The hypoxia-mimetic agent cobalt chloride induces cell cycle arrest and alters gene expression in U266 multiple myeloma cells

  • Authors:
    • Seunghee Bae
    • Hye-Jung Jeong
    • Hwa Jun Cha
    • Karam Kim
    • Yeong Min Choi
    • In-Sook An
    • Hyea Jung Koh
    • Dae Jin Lim
    • Su-Jae Lee
    • Sungkwan An
  • View Affiliations

  • Published online on: August 31, 2012     https://doi.org/10.3892/ijmm.2012.1115
  • Pages: 1180-1186
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypoxia is a common feature of tumors that occurs across a wide variety of malignancies. Multiple myeloma is an incurable malignant disorder of plasma cells in the bone marrow. Although bone marrow hypoxia is crucial for normal hematopoiesis, the effect of hypoxia on multiple myeloma is poorly understood. In this study, we demonstrated that cobalt chloride (CoCl2)-mediated hypoxia decreased cell viability and altered gene expression in U266 human multiple myeloma cells. CoCl2 induced the loss of cell viability in a concentration-dependent manner. In addition, FACS analysis revealed that the loss of cell viability was related to apoptosis. Using microarray analysis, we identified mRNA expression profile changes in response to CoCl2 treatment in U266 cells. Four hundred and fifty-two mRNAs exhibited >2-fold changes in expression in CoCl2-treated U266 cells compared to their expression in control cells. A follow-up bioinformatics study revealed that a great number of genes with altered expression were involved in apoptosis, cell cycle, transcription and development. In conclusion, these results provide novel evidence that CoCl2-mediated hypoxia affects the expression profiles of genes that are functionally related to apoptosis and angiogenesis in U266 multiple myeloma cells.

Introduction

Hypoxia, defined as areas with oxygen tension values of 10 mmHg or lower, is a common feature of solid tumors that occurs across a wide variety of malignancies (1,2). Once cells encounter hypoxia, a large number of genes that stimulate adaptation to oxygen deprivation are induced. These hypoxia-inducible genes are primarily controlled by the transcription factor hypoxia-inducible factor 1 (HIF-1), including those involved in cell cycle arrest, apoptosis, erythropoiesis, angiogenesis, glycolytic metabolism, and tumor invasiveness (2,3). HIF-1 is a heterodimer consisting of a constitutively expressed HIF-1β subunit and a HIF-1α subunit that is regulated in an oxygen-dependent manner (3). Under hypoxia, HIF-1α can block its own degradation and thus induce its accumulation (3). Therefore, HIF-1α is overexpressed in many cancer types and is associated with poor prognosis in cancers of the breast, brain, cervix, ovary, and uterus (3).

HIF-1α activation in response to hypoxia stimulates the transcription of several of its target genes. Vascular endothelial growth factor (VEGF), which is known to promote angiogenesis, is strongly induced by HIF-1α (4). In addition, matrix metalloproteinase (MMP)-1 and MMP-2 are induced to promote intravasation (5,6). Additionally, the epithelial-mesenchymal transition-inducing genes E-cadherin, SNAIL, TWIST, TCF3, and ZEB1 are up-regulated in a HIF-1α-dependent manner (79). Interestingly, HIF-1α also induces cell cycle arrest by up-regulating p53, p21, and E1B 19K/Bcl-2 binding protein Nip3 expression (1012). These findings suggest that there are different adaptive responses to oxygen deprivation.

Cobalt chloride (CoCl2), a commonly used hypoxia-mimetic agent, artificially induces hypoxia and can block the degradation and thus induce the accumulation of HIF-1α protein (13,14). Many reports have indicated that both CoCl2 and hypoxia regulate a similar group of genes on a global gene expression level (1517). In addition, the reports demonstrated that hypoxia and CoCl2 induced different effects in many different types of cells. For example, CoCl2 induced apoptosis in rat C6 glioma cells, human alveolar macrophages, neuronal PC12 cells, and HeLa human cervical cancer cells, although it inhibited apoptotic death in HepG2 hepatoma cells (1822). Moreover, CoCl2 induced prostate tumor cell adhesion, metastasis, and angiogenesis (2325) and stimulated oral squamous carcinoma cell growth (26).

