Macrophage migration inhibitory factor promotes Warburg effect via activation of the NF‑κB/HIF‑1α pathway in lung cancer

  • Authors:
    • Jun Li
    • Junhua Zhang
    • Fengjiao Xie
    • Jiangzhou Peng
    • Xu Wu
  • View Affiliations

  • Published online on: November 22, 2017     https://doi.org/10.3892/ijmm.2017.3277
  • Pages: 1062-1068
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Macrophage migration inhibitory factor (MIF) is upregulated in various solid tumors, a process that is associated with tumor progression and metastasis. The present study aimed to investigate the role and the underlying mechanism of MIF in human lung cancer. Human lung cancer H358, H460, H524, H1650, H838, H1975 and A549 cell lines were used to examine the expression of MIF by real time‑quantitative polymerase chain reaction and western blotting. The lentivirus was used to overexpress MIF and the expression of MIF and hypoxia‑inducible factor 1‑α (HIF‑1α) were knocked down by shRNA or siRNA. The proliferation of cell lines was detected by MTT assay. Glucose uptake, adenosine 5'‑triphosphate (ATP) production, the glycolytic rate and lactate production were used to examine the Warburg effect in cells. BAY 11‑7082 (BAY) was used to inhibit the nuclear translocation of nuclear factor‑κB (NF‑κB), which was detected using immunofluorescence. It was revealed that overexpression of MIF promoted cell proliferation and the Warburg effect in lung cancer, whereas knockdown of MIF inhibited cell proliferation and the Warburg effect. Mechanistically, MIF promoted the Warburg effect by upregulating HIF‑1α. Knockdown of HIF‑1α largely abolished the promotional effect of MIF on the Warburg effect. Additionally, the results in the current study provided evidence that MIF regulates HIF‑1α through NF‑κB. In conclusion, the findings of the present study demonstrated that MIF is a key component in lung cancer progression through promoting the Warburg effect, and that the novel MIF/NF‑κB/HIF‑1α axis may prove to be useful for the development of new strategies for treating patients with lung cancer.

Introduction

Lung cancer is the most common cancer and the leading cause of cancer-associated mortality in the world (1). It is estimated that the 5-year survival rate is <16% (1,2). However, the underlying molecular mechanisms of lung cancer pathogenesis are poorly understood. Therefore, novel therapeutic targets for human lung cancer are urgently warranted.

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine pro duced by a variety of cell types (35), which has been involved in numerous inflammatory diseases, including rheumatoid arthritis (6), atherosclerosis (7) and hepatitis (8). In addition, MIF is overexpressed in various solid tumors (911), and regulates autonomous properties of tumor cells, including proliferation, apoptosis, and chemoresistance. Furthermore, MIF is involved in the metastatic potential of tumors, including hepatoma, gastric cancer and lung cancer (1215). Overwhelming evidence has been produced to highlight the essential role of MIF in tumor progression and tumorigenesis. MIF promotes sustained extracellular signal-regulated kinase activation, which is a major outcome of mutations in Ras, occurring in a third of human tumors (16,17). MIF signal transduction is necessary for maximal tumor-associated evasion from cell senescence (18) and inhibition of the tumor suppressor, p53 (19,20). MIF has been shown to support hypoxic adaptation by inducing stabilization of hypoxia-inducible factor-1α (HIF-1α) (21). In a hypoxic environment, MIF may also promote tumor growth through a marked enhancement of angiogenesis (16,21). Recently, an expanding body of studies on lung cancer have identified MIF as a key factor of tumor growth. MIF has also been identified in high quantities in the non-small cell lung cancer cells that subsequently serve to promote neovascularization (22,23). MIF overexpression promoted lung tumor growth and progression (24), whereas specific knockdown of MIF expression or functional inhibition significantly reduced migration and invasion of lung adenocarcinoma cells (25,26). The present study will provide persuasive evidence that MIF is a promising therapeutic target in human lung cancer.

The majority of cancer cells preferentially use aerobic glycolysis instead of oxidative phosphorylation to meet their increased energetic and biosynthetic demands, despite the presence of oxygen, which has long been known as the Warburg effect (27,28). The Warburg effect serves a critical role in tumorigenesis, and can be targeted for cancer therapy (29) MIF has been shown to exert an important role in the control of glucose disposal and carbohydrate metabolism (30,31). However, the role of MIF on the Warburg effect in lung cancer has yet to be elucidated.

