth factor, β-2 Positive 2.21 
THBS1 Thrombospondin 1 Negative −4.13 
VEGFA Vascular endothelial growth factor A Positive 3.71 4.04 
VEGFB Vascular endothelial growth factor B Positive 3.77 5.19 
Fold changea
Gene symbolGene titleAngiogenic effect4 h24 h
CTGF Connective tissue growth factor Positive 3.17 3.42 
CXCL10 Chemokine (C-X-C motif) ligand 10 Negative −7.8 
CXCL14 Chemokine (C-X-C motif) ligand 14 Negative −2.1 −28.2 
CXCL3 Chemokine (C-X-C motif) ligand 3 Positive 7.14 2.05 
FGF2 Fibroblast growth factor 2 (basic) Positive 2.6 
HYOU1 Hypoxia upregulated 1 Positive 6.34 
IL-6 Interleukin 6 (interferon, β-2) Positive 2.12 2.59 
IL-8 Interleukin 8 Positive 3.5 −3 
MANF Pmesencephalic astrocyte-derived neurotrophic factor Positive 2.71 6.34 
NGF Nerve growth factor (β-polypeptide) Positive 2.71 
NRG1 Neuregulin 1 Positive 2.3 
PDGFA Platelet-derived growth factor α polypeptide Positive 
TGFB2 Transforming growth factor, β-2 Positive 2.21 
THBS1 Thrombospondin 1 Negative −4.13 
VEGFA Vascular endothelial growth factor A Positive 3.71 4.04 
VEGFB Vascular endothelial growth factor B Positive 3.77 5.19 

NOTE: UM-SCC-81B cells were treated with 0.1 mmol/L glucose for 0, 4, and 24 hours. Total mRNA was extracted and subjected to cDNA array analysis. Positive, proangiogenic; negative, antiangiogenic; -, no change.

aP < 0.05.

Multiple ER stressors can induce the angiogenic phenotype in different tumor types

To investigate whether the tumor angiogenic response was not limited to GD only, UM-SCC-81B cells were treated with chemical ER stressors, TM (1 μg/mL) and TG (1 μmol/L). Like GD, TM and TG were able to activate the UPR (Fig. 3A) and significantly increase VEGF secretion (Fig. 3B).

Figure 3.

Tumor cells express proangiogenic mediators in response to different ER stressors. Tumor cell lines (UM-SCC-81B, MCF7, and U87) were treated with GD (2 mmol/L), TM (1 μg/mL), and TG (1 μmol/L). A, UPR markers were detected by Western blot analysis. B, VEGF secretion was quantified with ELISA. *, P < 0.05.

Figure 3.

Tumor cells express proangiogenic mediators in response to different ER stressors. Tumor cell lines (UM-SCC-81B, MCF7, and U87) were treated with GD (2 mmol/L), TM (1 μg/mL), and TG (1 μmol/L). A, UPR markers were detected by Western blot analysis. B, VEGF secretion was quantified with ELISA. *, P < 0.05.

Close modal

To eliminate the possibility of a cell-specific response, the breast cancer cell line MCF7 and the glioma cell line U87 were both subjected to GD (2 mmol/L), TG (1 μmol/L), and TM (1 μg/mL) treatment with subsequent assessment of UPR activation and VEGF secretion. We observed consistent UPR activation in both MCF7 and U87 (Fig. 3A) and increased expression of VEGF (Fig. 3B) with all 3 stressors. In addition, 2 other HNSCC cell lines, UM-SCC-11B and UM-SCC-17B showed upregulation of the proangiogenic mediators VEGF, FGF2, and IL-6 upon GD treatment (Supplementary Fig. S3A–S3D). Collectively, these data indicate that the UPR plays an important role in regulating production of angiogenic mediators in tumor cells regardless of their origin.

HIF1α activation is not required for GD-induced UPR-mediated production of proangiogenic mediators

Hypoxia promotes the production of multiple proangiogenic mediators, including VEGF, platelet-derived growth factor (27), IL-6 (28), FGF-2 (29), and placental growth factor (30). Stein and colleagues reported that hypoxia transcriptionally increases VEGF expression and stabilizes its mRNA through HIF1α (17), and HIF1α has been reported to be involved in GD-induced VEGF expression in mouse embryonic cells (31). To address whether HIF1α is involved in GD-induced VEGF expression through UPR activation in tumor cells used, we knocked down the expression of HIF1α with shRNA. As seen from Fig. 4A, HIF1α knockdown was neither able to inhibit GD-induced upregulation of Grp78, XBP1-s, and phosphorylation of PERK nor inhibit GD-induced expression of VEGF, FGF-2, and IL-6 (Fig. 4B and C), However, knockdown of HIF1α inhibited CoCl2 (a mimetic of hypoxia)–induced VEGF and IL-6 expression at mRNA levels but not FGF2 (Fig. 4C). This may be because IL-6 (28) and VEGF (32) are transcriptionally controlled by hypoxia, whereas FGF-2 is not (29). Collectively, the results indicate that HIF1α is not involved in UPR-mediated production of angiogenic mediators.