Multiple myeloma (MM) is a B-cell malignancy selectively localized in the bone marrow (BM) (27). Unlike other organs, the normal BM microenvironment is hypoxic. This physiological hypoxia is crucial for normal marrow hematopoiesis (28). In several studies, oxygen levels in BM in mice with MM were depressed (29,30). This hypoxia is an important microenvironmental stimulus for activities critical for MM disease progression such as MM cell growth, drug resistance, and angiogenesis (29,30). However, how hypoxia and MM affect each other is poorly understood. Therefore, an investigation of the effects of hypoxia on myeloma cells is necessary.

Materials and methods

Cell culture

The human multiple myeloma cell line U266 was purchased from the Korean Cell Line Bank (Seoul, Korea) and cultured in RPMI-1640 medium containing 10% fetal bovine serum and antibiotics at 37°C in a humidified chamber containing 5% CO2. Cells were seeded into 60-mm culture dishes (4×105 cells per dish) 1 day before CoCl2 (Sigma, St. Louis, MO, USA) treatment.

Hypoxia induction

CoCl2 was dissolved in dimethylsulfoxide (DMSO; Sigma) and added to U266 cultures at the indicated final concentrations. DMSO was added to culture medium alone as a vehicle control. Cells were treated for 24 h.

RNA preparation and cDNA synthesis

Total RNA was extracted from cells by using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions after the 24-h CoCl2 treatment. For the microarray studies, both the quality and concentration of the RNA samples were determined using an Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA) and an Ultrospec 3300 Pro UV/Visible Spectrophotometer (Amersham Biosciences, UK). The recommended RNA quality parameters for microarray analysis are as follows: a UV spectroscopy A260/A280 ratio of 1.8–2.0 and an A260/A230 ratio >1.8, an 18S/28S rRNA ratio of 1.8–2.1, and an RNA integrity number >8.0. To synthesize cDNA, 1 μg of RNA was incubated with oligo-dT primers at 94°C for 10 min and reverse-transcribed with reverse transcriptase (Enzynomics, Seoul, Korea) at 37°C for 1 h.

DNA microarray analysis

DNA microarray analysis was performed using the HumanHT-12 v4.0 Expression Beadchip kit (Illumina, San Diego, CA, USA) according to the instruction manual. Derived data were analyzed using Genespring GX 11 (Agilent Technologies). The raw data were filtered using FLAG and t-tests. Significant genes were determined using the fluorescence ratio between the control and CoCl2-treated samples, and genes displaying a >2-fold increase or decrease were selected for analysis.

Immunoblotting and polymerase chain reaction (PCR)

Immunoblot analysis was performed as described previously (31). A primary antibody specific for HIF-1α was purchased from Cell Signaling Technology (Danvers, MA, USA). Anti-β-actin antibody was purchased from Sigma. The expression level of VEGF mRNA was determined by quantitative real-time (RT)-PCR using specific primers (forward, 5′-AAGGAGGAGGGCAGAATCAT-3′; reverse, 5′-GCTGTAGGAAGCTCATCTCT-3′). RT-PCR analysis was performed using Line-Gene K software (Bioer Technology Co. Ltd., Hangzhou, China).

Cell viability assay and FACS analysis

Cell proliferation was determined using the WST-1 assay (EZ-Cytox Cell Viability Assay kit, ITSBIO, Korea) according to the manufacturer’s instructions. In brief, 4×105 cells were seeded in 60-mm dishes. The cells were cultured for 24 h and treated with the indicated concentrations of CoCl2 for 24 h. At each time-point, the kit solution was added to cultured cells, which were incubated at 37°C for 30 min. Cell viability was measured using an iMark microplate reader (Bio-Rad, Hercules, CA, USA) at 450 nm by using a 620-nm reference filter. Cell cycle distribution was determined using FACS analysis. U266 cells were collected and fixed by resuspending them in 70% ethanol for 1 h, centrifuged, and washed in cold PBS. The cell pellets were resuspended in PBS containing 50 μg/ml propidium iodide (Sigma) and 100 μg/ml RNase, incubated at room temperature for 30 min, and then analyzed using a FACScalibur (BD Biosciences, San Jose, CA, USA).