In the present study, it is demonstrated that MIF promotes cell proliferation and the Warburg effect in lung cancer. The study also demonstrates that blocking the nuclear factor-κB (NF-κB)/HIF-1α signaling pathway largely abolishes the promotional effects of MIF on the Warburg effect. This novel MIF/NF-κB/HIF-1α axis may be valuable in the development of novel strategies for treating patients with lung cancer.

Materials and methods

Cells and reagents

Procedures concerning cell culture were performed as previously described (32). Seven human lung cancer cell lines (H358, H460, H524, H1650, H838, H1975 and A549) were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured according to their protocol. All the cell lines used in the present study were authenticated through short-tandem repeat profiling less than 6 months prior to the initiation of this project, and the cells had not been in culture for more than 2 months.

RNA interference

These procedures were also performed as previously described (32). Lentiviral plasmids expressing MIF, short hairpin (sh)MIF and negative control were purchased from Cyagen Biosciences, Inc. (Shanghai, China). The specific sequences against MIF were as follows: 5′-TCATCGTAAACACCAACGT-3′ and 5′-GCGCAGAACCGCTCCTACA-3′). Short interfering RNAs (siRNAs) against HIF-1α (the specific sequences, 5′-AGUUAGUUCAAACUGAGUUAAUCCC-3′) were purchased from Ruibo Biotech Co., Ltd. (Guangzhou, China). Scrambled siRNA (Ruibo Biotech Co., Ltd.) was used as a non-specific siRNA control. Approximately 2×105 cells/well were seeded in a 60-mm culture dish on the day prior to transfection. Transfection with 50 nmol siRNA was performed according to the manufacturer's protocol using the Lipofectamine™ RNAiMAX transfection reagent (Thermo Fisher Scientific, Inc., Waltham, MA, USA). After transfection for 48 h in a incubator with 5% CO2 at 37°C, real time-quantitative polymerase chain reaction (RT-qPCR) and western blot assays were performed.

RNA extraction and RT-qPCR

Total RNA was extracted from cells and isolated using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. First-strand cDNA was synthesized from 1 µg total RNA using a RevertAid™ First Strand cDNA Synthesis kit (MBI Fermentas, Burlington, ON, Canada). The RT-qPCR was performed using SYBR-Green Master mix (LightCycler 480; Roche, Basel, Switzerland). The primers used in the present study were as follows: MIF forward, 5′-ACTAAGAAAGACCCGAGGC-3′ and reverse, 5′-GGGGCACGTTGGTGTTTAC-3′; GAPDH forward, 5′-AGCCACATCGCTCAGACA-3′ and reverse, 5′-TGGACTCCACGACGTACT-3′. The reaction conditions included the denaturation at 95°C for 15 sec, renaturation at 60°C for 60 sec, with 40 cycles in total. The results were quantified with 2−ΔΔCT.

Western blot analysis

Total proteins were obtained from the cultured cells. The bicinchoninic acid (BCA) method was used to determine the protein concentrations. Equal amounts of samples (4 samples per group) were loaded onto 10% SDS-PAGE gels and the proteins were transferred to polyvinyl difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% skimmed milk at room temperature for 1 h, and then incubated overnight at 4°C with the primary antibodies [rabbit monoclonal antibody against MIF (1:1,000, ab175189); mouse monoclonal antibody against HIF-1α (1:1,000, ab16066) (both from Abcam, Cambridge, MA, USA); rabbit monoclonal antibody against NF-κB (1:1,000, #8242); mouse monoclonal antibody against β-actin (1:1,000, #3700) (both from Cell Signaling Technology, Inc., Danvers, MA, USA)]. The blots were detected using a SuperSignal ECL kit (Invitrogen; Thermo Fisher Scientific, Inc.) in a western blotting detection system (Kodak Digital Science, Rochester, NY, USA) and quantified by densitometry using ImageJ analysis software (National Institutes of Health, Bethesda, MD, USA).