Figure 4.

HIF1α is not involved in production of UPR-mediated proangiogenic mediators. Stable cell lines 81B-scshRNA, 81B-shHIF1α1, and 81B-shHIF1α2 were established with lentiviral vectors and treated with GD (2 mmol/L) or CoCl2 for 24 hours. Cells cultured in regular glucose (25 mmol/L) were used as untreated control (NT). A, UPR markers and HIF1α were assessed with Western blot analysis. Grp78 expression was determined by qPCR. B, ELISA shows VEGF levels in the supernatant. C, expression of angiogenic factors was quantified with qPCR and normalized to untreated control. *, P < 0.05.

Figure 4.

HIF1α is not involved in production of UPR-mediated proangiogenic mediators. Stable cell lines 81B-scshRNA, 81B-shHIF1α1, and 81B-shHIF1α2 were established with lentiviral vectors and treated with GD (2 mmol/L) or CoCl2 for 24 hours. Cells cultured in regular glucose (25 mmol/L) were used as untreated control (NT). A, UPR markers and HIF1α were assessed with Western blot analysis. Grp78 expression was determined by qPCR. B, ELISA shows VEGF levels in the supernatant. C, expression of angiogenic factors was quantified with qPCR and normalized to untreated control. *, P < 0.05.

Close modal

PERK/ATF4 pathway is involved in UPR-mediated angiogenesis

Upon UPR activation, approximately one third of UPR-responsive gene transcription requires phosphorylation of eIF2α, suggesting signaling from PERK regulates the UPR at transcriptional level (33) and is important for survival of tumor cells (34). We therefore, focused on the role of PERK/ATF4 pathway in the production of angiogenic mediators. During UPR, PERK activation leads to eIF2α phosphorylation, an increase of ATF4 expression, and subsequent upregulation of CHOP and Grp78 (35, 36). To examine the role of PERK/ATF4 pathway in angiogenic mediator production, lentiviral vectors containing shRNA against PERK and ATF4 were used to infect UM-SCC-81B cells, generating stable cell lines with specific knockdown of PERK and ATF4, respectively. As shown in Fig. 5A, shRNAs inhibit more than 70% of PERK expression and lead to reduced expression of its downstream genes (Grp78 and ATF4) upon GD treatment. In addition, PERK knockdown in UM-SCC-81B cell line decreased VEGF expression at both protein and mRNA levels (P < 0.05; Fig. 5B and C). Expression levels of FGF-2 and IL-6 were also significantly suppressed (Fig. 5C). To further corroborate the results above, PERK knockout (PERK −/−) MEFs were treated with GD (2 mmol/L). In PERK −/− cells, expression of VEGF was significantly suppressed as compared with PERK +/+ cells (Supplementary Fig. S4A–S4C).

Figure 5.

PERK is involved in UPR-mediated angiogenic factor production. Stable cell lines 81B-scshRNA, 81B-shPERK3, and 81B-shPERK7 were established with lentiviral vectors and treated without (NT) or with low glucose (2 mmol/L) for 24 hours. A, UPR markers (PERK, ATF4, and Grp78) were assessed with Western blot analysis. The relative density of PERK was measured and normalized to NT. B, secreted VEGF was measured with ELISA. C, expression of angiogenic factors (IL-6, FGF-2, and VEGF) was quantified with qPCR and normalized to NT. *, P < 0.05.

Figure 5.

PERK is involved in UPR-mediated angiogenic factor production. Stable cell lines 81B-scshRNA, 81B-shPERK3, and 81B-shPERK7 were established with lentiviral vectors and treated without (NT) or with low glucose (2 mmol/L) for 24 hours. A, UPR markers (PERK, ATF4, and Grp78) were assessed with Western blot analysis. The relative density of PERK was measured and normalized to NT. B, secreted VEGF was measured with ELISA. C, expression of angiogenic factors (IL-6, FGF-2, and VEGF) was quantified with qPCR and normalized to NT. *, P < 0.05.