Statistical analysis

Statistical analysis was performed using the χ2 test or Fisher’s exact test and Spearman rank correlation coefficient analysis. p<0.05 was considered significant.

Results and Discussion

We first examined whether CoCl2 stimulation induces hypoxia in MM U266 cells. The cells were treated with the indicated concentrations of CoCl2 for 4 h, and then the level of hypoxia was determined by immunoblotting against the hypoxia marker HIF-1α. As shown in Fig. 1A, the cellular protein levels of HIF-1α were greatly increased in a CoCl2 concentration-dependent manner. Further, to test whether the accumulated HIF-1α is functionally active in the cells, the expression level of a well-known HIF-1α target gene, VEGF, was verified by RT-PCR using its specific primers (see Materials and methods). As shown in Fig. 1B, the mRNA expression levels of VEGF were up-regulated by CoCl2 stimulation in U266 cells. Therefore, the hypoxia-mimetic agent CoCl2 induced hypoxia in U266 cells.

As hypoxia has dual roles in cell proliferation and cell death, we examined the effect of hypoxia on MM cell proliferation. U266 cells were stimulated with CoCl2 at the indicated concentrations for 24 h, and then WST-1-based cell viability assay was used to determine the level of cell proliferation. As shown in Fig. 2, CoCl2 decreased cell viability in a concentration-dependent manner. We next examined whether the loss of cell viability is related to cell cycle arrest or apoptosis. Interestingly, FACS analysis revealed that CoCl2 stimulation induces apoptosis, but not cell cycle arrest. These results indicate that the hypoxia induced cell death in U266 cells.

Although hypoxia alters cell type-specific gene expression patterns (32), its effects have been not fully studied in MM cells. Therefore, we analyzed mRNA profiles using Illumina Human HT-12 v4.0 Beadchip kits in hypoxia-stimulated and control U266 cells. A total of 47,000 human mRNAs were selected to analyze gene expression profiles. Those human genes were continually filtered with FLAG to obtain more defined data using Agilent GeneSpring GX 11 software. We selected total 452 human mRNAs as a result of FLAG filtration, and the genes displaying >2-fold differences in expression between the control and CoCl2-treated cells are shown in Fig. 3A. In addition, the mRNA microarray was visualized on a scatter plot (Fig. 3B). The data of the scatter blot demonstrated that many genes displayed >2-fold differences in expression levels between control and CoCl2-treated cells, as indicated by their distance from the diagonal line. The numbers of up- and downregulated mRNAs exhibiting >2-fold changes were derived from the expression patterns in each group. We found that 293 up-regulated and 159 down-regulated mRNA displayed meaningful changes in their transcription profiles in response to CoCl2 treatment (Fig. 3C). The 50 genes for which their expression was most strongly altered by CoCl2 are listed in Table I. To further analyze the changes in gene expression patterns, we sorted the genes displaying >2-fold changes in expression after CoCl2 treatment into several groups according to common features such as homology or biochemical activity by using the Gene Set Enrichment Analysis bioinformatics tool (www.broadiinstitute.org/gsea), and we revealed that the sorted gene sets were categorized into transcription factors, cell differentiation markers, cytokines and growth factors, protein kinases, tumor suppressors, and oncogenes (Table II). Moreover, we further identified the cellular process pattern of CoCl2-induced gene expression changes by using the gene ontology database. Although the analysis of cellular process pattern was firstly performed using genes with >2-fold changes in expression, the pattern results were not sufficient for elucidation because the p-values of each category were >0.05; therefore, the analysis was re-performed on genes with >1.5-fold changes in expression, and the patterns with p-values <0.05 were selected. As shown in Fig. 4A, we first identified the genomic region responsible for gene expression changes induced in response to CoCl2 treatment in U266 cells by sorting the genes according to their chromosomal positions and revealed that the regions within ch11q13 and ch11q21 (up-regulated genes) and ch19p13 and ch6p21 (down-regulated genes) were the top two chromosome regions. Next, we grouped the genes according to cellular process and found that the up-regulated genes were related to apoptosis, development, cell cycle regulation, immunity, transcription, and B cell activation; however, the downregulated genes were related to cell cycle regulation, transcription, and kinase activity (Fig. 4B). In particular, there are more changes among genes associated with both apoptosis and development, implying that in MM, hypoxia exerts both antitumor effects, and positively regulates cell proliferation pathways, although cell viability was decreased in hypoxic conditions.