Cell proliferation

For the cell counting kit-8 (CCK-8) assay, ~5×103 cells/well were seeded into 96-well plates. CCK-8 solution (10 µl) was added to each well at the indicated time-points of 1, 3, 5 and 7 days post-transfection, and the mixture was incubated for 2 h at 37°C. Subsequently, the absorbance was measured at 450 nm using a spectrophotometer.

Analysis of glucose uptake, the glycolytic rate and lactate production in cells

These procedures were performed as previously described (32). Glucose uptake levels were determined by measuring the uptake of [3H]2-deoxyglucose. Briefly, cells cultured in 12-well plates were pre-incubated in glucose-free media for 30 min prior to the addition of [3H]2-deoxyglucose (1 µCi/well) to the cells. After incubation for 30 min, the cells were washed with phosphate-buffered saline (PBS) and lysed in 1% SDS. The radioactivity of the cell lysates was determined in a liquid scintillation counter, and normalized to the protein concentrations of the cell lysates. Cellular glycolytic rates were measured by monitoring the conversion of [5-3H]glucose into 3H2O. Briefly, the cells (1×106) were collected and washed once in PBS prior to resuspension in 1 ml Krebs buffer without glucose for 30 min at 37°C. The cells were collected and resuspended in 0.5 ml Krebs buffer containing 10 mM glucose and 5 µCi [5-3H]glucose for 1 h at 37°C. Triplicate 100 µl aliquots were transferred to uncapped PCR tubes containing 100 µl 0.2 N HCl, and the tubes were transferred to scintillation vials containing 0.5 ml H2O. The scintillation vials were then sealed and left for 48 h. The quantities of diffused and undiffused 3H were subsequently determined in a liquid scintillation counter. Lactate production levels were measured using a Lactate Fluorimetric/Colorimetric Assay kit (BioVision, Inc., Milpitas, CA, USA). Cells were plated in 100-mm culture dishes at a density of 1×106 cells/plate. After incubation for 24 h at 37°C, the culture medium was replaced with fetal bovine serum (FBS)-free DMEM. The lactate levels in the culture medium were examined with the Lactate Fluorimetric/Colorimetric Assay kit, and normalized against the number of cells.

Adenosine 5′-triphosphate (ATP) measurements

ATP measurements were obtained using an ATP determination kit (Beyotime Institute of Biotechnology, Shanghai, China) according to the manufacturer's protocol. The ATP content was determined based on comparison with a concurrent standard curve.

Immunofluorescence

The cells were fixed using 4% paraformaldehyde for 15 min. After washing and permeabilization with 0.1% Triton X-100 for 5 min, the cells were blocked with a 5% solution of BSA (for 1 h at room temperature). Primary antibodies of NF-κB (rabbit monoclonal antibody, 1:200, $8242; Cell Signaling Technology, Inc.) were applied overnight at 4°C. The corresponding secondary antibodies [goat anti-rabbit IgG (H+L), Alexa Fluor Plus 647 (#A32733; Invitrogen; Thermo Fisher Scientific, Inc.)] were added for a further incubation for 30 min at room temperature. To visualize the nuclei, fixed cells were incubated with 4′,6-diamidino-2-phenylindole for 10 min.

Statistical analysis

Data are expressed as the means ± standard deviation of at least three separate experiments. A one-way ANOVA and Student-Newman-Keuls tests were used for statistical analysis, and a value of P<0.05 was considered to indicate a statistically significant difference. All analyses were performed using SPSS version 13.0 (Statistical Software for Social Sciences, Chicago, IL, USA).

Results

MIF promotes lung cancer cell proliferation

First, the expression of MIF in seven lung cancer cell lines was detected using RT-qPCR and western blotting (Fig. 1A and B). High expression of MIF was observed in the H358 and A549 cell lines, whereas low expression of MIF was found in the H524 and H460 cell lines (Fig. 1A and B). Based on the expression level of MIF in these lung cancer cells, stable cell lines were constructed either with silenced MIF in H358 cells, or ectopic expression of MIF in H524 cells (Fig. 1C). Subsequently, the effect of MIF on the biological behaviors of these cell lines was investigated. Cell proliferation assays indicated that silencing MIF significantly inhibited cell proliferation (Fig. 1D); by contrast, the ectopic expression of MIF increased the cell proliferative ability of H524 cells (Fig. 1E).