Close modal

As a downstream effector of PERK, it is conceivable that ATF4 is also involved in GD-induced angiogenic factor production. Studies have shown that homocysteine, an ER stressor, and arsenite increase VEGF transcription in an ATF4-dependent manner (25, 37). By knocking down ATF4, we observed decreased expression of Grp78 (Fig. 6A) and proangiogenic factors (VEGF, IL-6, and FGF-2) were significantly suppressed (P < 0.05; Fig. 6B and C).

Figure 6.

ATF4 is involved in UPR-mediated angiogenic factor production. Stable cell lines 81B-scshRNA, 81B-shATF4-5, and 81B-shATF4-9 were established with lentiviral vectors and treated without (NT) or with GD (2 mmol/L) for 24 hours. A, Grp78 and ATF4 were assessed with Western blot analysis and qPCR. B, angiogenic factor expression was quantified with qPCR. C, secreted VEGF was quantified with ELISA. D, UM-SCC-81B cells were treated with GD (2 mmol/L) for 18 hours and processed for ChIP assay. PCR products of VEGF promoter fragments (AsnSyn site), asparagine synthetase (NSRE-1 site), and GAPDH genes were resolved in 1% agarose gel and stained with SYBR Green. The input was obtained by amplification of the initial unfractionated cell extracts. VEGF signal was quantified and normalized to GAPDH. *, P < 0.05.

Figure 6.

ATF4 is involved in UPR-mediated angiogenic factor production. Stable cell lines 81B-scshRNA, 81B-shATF4-5, and 81B-shATF4-9 were established with lentiviral vectors and treated without (NT) or with GD (2 mmol/L) for 24 hours. A, Grp78 and ATF4 were assessed with Western blot analysis and qPCR. B, angiogenic factor expression was quantified with qPCR. C, secreted VEGF was quantified with ELISA. D, UM-SCC-81B cells were treated with GD (2 mmol/L) for 18 hours and processed for ChIP assay. PCR products of VEGF promoter fragments (AsnSyn site), asparagine synthetase (NSRE-1 site), and GAPDH genes were resolved in 1% agarose gel and stained with SYBR Green. The input was obtained by amplification of the initial unfractionated cell extracts. VEGF signal was quantified and normalized to GAPDH. *, P < 0.05.

Close modal

The ChIP assay is widely used to show the interaction between transcription factor activation and cis-acting elements. The “AsnSyn” site in the VEGF gene is an important ATF4 binding site (25, 26). To further verify the role of ATF4 in regulating VEGF expression, we examined the binding of ATF4 to the “AsnSyn” site using ChIP assay. As shown in Fig. 6D, specific PCR product was present in GD-treated samples immunoprecipitated with antibody against ATF4. In addition, GD also stimulated formation of a complex between ATF4 and a functionally important NSRE-1 site in the promoter of asparagine synthetase (an ATF4 target gene) as mentioned earlier (26, 38). PCR bands for GAPDH were observed in the samples immunoprecipitated with anti-RNA polymerase II with or without GD treatment. No PCR products were observed in rabbit IgG immunoprecipited samples (Fig. 6D, left). The quantification from ChIP assay clearly shows that GD promotes strong interaction between the VEGF “AsnSyn” site and ATF4 (Fig. 6D, right). Collectively, these results show that the PERK/ATF4 pathway plays a pivotal role in GD-induced VEGF production.

UPR activation in tumor cells promotes tumor vascularization and proliferation

To determine how the UPR stimulates angiogenesis, we used conditioned medium (CM) collected from GD-treated tumor cells (GDCM) to induce HDMEC proliferation and sprout formation on collagen gels. When exposed to angiogenic stimuli, HDMECs proliferate, migrate, and organize into capillary-like structures (39, 40). To assess sprout formation, HDMECs were grown in the presence of CM collected from UM-SCC-81B-scshRNA and UM-SCC-81B-shPERK7 with or without GD treatment. GDCM showed strong ability in promoting capillary-like sprout formation on collagen as compared with CM collected from cells grown in normal glucose (NGCM). PERK knockdown, however, decreased the ability of GDCM in inducing sprout formation (Fig. 7A).

Figure 7.