Table I.

Top 50 genes altered by treatment with cobalt chloride.a

Table I.

Top 50 genes altered by treatment with cobalt chloride.a

Up-regulated genes
Down-regulated genes
Gene nameF.CGene nameF.CGene nameF.CGene nameF.C
OKL3810.5MT1A4.19HEMGN7.56XK2.69
RN7SK8.34C17ORF914.12RHOXF2B5.29EIF2AK22.66
EMP17.15RAB7B4.07TSPAN325.12ACSM32.61
TXNRD17.14MT1X4.01RHOXF25.00VEGFB2.60
CDK5RAP26.40CTH3.94TESC4.52SLC10A42.60
SRXN16.35SLC3A23.83ARHGAP224.11BOLA32.56
C12ORF486.33UGDH3.81KISS1R3.76C5ORF132.53
AGPAT96.03ARRDC43.77GYPA3.75GAD12.48
NDRG15.90SLCO2B13.74GYPE3.67HADH2.48
UPP15.79NOTCH13.52RHAG3.32MYB2.47
CTSL15.74MT1E3.48GAD13.24SPR2.47
SLC7A115.44CD533.38STAT5A3.22TFDP22.46
CD445.32SERPINE23.40CA13.21METTL7A2.45
FLNB5.23ITPR13.37SCARB13.13ATP5G12.45
INHBE5.07RN5S93.32ELOVL63.05C4ORF182.43
GCLM4.78DCUN1D33.28NME43.05NMRAL12.42
FAM129B4.78ISG203.25NFE23.02BEX42.42
BCL64.74CEBPB3.24CCDC342.99CYP3A52.42
EID34.70PRSS23.22ATF52.96CFD2.42
MT2A4.66KLF63.18KCNH22.94SLC1A32.42
DUSP54.62PHLDA13.16STS-12.83SOX212.41
XIRP14.59PAEP3.13ACPP2.82RELN2.41
OSGIN14.31KIAA16663.12ZNF1212.73SPIN42.40
SLC3A24.28IER53.10GATA12.73EVL2.39
ADM4.24LGMN3.09LIPH2.71PIK3R22.39

a The list was selected to show only the top 50 genes displaying >2-fold expression changes after FLAG sorting. Predicted genes were not selected in this list.

Table II.

Genes sharing a common feature such as homology or biochemical activity.a

Table II.

Genes sharing a common feature such as homology or biochemical activity.a

Cytokines and growth factorsTranscription factorsHomeo-domain proteinsCell differentiation markersProtein kinasesTranslocated cancer genesOncogenesTumor suppressors
Tumor suppressor01 (1/0)000002 (1/1)
Oncogenes05 (2/3)001 (1/0)8 (6/2)9 (6/3)
Translocated cancer genes04 (2/2)001 (1/0)8 (6/2)
Protein kinases000010 (6/4)
Cell differentiation markers1 (1/0)0018 (14/4)
Homeodomain proteins002 (0/2)
Transcription factors033 (18/15)
Cytokines and growth factors15 (12/3)

a Genes displaying >2-fold expression changes after FLAG sorting were sorted according to shared biological features. Predicted genes were not selected in this list. The numbers of up- and down-regulated genes are presented in parentheses (up/down).