MIF promotes the Warburg effect in lung cancer

The majority of cancer cells preferentially use aerobic glycolysis instead of oxidative phosphorylation to meet their increased energetic and biosynthetic demands, a phenomenon known as the Warburg effect (27). The Warburg effect is involved in tumor development, and is able to fulfill the energetic demands of the membrane transport activities required for proliferation and migration (33). Since MIF has previously been reported to regulate glucose uptake and catabolism (30,31), the issue of whether MIF may regulate the Warburg effect in lung cancer cells was investigated. Lactate dehydrogenase A (LDHA) is required for the conversion of pyruvate into lactate, which is associated with the Warburg effect. As predicted, overexpression or knockdown of MIF increased or decreased LDHA activation (Fig. 2A). Furthermore, overexpression of MIF significantly promoted the Warburg effect in H524 cells, whereas knockdown of MIF significantly inhibited the Warburg effect in H358 cells (Fig. 2B–E). These findings indicated that MIF promotes the Warburg effect in lung cancer cells.

MIF promotes the Warburg effect through activation of HIF-1α

How MIF regulates the Warburg effect in lung cancer was subsequently investigated. HIF-1α-induced transcriptional regulation controls several key genes that are crucial to deregulated glucose metabolism in cancers; that is, the Warburg effect (34). Furthermore, MIF has been shown to support hypoxic adaptation by inducing stabilization of HIF-1α (21). Consequently, the aim was to determine the association between MIF and HIF-1α on the Warburg effect in lung cancer. Knockdown of MIF decreased the protein expression of HIF-1α in H358 cells, whereas ectopic expression of MIF significantly upregulated HIF-1α in H524 cells (Fig. 3A). H358 cells were transfected with non-targeting negative control RNA and HIF-1α-specific siRNA. The specificity of siRNA was examined with western blotting, which confirmed that the siHIF-1α reduced the expression of HIF-1α (Fig. 3B). In addition, knockdown of HIF-1α expression significantly inhibited HIF-1α upregulation induced by MIF overexpression in H524 cells (Fig. 3B). Subsequently, it was shown that knockdown of HIF-1α expression significantly inhibited cell proliferation, LDHA activation and the Warburg effect in H358 cells (Fig. 3C-H). By contrast, MIF overexpression promoted cell proliferation, LDHA activation, and the Warburg effect in H524 cells, and silencing HIF-1α clearly abolished the promotional effect of MIF on cell proliferation, LDHA activation, and the Warburg effect (Fig. 3I–N). Taken together, these results strongly suggested that MIF promotes the Warburg effect through activation of HIF-1α.

MIF regulates HIF-1α through activation of NF-κB

Subsequently, the molecular mechanisms underlying MIF regulation of the Warburg effect were investigated. NF-κB and its associated signaling pathway, which is frequently activated in various types of human cancers, serves a pivotal role in tumorigenesis (27). The activated NF-κB signaling pathway has been shown to promote the Warburg effect in cancer cells (35). In the present study, H358 cells were treated with BAY 11-7082 (BAY; an NF-κB inhibitor) to block NF-κB activation. Nuclear translocation of NF-κB was significantly reduced, a result that was confirmed by western blotting and immunofluorescence (Fig. 4A and B). Ectopic expression of MIF significantly increased the translocation of NF-κB to the nucleus in H524 cells, whereas BAY largely inhibited the translocation of NF-κB to the nucleus induced by MIF overexpression, as determined by western blotting and the immunofluorescence experiments (Fig. 4A and B). Blocking NF-κB activation with BAY significantly reduced the expression of HIF-1α in H358 cells (Fig. 4C). MIF overexpression upregulated HIF-1α expression in H524 cells, whereas blocking NF-κB activation with BAY clearly inhibited the HIF-1α upregulation induced by MIF (Fig. 4C). These data suggested that MIF regulates HIF-1α through activation of NF-κB.