UPR regulates blood vessel formation and tumor progression. A, sprout formation assay was used to evaluate the ability of CM from GD-treated UM-SCC-81B cells (with or without PERK knockdown) in inducing HDMEC sprout formation (arrowheads show representative sprouts). HDMEC proliferation in response to CM was measured with MTT assay; NGCM plus 50 ng/mL VEGF (NGCM-V) or NGCM plus 50 ng/mL FGF2 (NGCM-F) was used as positive control. Cell viability was normalized to the samples treated with NGCM (100%). B, tumor volume (measured every 3 days) and tumor weight (at endpoint) were used to evaluate tumor progression. C, Ki67 and Grp78 expression in xenograft tumors was examined by IHC staining, and Ki67 expression was quantified. D, blood vessel density was detected by CD31 staining and defined as the number of blood vessels per field. *, P < 0.05.

Figure 7.

UPR regulates blood vessel formation and tumor progression. A, sprout formation assay was used to evaluate the ability of CM from GD-treated UM-SCC-81B cells (with or without PERK knockdown) in inducing HDMEC sprout formation (arrowheads show representative sprouts). HDMEC proliferation in response to CM was measured with MTT assay; NGCM plus 50 ng/mL VEGF (NGCM-V) or NGCM plus 50 ng/mL FGF2 (NGCM-F) was used as positive control. Cell viability was normalized to the samples treated with NGCM (100%). B, tumor volume (measured every 3 days) and tumor weight (at endpoint) were used to evaluate tumor progression. C, Ki67 and Grp78 expression in xenograft tumors was examined by IHC staining, and Ki67 expression was quantified. D, blood vessel density was detected by CD31 staining and defined as the number of blood vessels per field. *, P < 0.05.

Close modal

During sprout formation assay, we noted consistently higher confluence rates in cells cultured in the presence of GDCM, despite equal seeding of cells, suggesting GDCM might be able to promote cell proliferation. MTT assay verified this observation. As shown in Fig. 7A, GDCM promoted stronger cell proliferation as compared with NGCM (P < 0.05). NGCM supplemented with FGF2 showed the strongest ability in promoting HDMEC proliferation (Fig. 7A).

The effect of PERK knockdown on tumor proliferation and blood vessel formation was also evaluated in vivo. As seen, PERK knockdown significantly reduced the tumor volume and slowed down tumor progression (Fig. 7B). Knockdown of PERK reduced Ki67-positive cells from 44.6% in controls to 12% in PERK knockdown tumors, suggesting that PERK plays an important role in tumor growth (Fig. 7C). As expected, suppressing PERK expression in tumor cells also reduced the blood vessel density from 8 to 4 per field, which can reduce tumor volume due to a decreased blood supply (Fig. 7D). To confirm these results, a rapid growing tumor cell line (UM-SCC-74B) was used in vivo. In UM-SCC-74B, PERK knockdown significantly reduced tumor proliferation as shown by tumor volume, tumor weight, and Ki67 staining (Supplementary Fig. S5A and S5B). And blood vessel density in UM-SCC-74B tumors decreased from 17 to 8 per field (Supplementary Fig. S5C).

TME is often hypoxic and nutrient deficient due to an inadequate blood supply. This in turn leads to activation of the UPR. The main function of the UPR is to maintain or restore ER homeostasis in response to environmental stress. It is therefore not surprising that one mechanism for relieving this stress is by increasing the blood supply to tumors. It has been shown that in prostate and ovarian carcinoma, tumor cells subjected to GD show elevated levels of VEGF production (41, 42). However, due to the complex nature of the angiogenic process, it is likely that multiple proangiogenic mediators produced at the tumor site accumulate in sufficient quantities to offset antiangiogenic mediators. With immunostaining, we found strong expression of the UPR markers Grp78 and CHOP in tumors from patients with HNSCC, whereas in normal human mucosa there was only weak expression of Grp78 and CHOP. Meanwhile, LCM analysis revealed expression levels of proangiogenic factors were elevated in tumor tissues, whereas the antiangiogenic mediator CXCL14 was significantly reduced. This supports the notion that UPR activation coordinates the expression of pro- and antiangiogenic mediators to facilitate blood vessel formation, which subsequently reduces stress.

Similar to tumor samples, tumor cell lines treated with GD showed UPR activation along with simultaneous increase of proangiogenic genes, such as VEGF, FGF-2, IL-6, CTGF, etc., and decrease in a number of antiangiogenic genes (CXCL14, CXCL10, and THBS1), with CXCL14 showing the most significant reduction (28.21-fold). These results confirm that UPR plays a pivotal role in the angiogenic switch in tumors.