Although both control BM and myeloma-infiltrated BM are hypoxic, the hypoxic level in myelomatous BM was revealed to be much lower than that in native BM, as confirmed by the lower expression level of HIF-1α in myelomatous BM than in native BM (29). These results imply that BM hypoxia is decreased during the progression of myeloma. Interestingly, using the 5T2 multiple myeloma mouse model, native BM hypoxia induced both apoptotic cell death of CD45 MM cells and exerted tumor-initiating effects on CD45+ MM cells (29). Additionally, blocking angiogenesis in this model restores the native BM hypoxia and results in apoptosis and suppressed proliferation of the MM cells (29). Furthermore, VEGF, a key angiogenic factor and a HIF-1α-responsive gene, is induced in CD45 5T2MM cells but not in CD45+ 5T2MM cells (29). These data indicate that although BM hypoxia decreased the proliferation of CD45 MM cells, angiogenesis was induced, thus permitting outgrowth of the CD45 MM cells. Similarly, the microarray data in this study revealed that up-regulated genes were associated with both apoptosis and development. The MM U266 cells used in this study were CD45. Therefore, although hypoxia induces U266 cell death by up-regulating apoptosis-related gene expression, resistance mechanisms for hypoxia were also induced via the up-regulation of development-related genes including VEGF and CD44.

In the microarray data, oxidative stress induced growth inhibitor 1 (OKL38) was the gene most strongly up-regulated by CoCl2 treatment (fold change = 10.52). OKL38 is a tumor suppressor gene, and its overexpression inhibits tumor cell growth and induces apoptosis (33,34). Although the effect in hypoxia on OKL38 has not been researched, hypoxia-mediated cell death could be dependent on OKL38 expression. In addition, CD44 was highly up-regulated by CoCl2 in U266 cells (fold change = 5.32). CD44 is an antigen glycoprotein that plays an important role in aspects of cancer progression including growth promotion, angiogenesis, and metastasis (35); however, its cellular function in hypoxia has not yet been elucidated. Therefore, the hypoxia-induced up-regulation of CD44 in U266 cells indicates that CD44 is a novel key protein controlling hypoxia resistance mechanisms including angiogenesis in MM cells.

In conclusion, we first examined the diversity of gene expression responses to hypoxia in MM cells and found significant differences in gene expression. Additionally, although hypoxia induced apoptosis, the gene expression patterns indicated the activation of both antitumor and hypoxia resistance processes. Therefore, our results may provide a useful approach to understanding cellular responses to hypoxia.

Acknowledgements

We are grateful to all members of our research group for their support and advice regarding this study. This study resulted from the Konkuk University Research Support Program.

References

1. 

X LuY KangHypoxia and hypoxia-inducible factors: master regulators of metastasisClin Cancer Res1659285935201010.1158/1078-0432.CCR-10-136020962028

2. 

AL HarrisHypoxia, a key regulatory factor in tumour growthNat Rev Cancer23847200210.1038/nrc70411902584

3. 

GL SemenzaTargeting HIF-1 for cancer therapyNat Rev Cancer3721732200310.1038/nrc1187

4. 

R SullivanCH GrahamHypoxia-driven selection of the metastatic phenotypeCancer Metastasis Rev26319331200710.1007/s10555-007-9062-217458507

5. 

GP GuptaDX NguyenAC ChiangMediators of vascular remodelling co-opted for sequential steps in lung metastasisNature446765770200710.1038/nature0576017429393

6. 

KG ShyuFL HsuMJ WangBW WangS LinHypoxia-inducible factor 1alpha regulates lung adenocarcinoma cell invasionExp Cell Res31311811191200710.1016/j.yexcr.2007.01.01317335808

7. 

T ImaiA HoriuchiC WangHypoxia attenuates the expression of E-cadherin via up-regulation of SNAIL in ovarian carcinoma cellsAm J Pathol16314371447200310.1016/S0002-9440(10)63501-814507651

8. 