Discussion

An improved understanding of the mechanism underlying human lung cancer and novel therapeutic targets is urgently required for this deadly cancer. In the present study, it has been demonstrated that MIF is a key player in lung cancer progression via promoting the Warburg effect. It was also shown that MIF regulates the Warburg effect through the NF-κB/HIF-1α pathway. This novel MIF/NF-κB/HIF-1α axis may be useful for development of novel strategies for treating patients with lung cancer.

MIF, a pro-inflammatory cytokine, has been highlighted as a key factor in lung cancer. High expression of MIF in patients with lung cancer has been shown to lead to a worse prognosis (23,36), and it has been suggested as a potential biomarker in non-small cell lung cancer (37). It has been reported that MIF overexpression directly enhanced lung cancer growth and metastasis (24). In addition, functional inhibition of MIF in lung cancer cell lines suppressed cell proliferation and numerous hallmarks of tumor development, including angiogenesis and metastasis (25,26,38). In agreement with a previous study on cell proliferation (24), the present study has confirmed that MIF overexpression promotes cell proliferation, whereas silencing MIF significantly inhibited proliferation in human lung cancer cells. Our data indicate that MIF promotes cell proliferation and tumor growth in lung cancer.

The Warburg effect is one of the characteristics of cancer cells (28,39). The switch from oxidative phosphorylation to glycolysis provides an essential mechanism that enables cancer cells to meet their significantly increased energetic and biosynthetic demands to support their rapid growth and proliferation (27). Understanding the mechanism underlying the Warburg effect is important for the development of novel strategies for cancer therapy. MIF has been reported to exert an important role in the control of glucose disposal and carbohydrate metabolism (30,31). Brock et al (40) revealed that MIF promotes glucose uptake and ATP generation in non-small cell lung carcinomas. Consistent with these discoveries (27,40), our group has observed that overexpression of MIF significantly promotes the Warburg effect in H524 cells, whereas knockdown of MIF significantly inhibited the Warburg effect in H358 cells. The results of the present study suggest that MIF promotes the Warburg effect in lung cancer, indicating that MIF inhibition represents a potentially important therapeutic strategy for the treatment of human lung cancer.

How does MIF regulate the Warburg effect in lung cancer? It is essential to understand its mechanism. HIF-1 activation has been associated with angiogenesis, erythropoiesis and modulation of key enzymes involved in aerobic glycolysis, thereby modulating key processes required for the Warburg effect (41). The Warburg effect is due, at least in part, to the failure of cancer cells to appropriately downregulate HIF under well-oxygenated conditions (42). Furthermore, MIF has been shown to support hypoxic adaptation by inducing stabilization of HIF-1α (21). The results in the current study have provided evidence that MIF regulates the Warburg effect through activation of HIF-1α. NF-κB and its associated signaling pathway, which is frequently activated in various types of human cancers, exerts a pivotal role in tumorigenesis (27). Activated NF-κB signaling has been shown to promote the Warburg effect in cancer cells (35). MIF has been reported to activate NF-κB to promote breast cancer metastasis (12). Furthermore, emerging evidence indicates cross-talk between the HIF and NF-κB pathways. For example, inflammatory stimuli, such as tumor necrosis factor-α, activate HIF-1α in an NF-κB-dependent manner (43). Furthermore, NF-κB increases HIF-1β stabilization (44), suggesting a potential role of NF-κB in regulation of the HIF pathways. The results in the present study have demonstrated that MIF regulates HIF-1α through activation of NF-κB in lung cancer. Taken together, these results suggest that MIF regulates the Warburg effect through activation of the NF-κB/HIF-1α pathway.

In conclusion, the findings of the current study have demonstrated that MIF promotes the Warburg effect through the NF-κB/HIF-1α pathway in lung cancer. These results contribute towards an improved understanding of the role of MIF in human lung cancer, and provide evidence that MIF inhibition may be therapeutically beneficial. However, additional studies are required; for example, a further assessment of the functions of MIF is required to investigate its biological significance in lung cancer in vivo. Mechanistic details underlying the promotion of the Warburg effect by MIF in lung cancer also require further characterization.