Pereira and colleagues recently reported that treatment of the human medulloblastoma cell line (Daoy) with the chemical ER stressor TG, induced expression of proangiogenic factors including IL-8, angiogenin, angiopoietin-2, etc., and reduced expression of the antiangiogenic protein vasohibin (21). Interestingly, our studies revealed major differences in the spectrum of genes involved in angiogenesis as compared with that reported by Pereira and colleagues, which suggests different stressors induce a distinct pattern of angiogenic mediator expression. Indeed, we found that GD (2 mmol/L) and TM (1 μg/mL) can induce strong VEGF expression but not IL-8 expression. TG, at 100 nmol/L, cannot induce strong VEGF expression but can induce significant IL-8 expression in UM-SCC-81B cells (Supplementary Fig. S6A and S6B). Higher TG concentration (1 μmol/L) can, however, induce VEGF (Fig. 3A). This apparent discrepancy may be because TG releases calcium from the ER into the cytosol and IL-8 expression is dependent on the change in cytosolic calcium (43). We contend that activation of the UPR and subsequent activation of the angiogenic switch by GD more closely resembles the events occurring in the tumor microenvironment. Nevertheless, these observations corroborate our hypothesis that UPR activation in tumors induces the angiogenic switch. Indeed, HDMEC treated with GDCM produce more capillary-like sprouts on collagen gel and suppressing PERK/ATF4 pathway of the UPR reduces tube formation ability of GDCM in inducing tube formation.

It has been reported that both GD and hypoxia activate the UPR (44). The UPR component PERK, has been linked to translation inhibition under hypoxic stress (45) and XBP1 has been shown to be essential to the survival of transformed cells in response to hypoxia (46). The best-characterized stress-induced regulator of tumor angiogenesis is HIF1α. Upregulation of VEGF in response to GD was found to be HIF1α-dependent in mouse embryonic stem cells (31), suggesting the TME-related stimuli, GD, and hypoxia, share similar signaling pathways. However, others and we showed that while GD could activate UPR, it could not promote accumulation of HIF1α (21, 47). Furthermore, although the hypoxia mimetic, CoCl2 promoted the accumulation of HIF1α, it was unable to promote overexpression of UPR markers Grp78, XBP1-s, and phosphorylation of PERK. Knockdown of HIF1α had no effect on GD-induced VEGF, IL-6, and FGF-2 expression. These results suggest that the pathways activated following GD and CoCl2 treatment are different and HIF1α is not involved in angiogenic mediator production that is associated with GD-induced UPR. Our studies confirm earlier observations that HIF1α was not involved in UPR-mediated VEGF expression (21, 22).

The PERK/ATF4 pathway has been reported to mediate VEGF mRNA expression in mouse cell lines (MEF, neruo2A), human umbilical vein endothelial cells (HUVEC), human retinal pigment epithelial cell (ARPE-19), and hepatic carcinoma cell line (HepG2) following activation of the UPR with TG or TM (21, 22, 25, 26). However, its role in GD-induced UPR-mediated angiogenesis in human tumors has not been reported. We show here that the PERK/ATF4 pathway of the UPR is involved not only in VEGF expression but also FGF-2 and IL-6 expression in human tumor cells treated with GD. An approximately 70% knockdown of PERK is sufficient to reduce expression of these proangiogenic mediators. In vivo results confirmed that PERK plays an important role in tumor proliferation and neovascularization. Direct knockdown of the downstream target of PERK, ATF4, displayed an even stronger inhibitory effect in reducing VEGF, IL-6, and FGF-2 expression. This suggests a central role of ATF4 in the production of proangiogenic mediators. A possible explanation is that as an ATF, ATF4 regulates the expression of these genes directly.

In summary, our studies show that GD-induced UPR activation initiates an angiogenic switch that alters the balance of pro- and antiangiogenic mediators. The resulting proangiogenic environment could function to relieve the stress by increasing the blood supply to tumors. We also found that the PERK/ATF4 arm of UPR signaling is a pivotal pathway responsible for upregulating the production of multiple proangiogenic mediators. In conclusion, these results suggest that the role of UPR-mediated stress response must be taken into consideration as a potential target in the design of new cancer therapies.

No potential conflicts of interest were disclosed.