MH YangMZ WuSH ChiouDirect regulation of TWIST by HIF-1alpha promotes metastasisNat Cell Biol10295305200810.1038/ncb169118297062

9. 

B KrishnamacharyD ZagzagH NagasawaHypoxiainducible factor-1-dependent repression of E-cadherin in von Hippel-Lindau tumor suppressor-null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1BCancer Res6627252731200610.1158/0008-5472.CAN-05-371916510593

10. 

M KoshijiY KageyamaEA PeteI HorikawaJC BarrettLE HuangHIF-1alpha induces cell cycle arrest by functionally counteracting MycEMBO J2319491956200410.1038/sj.emboj.760019615071503

11. 

WG AnM KanekalMC SimonE MaltepeMV BlagosklonnyLM NeckersStabilization of wild-type p53 by hypoxiainducible factor 1alphaNature392405408199810.1038/329259537326

12. 

K GuoG SearfossD KrolikowskiHypoxia induces the expression of the pro-apoptotic gene BNIP3Cell Death Differ8367376200110.1038/sj.cdd.440081011550088

13. 

Y HuangKM DuZH XueCobalt chloride and low oxygen tension trigger differentiation of acute myeloid leukemic cells: possible mediation of hypoxia-inducible factor-1alphaLeukemia1720652073200310.1038/sj.leu.2403141

14. 

JY JungWJ KimInvolvement of mitochondrial- and Fas-mediated dual mechanism in CoCl2-induced apoptosis of rat PC12 cellsNeurosci Lett3718590200410.1016/j.neulet.2004.06.06915519734

15. 

Z JiG YangS ShahzidiInduction of hypoxia-inducible factor-1alpha overexpression by cobalt chloride enhances cellular resistance to photodynamic therapyCancer Lett244182189200610.1016/j.canlet.2005.12.01016427735

16. 

SG LeeH LeeHM RhoTranscriptional repression of the human p53 gene by cobalt chloride mimicking hypoxiaFEBS Lett507259263200110.1016/S0014-5793(01)02989-111696352

17. 

A VengellurBG WoodsHE RyanRS JohnsonJJ LaPresGene expression profiling of the hypoxia signaling pathway in hypoxia-inducible factor 1alpha null mouse embryonic fibroblastsGene Expr11181197200310.3727/00000000310874906214686790

18. 

SJ YangJ PyenI LeeH LeeY KimT KimCobalt chloride-induced apoptosis and extracellular signal-regulated protein kinase 1/2 activation in rat C6 glioma cellsJ Biochem Mol Biol37480486200410.5483/BMBRep.2004.37.4.48015469737

19. 

J ArayaM MaruyamaA InoueInhibition of proteasome activity is involved in cobalt-induced apoptosis of human alveolar macrophagesAm J Physiol Lung Cell Mol Physiol283L849858200210.1152/ajplung.00422.200112225962

20. 

W ZouM YanW XuCobalt chloride induces PC12 cells apoptosis through reactive oxygen species and accompanied by AP-1 activationJ Neurosci Res64646653200110.1002/jnr.111811398189

21. 

HJ KimSJ YangYS KimTU KimCobalt chloride-induced apoptosis and extracellular signal-regulated protein kinase activation in human cervical cancer HeLa cellsJ Biochem Mol Biol36468474200310.5483/BMBRep.2003.36.5.46814536030

22. 

JP PiretD MottetM RaesC MichielsCoCl2, a chemical inducer of hypoxia-inducible factor-1, and hypoxia reduce apoptotic cell death in hepatoma cell line HepG2Ann NY Acad Sci973443447200212485908

23. 

XH LiuA KirschenbaumS YaoUp-regulation of vascular endothelial growth factor by cobalt chloride-simulated hypoxia is mediated by persistent induction of cyclooxygenase-2 in a metastatic human prostate cancer cell lineClin Exp Metastasis17687694199910.1023/A:1006728119549

24. 

T Van LieshoutJ StaniszV EspirituM RichardsonG SinghA hypoxic response induced in MatLyLu cells by cobalt chloride results in an enhanced angiogenic response by the chick chorioallantoic membraneInt J Oncol237457502003

25. 