Acknowledgments

This study was supported by the Foundation of the President of Nanfang Hospital, Southern Medical University, Guangzhou, China (grant no. 2013B016)

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Herbst RS, Heymach JV and Lippman SM: Lung cancer. N Engl J Med. 359:1367–1380. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Daryadel A, Grifone RF, Simon HU and Yousefi S: Apoptotic neutrophils release macrophage migration inhibitory factor upon stimulation with tumor necrosis factor-alpha. J Biol Chem. 281:27653–27661. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Shimizu T: Role of macrophage migration inhibitory factor (MIF) in the skin. J Dermatol Sci. 37:65–73. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Verschuren L, Lindeman JH, van Bockel JH, Abdul-Hussien H, Kooistra T and Kleemann R: Upregulation and coexpression of MIF and matrix metalloproteinases in human abdominal aortic aneurysms. Antioxid Redox Signal. 7:1195–1202. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Morand EF, Leech M and Bernhagen J: MIF: A new cytokine link between rheumatoid arthritis and atherosclerosis. Nat Rev Drug Discov. 5:399–410. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Chen D, Xia M, Hayford C, Tham L, Semik V, Hurst S, Chen Y, Tam HH, Pan J, Wang Y, et al: Expression of human tissue factor pathway inhibitor on vascular smooth muscle cells inhibits secretion of macrophage migration inhibitory factor and attenuates atherosclerosis in ApoE−/− mice. Circulation. 131:1350–1360. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Assis DN, Leng L, Du X, Zhang CK, Grieb G, Merk M, Garcia AB, McCrann C, Chapiro J, Meinhardt A, et al: The role of macrophage migration inhibitory factor in autoimmune liver disease. Hepatology. 59:580–591. 2014. View Article : Google Scholar

9 

Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K and Muschel R: Macrophage migration inhibitory factor: A key cytokine and therapeutic target in colon cancer. Cytokine Growth Factor Rev. 26:451–461. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Tas F, Karabulut S, Serilmez M, Ciftci R and Duranyildiz D: Serum levels of macrophage migration-inhibitory factor (MIF) have diagnostic, predictive and prognostic roles in epithelial ovarian cancer patients. Tumour Biol. 35:3327–3331. 2014. View Article : Google Scholar

11 

Meyer-Siegler KL, Vera PL, Iczkowski KA, Bifulco C, Lee A, Gregersen PK, Leng L and Bucala R: Macrophage migration inhibitory factor (MIF) gene polymorphisms are associated with increased prostate cancer incidence. Genes Immun. 8:646–652. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Lv W, Chen N, Lin Y, Ma H, Ruan Y, Li Z, Li X, Pan X and Tian X: Macrophage migration inhibitory factor promotes breast cancer metastasis via activation of HMGB1/TLR4/NF kappa B axis. Cancer Lett. 375:245–255. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Zhao YM, Wang L, Dai Z, Wang DD, Hei ZY, Zhang N, Fu XT, Wang XL, Zhang SC, Qin LX, et al: Validity of plasma macrophage migration inhibitory factor for diagnosis and prognosis of hepatocellular carcinoma. Int J Cancer. 129:2463–2472. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Morris KT, Nofchissey RA, Pinchuk IV and Beswick EJ: Chronic macrophage migration inhibitory factor exposure induces mesenchymal epithelial transition and promotes gastric and colon cancers. PLoS One. 9:e986562014. View Article : Google Scholar : PubMed/NCBI

15 

Cates JM, Friedman DB, Seeley EH, Dupont WD, Schwartz HS, Holt GE, Caprioli RM and Young PP: Proteomic analysis of osteogenic sarcoma: Association of tumour necrosis factor with poor prognosis. Int J Exp Pathol. 91:335–349. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Xu L, Li Y, Sun H, Zhen X, Qiao C, Tian S and Hou T: Current developments of macrophage migration inhibitory factor (MIF) inhibitors. Drug Discov Today. 18:592–600. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Kyriakis JM: Thinking outside the box about Ras. J Biol Chem. 284:10993–10994. 2009. View Article : Google Scholar :