Conception and design: Y. Wang, F. Visioli, J.E. Nör, P.J. Polverini

Development of methodology: Y. Wang, F. Visioli, Z. Dong, J.E. Nör

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Wang, G.N. Alam, Y. Ning, F. Visioli, Z. Dong

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Wang, G.N. Alam, J.E. Nör, P.J. Polverini

Writing, review, and/or revision of the manuscript: Y. Wang, G.N. Alam, J.E. Nör, P.J. Polverini

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Wang, Y. Ning, Z. Dong

Study supervision: Y. Wang, P.J. Polverini

The authors thank members from Jacques E. Nör's laboratory, Dr. Andrew Fribley, and members from Randal J. Kaufman's Laboratory for helpful discussions and assistance. The authors also thank University of Michigan core facilities for their technical assistance.

This work is supported by The Sharon and Lauren Daniels Cancer Research Fund and University of Michigan, Office of the Provost, grant P50-CA97248, from the NIH/NCI, and grant R01-DE21139 from the NIH/NIDCR.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Mahadevan
NR
,
Zanetti
M
. 
Tumor stress inside out: cell-extrinsic effects of the unfolded protein response in tumor cells modulate the immunological landscape of the tumor microenvironment
.
J Immunol
2011
;
187
:
4403
9
.
2.
Schroder
M
,
Kaufman
RJ
. 
The mammalian unfolded protein response
.
Annu Rev Biochem
2005
;
74
:
739
89
.
3.
Li
X
,
Zhang
K
,
Li
Z
. 
Unfolded protein response in cancer: the physician's perspective
.
J Hematol Oncol
2011
;
4
:
8
.
4.
Xu
C
,
Bailly-Maitre
B
,
Reed
JC
. 
Endoplasmic reticulum stress: cell life and death decisions
.
J Clin Invest
2005
;
115
:
2656
64
.
5.
Merksamer
PI
,
Papa
FR
. 
The UPR and cell fate at a glance
.
J Cell Sci
2010
;
123
:
1003
6
.
6.
Bertolotti
A
,
Zhang
Y
,
Hendershot
LM
,
Harding
HP
,
Ron
D
. 
Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response
.
Nat Cell Biol
2000
;
2
:
326
32
.
7.
Lu
PD
,
Harding
HP
,
Ron
D
. 
Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response
.
J Cell Biol
2004
;
167
:
27
33
.
8.
Ma
Y
,
Brewer
JW
,
Diehl
JA
,
Hendershot
LM
. 
Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response
.
J Mol Biol
2002
;
318
:
1351
65
.
9.
Rutkowski
DT
,
Arnold
SM
,
Miller
CN
,
Wu
J
,
Li
J
,
Gunnison
K M
, et al
Adaptation to ER stress is mediated by differential stabilities of pro-survival and pro-apoptotic mRNAs and proteins
.
PLoS Biol
2006
;
4
:
e374
.
10.
Luo
S
,
Baumeister
P
,
Yang
S
,
Abcouwer
SF
,
Lee
AS
. 
Induction of Grp78/BiP by translational block: activation of the Grp78 promoter by ATF4 through and upstream ATF/CRE site independent of the endoplasmic reticulum stress elements
.
J Biol Chem
2003
;
278
:
37375
85
.
11.
Yoshida
H
,
Matsui
T
,
Yamamoto
A
,
Okada
T
,
Mori
K
. 
XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor
.
Cell
2001
;
107
:
881
91
.
12.
Calfon
M
,
Zeng
H
,
Urano
F
,
Till
J H
,
Hubbard
SR
,
Harding
HP
, et al
IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA
.
Nature
2002
;
415
:
92
6
.
13.
Lee
AH
,
Iwakoshi
NN
,
Glimcher
LH
. 
XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response
.
Mol Cell Biol
2003
;
23
:
7448
59
.
14.
Ye
J
,
Rawson
RB
,
Komuro
R
,
Chen
X
,
Dave
UP
,
Prywes
R
, et al
ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs
.
Mol Cell
2000
;
6
:
1355
64
.
15.
Park
SH
,
Kim
KW
,
Lee
YS
,
Baek