N LuH ZhouYH LinZQ ChenY PanXJ LiOxidative stress mediates CoCl(2)-induced prostate tumour cell adhesion: role of protein kinase C and p38 mitogen-activated protein kinaseBasic Clin Pharmacol Toxicol1014146200710.1111/j.1742-7843.2007.00074.x17577315

26. 

MH RyuJH ParkJE ParkJ ChungCH LeeHR ParkCobalt chloride stimulates phosphoinositide 3-kinase/Akt signaling through the epidermal growth factor receptor in oral squamous cell carcinomaBiocell341521201020506627

27. 

RA KyleSV RajkumarMultiple myelomaBlood11129622972200810.1182/blood-2007-10-07802218332230

28. 

K ParmarP MauchJA VergilioR SacksteinJD DownDistribution of hematopoietic stem cells in the bone marrow according to regional hypoxiaProc Natl Acad Sci USA10454315436200710.1073/pnas.070115210417374716

29. 

K AsosinghH De RaeveM de RidderRole of the hypoxic bone marrow microenvironment in 5T2MM murine myeloma tumor progressionHaematologica90810817200515951294

30. 

J HuDR HandisidesE Van ValckenborghTargeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrugBlood11615241527201010.1182/blood-2010-02-26912620530289

31. 

SY KimS BaeKH ChoiS AnHydrogen peroxide controls Akt activity via ubiquitination/degradationOncol Rep2615611566201121874266

32. 

JT ChiZ WangDS NuytenGene expression programs in response to hypoxia: cell type specificity and prognostic significance in human cancersPLoS Med3e47200610.1371/journal.pmed.003004716417408

33. 

T WangD XiaN LiBone marrow stromal cell-derived growth inhibitor inhibits growth and migration of breast cancer cells via induction of cell cycle arrest and apoptosisJ Biol Chem28043744382200510.1074/jbc.M40870820015569677

34. 

H HuynhCY NgCK OngKB LimTW ChanCloning and characterization of a novel pregnancy-induced growth inhibitor in mammary glandEndocrinology14236073615200110.1210/endo.142.8.829711459809

35. 

R MarhabaM ZollerCD44 in cancer progression: adhesion, migration and growth regulationJ Mol Histol35211231200410.1023/B:HIJO.0000032354.94213.6915339042

Related Articles

Journal Cover

November 2012
Volume 30 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bae S, Jeong H, Cha HJ, Kim K, Choi YM, An I, Koh HJ, Lim DJ, Lee S, An S, An S, et al: The hypoxia-mimetic agent cobalt chloride induces cell cycle arrest and alters gene expression in U266 multiple myeloma cells. Int J Mol Med 30: 1180-1186, 2012
APA
Bae, S., Jeong, H., Cha, H.J., Kim, K., Choi, Y.M., An, I. ... An, S. (2012). The hypoxia-mimetic agent cobalt chloride induces cell cycle arrest and alters gene expression in U266 multiple myeloma cells. International Journal of Molecular Medicine, 30, 1180-1186. https://doi.org/10.3892/ijmm.2012.1115
MLA
Bae, S., Jeong, H., Cha, H. J., Kim, K., Choi, Y. M., An, I., Koh, H. J., Lim, D. J., Lee, S., An, S."The hypoxia-mimetic agent cobalt chloride induces cell cycle arrest and alters gene expression in U266 multiple myeloma cells". International Journal of Molecular Medicine 30.5 (2012): 1180-1186.
Chicago
Bae, S., Jeong, H., Cha, H. J., Kim, K., Choi, Y. M., An, I., Koh, H. J., Lim, D. J., Lee, S., An, S."The hypoxia-mimetic agent cobalt chloride induces cell cycle arrest and alters gene expression in U266 multiple myeloma cells". International Journal of Molecular Medicine 30, no. 5 (2012): 1180-1186. https://doi.org/10.3892/ijmm.2012.1115