18 

Welford SM, Bedogni B, Gradin K, Poellinger L, Broome Powell M and Giaccia AJ: HIF1alpha delays premature senescence through the activation of MIF. Genes Dev. 20:3366–3371. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Fingerle-Rowson G, Petrenko O, Metz CN, Forsthuber TG, Mitchell R, Huss R, Moll U, Müller W and Bucala R: The p53-dependent effects of macrophage migration inhibitory factor revealed by gene targeting. Proc Natl Acad Sci USA. 100:9354–9359. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Martin J, Duncan FJ, Keiser T, Shin S, Kusewitt DF, Oberyszyn T, Satoskar AR and VanBuskirk AM: Macrophage migration inhibitory factor (MIF) plays a critical role in pathogenesis of ultraviolet-B (UVB)-induced nonmelanoma skin cancer (NMSC). FASEB J. 23:720–730. 2009. View Article : Google Scholar

21 

Winner M, Koong AC, Rendon BE, Zundel W and Mitchell RA: Amplification of tumor hypoxic responses by macrophage migration inhibitory factor-dependent hypoxia-inducible factor stabilization. Cancer Res. 67:186–193. 2007. View Article : Google Scholar : PubMed/NCBI

22 

White ES, Strom SR, Wys NL and Arenberg DA: Non-small cell lung cancer cells induce monocytes to increase expression of angiogenic activity. J Immunol. 166:7549–7555. 2001. View Article : Google Scholar : PubMed/NCBI

23 

White ES, Flaherty KR, Carskadon S, Brant A, Iannettoni MD, Yee J, Orringer MB and Arenberg DA: Macrophage migration inhibitory factor and CXC chemokine expression in non-small cell lung cancer: Role in angiogenesis and prognosis. Clin Cancer Res. 9:853–860. 2003.PubMed/NCBI

24 

Arenberg D, Luckhardt TR, Carskadon S, Zhao L, Amin MA and Koch AE: Macrophage migration inhibitory factor promotes tumor growth in the context of lung injury and repair. Am J Respir Crit Care Med. 182:1030–1037. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Mawhinney L, Armstrong ME, O' Reilly C, Bucala R, Leng L, Fingerle-Rowson G, Fayne D, Keane MP, Tynan A, Maher L, et al: Macrophage migration inhibitory factor (MIF) enzymatic activity and lung cancer. Mol Med. 20:729–735. 2015.PubMed/NCBI

26 

Rendon BE, Roger T, Teneng I, Zhao M, Al-Abed Y, Calandra T and Mitchell RA: Regulation of human lung adenocarcinoma cell migration and invasion by macrophage migration inhibitory factor. J Biol Chem. 282:29910–29918. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Liu J, Zhang C, Wu R, Lin M, Liang Y, Liu J, Wang X, Yang B and Feng Z: RRAD inhibits the Warburg effect through negative regulation of the NF-κB signaling. Oncotarget. 6:14982–14992. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Iansante V, Choy PM, Fung SW, Liu Y, Chai JG, Dyson J, Del Rio A, D'Santos C, Williams R, Chokshi S, et al: PARP14 promotes the Warburg effect in hepatocellular carcinoma by inhibiting JNK1-dependent PKM2 phosphorylation and activation. Nat Commun. 6:78822015. View Article : Google Scholar : PubMed/NCBI

30 

Benigni F, Atsumi T, Calandra T, Metz C, Echtenacher B, Peng T and Bucala R: The proinflammatory mediator macrophage migration inhibitory factor induces glucose catabolism in muscle. J Clin Invest. 106:1291–1300. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Waeber G, Calandra T, Roduit R, Haefliger JA, Bonny C, Thompson N, Thorens B, Temler E, Meinhardt A, Bacher M, et al: Insulin secretion is regulated by the glucose-dependent production of islet beta cell macrophage migration inhibitory factor. Proc Natl Acad Sci USA. 94:4782–4787. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Lv XB, Liu L, Cheng C, Yu B, Xiong L, Hu K, Tang J, Zeng L and Sang Y: SUN2 exerts tumor suppressor functions by suppressing the Warburg effect in lung cancer. Sci Rep. 5:179402015. View Article : Google Scholar : PubMed/NCBI