JH
,
Kim
MS
,
Lee
YM
, et al
Hypoglycemia-induced VEGF expression is mediated by intracellular Ca2+ and protein kinase C signaling pathway in HepG2 human hepatoblastoma cells
.
Int J Mol Med
2001
;
7
:
91
6
.
16.
Satake
S
,
Kuzuya
M
,
Miura
H
,
Asai
T
,
Ramos
MA
,
Muraguchi
M
, et al
Up-regulation of vascular endothelial growth factor in response to glucose deprivation
.
Biol Cell
1998
;
90
:
161
8
.
17.
Stein
I
,
Neeman
M
,
Shweiki
D
,
Itin
A
,
Keshet
E
. 
Stabilization of vascular endothelial growth factor mRNA by hypoxia and hypoglycemia and coregulation with other ischemia-induced genes
.
Mol Cell Biol
1995
;
15
:
5363
8
.
18.
Shweiki
D
,
Itin
A
,
Soffer
D
,
Keshet
E
. 
Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis
.
Nature
1992
;
359
:
843
5
.
19.
Ikeda
E
,
Achen
MG
,
Breier
G
,
Risau
W
. 
Hypoxia-induced transcriptional activation and increased mRNA stability of vascular endothelial growth factor in C6 glioma cells
.
J Biol Chem
1995
;
270
:
19761
6
.
20.
Marjon
PL
,
Bobrovnikova-Marjon
EV
,
Abcouwer
SF
. 
Expression of the pro-angiogenic factors vascular endothelial growth factor and interleukin-8/CXCL8 by human breast carcinomas is responsive to nutrient deprivation and endoplasmic reticulum stress
.
Mol Cancer
2004
;
3
:
4
.
21.
Ghosh
R
,
Lipson
KL
,
Sargent
KE
,
Mercurio
AM
,
Hunt
JS
,
Ron
D
, et al
Transcriptional regulation of VEGF-A by the unfolded protein response pathway
.
PLoS ONE
;
5
:
e9575
.
22.
Pereira
ER
,
Liao
N
,
Neale
GA
,
Hendershot
LM
. 
Transcriptional and post-transcriptional regulation of proangiogenic factors by the unfolded protein response
.
PLoS ONE
2010
;
5
:
e12521
.
23.
Zhang
Z
,
Neiva
KG
,
Lingen
MW
,
Ellis
LM
,
Nor
JE
. 
VEGF-dependent tumor angiogenesis requires inverse and reciprocal regulation of VEGFR1 and VEGFR2
.
Cell Death Differ
2010
;
17
:
499
512
.
24.
Kaneko
T
,
Zhang
Z
,
Mantellini
MG
,
Karl
E
,
Zeitlin
B
,
Verhaegen
M
, et al
Bcl-2 orchestrates a cross-talk between endothelial and tumor cells that promotes tumor growth
.
Cancer Res
2007
;
67
:
9685
93
.
25.
Roybal
CN
,
Hunsaker
LA
,
Barbash
O
,
Vander
Jagt DL
,
Abcouwer
SF
. 
The oxidative stressor arsenite activates vascular endothelial growth factor mRNA transcription by an ATF4-dependent mechanism
.
J Biol Chem
2005
;
280
:
20331
9
.
26.
Oskolkova
OV
,
Afonyushkin
T
,
Leitner
A
,
von Schlieffen
E
,
Gargalovic
PS
,
Lusis
AJ
, et al
ATF4-dependent transcription is a key mechanism in VEGF up-regulation by oxidized phospholipids: critical role of oxidized sn-2 residues in activation of unfolded protein response
.
Blood
2008
;
112
:
330
9
.
27.
Schweda
F
,
Blumberg
FC
,
Schweda
A
,
Nabel
C
,
Holmer
SR
,
Riegger
GA
, et al
Effects of chronic hypoxia on renal PDGF-A, PDGF-B, and VEGF gene expression in rats
.
Nephron
2000
;
86
:
161
6
.
28.
Yan
SF
,
Tritto
I
,
Pinsky
D
,
Liao
H
,
Huang
J
,
Fuller
G
, et al
Induction of interleukin 6 (IL-6) by hypoxia in vascular cells. Central role of the binding site for nuclear factor-IL-6
.
J Biol Chem
1995
;
270
:
11463
71
.
29.
Conte
C
,
Riant
E
,
Toutain
C
,
Pujol
F
,
Arnal
JF
,
Lenfant
F
, et al
FGF2 translationally induced by hypoxia is involved in negative and positive feedback loops with HIF-1alpha
.
PLoS ONE
2008
;
3
:
e3078
.
30.
Green
CJ
,
Lichtlen
P
,
Huynh
NT
,
Yanovsky
M
,
Laderoute
KR
,
Schaffner
W
, et al
Placenta growth factor gene expression is induced by hypoxia in fibroblasts: a central role for metal transcription factor-1
.
Cancer Res
2001
;
61
:
2696
703
.
31.
Ryan
HE
,
Lo
J
,
Johnson
RS
. 
HIF-1 alpha is required for solid tumor formation and embryonic vascularization
.
EMBO J
1998
;
17
:
3005
15
.
32.
Forsythe
JA
,
Jiang
BH
,
Iyer
NV
,
Agani
F
,
Leung
SW
,
Koos
RD
, et al
Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1
.
Mol Cell Biol
1996
;
16
:
4604
13
.
33.
Shen
X
,
Zhang
K
,
Kaufman
RJ
. 
The unfolded protein response—a stress signaling pathway of the endoplasmic reticulum
.
J Chem Neuroanat
2004
;
28
:
79
92
.
34.
Blais
JD
,
Addison
CL
,
Edge
R
,
Falls
T
,
Zhao
H
,
Wary
K
, et al
Perk-dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress
.
Mol Cell Biol
2006
;
26
:
9517
32
.
35.
Malhotra
JD
,
Kaufman
RJ
. 
The endoplasmic reticulum and the unfolded protein response
.
Semin Cell Dev Biol
2007
;
18
:
716
31
.
36.
Inageda
K
. 
Insulin modulates induction of glucose-regulated protein 78 during endoplasmic reticulum stress via augmentation of ATF4 expression in human neuroblastoma cells
.
FEBS Lett
2010
;
584
:
3649
54
.
37.
Roybal
CN
,
Yang
S
,
Sun
CW
,
Hurtado
D
,
Vander
Jagt DL
,
Townes
TM
, et al
Homocysteine increases the expression of vascular endothelial growth factor by a mechanism involving endoplasmic reticulum stress and transcription factor ATF4
.
J Biol Chem
2004
;
279
:
14844
52
.
38.
Gjymishka
A
,
Su
N
,
Kilberg
MS
. 
Transcriptional induction of the human asparagine synthetase gene during the unfolded protein response does not require the ATF6 and IRE1/XBP1 arms of the pathway
.
Biochem J
2009
;
417
:
695
703
.
39.
Nor
JE
,
Christensen
J
,
Mooney
DJ
,
Polverini
PJ
. 
Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression
.
Am J Pathol
1999
;
154
:
375
84
.
40.
DiPietro
LA
,
Nebgen
DR
,
Polverini
PJ
. 
Downregulation of endothelial cell thrombospondin 1 enhances in vitro angiogenesis
.
J Vasc Res
1994
;
31
:
178
85
.
41.
Yun
H
,
Lee
M
,
Kim
SS
,
Ha
J
. 
Glucose deprivation increases mRNA stability of vascular endothelial growth factor through activation of AMP-activated protein kinase in DU145 prostate carcinoma
.
J Biol Chem
2005
;
280
:
9963
72
.
42.
Adham
SA
,
Coomber
BL
. 
Glucose is a key regulator of VEGFR2/KDR in human epithelial ovarian carcinoma cells
.
Biochem Biophys Res Commun
2009
;
390
:
130
5
.
43.
Yu
Y
,
De Waele
C
,
Chadee
K
. 
Calcium-dependent interleukin-8 gene expression in T84 human colonic epithelial cells
.
Inflamm Res
2001
;
50
:
220
6
.
44.
Kaufman
RJ
,
Scheuner
D
,
Schroder
M
,
Shen
X
,
Lee
K
,
Liu
CY
, et al
The unfolded protein response in nutrient sensing and differentiation
.
Nat Rev Mol Cell Biol
2002
;
3
:
411
21
.
45.
Koumenis
C
,
Naczki
C
,
Koritzinsky
M
,
Rastani
S
,
Diehl
A
,
Sonenberg
N
, et al
Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha
.
Mol Cell Biol
2002
;
22
:
7405
16
.
46.
Romero-Ramirez
L
,
Cao
H
,
Nelson
D
,
Hammond
E
,
Lee
AH
,
Yoshida
H
, et al
XBP1 is essential for survival under hypoxic conditions and is required for tumor growth
.
Cancer Res
2004
;
64
:
5943
7
.
47.
Drogat
B
,
Auguste
P
,
Nguyen
DT
,
Bouchecareilh
M
,
Pineau
R
,
Nalbantoglu
J
, et al
IRE1 signaling is essential for ischemia-induced vascular endothelial growth factor-A expression and contributes to angiogenesis and tumor growth in vivo
.
Cancer Res
2007
;
67
:
6700
7
.