33 

Cairns RA: Drivers of the Warburg phenotype. Cancer J. 21:56–61. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Jia YY, Zhao JY, Li BL, Gao K, Song Y, Liu MY, Yang XJ, Xue Y, Wen AD and Shi L: miR-592/WSB1/HIF-1α axis inhibits glycolytic metabolism to decrease hepatocellular carcinoma growth. Oncotarget. 7:35257–35269. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Johnson RF and Perkins ND: Nuclear factor-κB, p53, and mitochondria: Regulation of cellular metabolism and the Warburg effect. Trends Biochem Sci. 37:317–324. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Kamimura A, Kamachi M, Nishihira J, Ogura S, Isobe H, Dosaka-Akita H, Ogata A, Shindoh M, Ohbuchi T and Kawakami Y: Intracellular distribution of macrophage migration inhibitory factor predicts the prognosis of patients with adenocarcinoma of the lung. Cancer. 89:334–341. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Gámez-Pozo A, Sánchez-Navarro I, Calvo E, Agulló-Ortuño MT, López-Vacas R, Díaz E, Camafeita E, Nistal M, Madero R, Espinosa E, et al: PTRF/cavin-1 and MIF proteins are identified as non-small cell lung cancer biomarkers by label-free proteomics. PLoS One. 7:e337522012. View Article : Google Scholar : PubMed/NCBI

38 

Guo Y, Hou J, Luo Y and Wang D: Functional disruption of macrophage migration inhibitory factor (MIF) suppresses proliferation of human H460 lung cancer cells by caspase-dependent apoptosis. Cancer Cell Int. 13:282013. View Article : Google Scholar : PubMed/NCBI

39 

Vousden KH and Ryan KM: p53 and metabolism. Nat Rev Cancer. 9:691–700. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Brock SE, Rendon BE, Yaddanapudi K and Mitchell RA: Negative regulation of AMP-activated protein kinase (AMPK) activity by macrophage migration inhibitory factor (MIF) family members in non-small cell lung carcinomas. J Biol Chem. 287:37917–37925. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Courtnay R, Ngo DC, Malik N, Ververis K, Tortorella SM and Karagiannis TC: Cancer metabolism and the Warburg effect: The role of HIF-1 and PI3K. Mol Biol Rep. 42:841–851. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Kaelin WG Jr: Cancer and altered metabolism: Potential importance of hypoxia-inducible factor and 2-oxoglutarate-dependent dioxygenases. Cold Spring Harb Symp Quant Biol. 76:335–345. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Yao G, Zhang Q, Doeppner TR, Niu F, Li Q, Yang Y, Kuckelkorn U, Hagemann N, Li W, Hermann DM, et al: LDL suppresses angiogenesis through disruption of the HIF pathway via NF-κB inhibition which is reversed by the proteasome inhibitor BSc2118. Oncotarget. 6:30251–30262. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Shin DH, Li SH, Yang SW, Lee BL, Lee MK and Park JW: Inhibitor of nuclear factor-kappaB alpha derepresses hypoxia-inducible factor-1 during moderate hypoxia by sequestering factor inhibiting hypoxia-inducible factor from hypoxia-inducible factor 1alpha. FEBS J. 276:3470–3480. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 41 Issue 2

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
Li J, Zhang J, Xie F, Peng J and Wu X: Macrophage migration inhibitory factor promotes Warburg effect via activation of the NF‑κB/HIF‑1α pathway in lung cancer. Int J Mol Med 41: 1062-1068, 2018
APA
Li, J., Zhang, J., Xie, F., Peng, J., & Wu, X. (2018). Macrophage migration inhibitory factor promotes Warburg effect via activation of the NF‑κB/HIF‑1α pathway in lung cancer. International Journal of Molecular Medicine, 41, 1062-1068. https://doi.org/10.3892/ijmm.2017.3277
MLA
Li, J., Zhang, J., Xie, F., Peng, J., Wu, X."Macrophage migration inhibitory factor promotes Warburg effect via activation of the NF‑κB/HIF‑1α pathway in lung cancer". International Journal of Molecular Medicine 41.2 (2018): 1062-1068.
Chicago
Li, J., Zhang, J., Xie, F., Peng, J., Wu, X."Macrophage migration inhibitory factor promotes Warburg effect via activation of the NF‑κB/HIF‑1α pathway in lung cancer". International Journal of Molecular Medicine 41, no. 2 (2018): 1062-1068. https://doi.org/10.3892/ijmm.2017.3277