The Role of Hypoxia-Inducible Factor Isoforms in Breast Cancer and Perspectives on Their Inhibition in Therapy
Abstract
:Simple Summary
Abstract
1. Hypoxia in Normal and Cancer Cells
2. Hypoxia-Inducible Factors: Characteristics and Functions
3. Activation of the Hypoxia-Inducible Factor in Cancers
4. Effects of the HIF Activation
4.1. Metabolism
4.2. Evasion of the Immune Response
4.3. Cell Survival and Death
4.4. Angiogenesis, Invasion, and Metastasis
4.5. Cancer Stem Cells
4.6. HIFs Activity and Hormone Receptors
5. Perspectives on HIF’s Inhibition in Breast Cancer—To Inhibit or Not to Inhibit?
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Höckel, M.; Vaupel, P. Tumor hypoxia: Definitions and current clinical, biologic, and molecular aspects. J. Natl. Cancer Inst. 2001, 93, 266–276. [Google Scholar] [CrossRef] [PubMed]
- Carreau, A.; El Hafny-Rahbi, B.; Matejuk, A.; Grillon, C.; Kieda, C. Why is the partial oxygen pressure of human tissues a crucial parameter? Small molecules and hypoxia. J. Cell. Mol. Med. 2011, 15, 1239–1253. [Google Scholar] [CrossRef] [PubMed]
- Vaupel, P.; Mayer, A. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev. 2007, 26, 225–239. [Google Scholar] [CrossRef] [PubMed]
- Shi, R.; Liao, C.; Zhang, Q. Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications. Cells 2021, 10, 678. [Google Scholar] [CrossRef]
- Semenza, G.L. Hypoxia-inducible factors in physiology and medicine. Cell 2012, 148, 399–408. [Google Scholar] [CrossRef]
- Duan, C. Hypoxia-inducible factor 3 biology: Complexities and emerging themes. Am. J. Physiol. Cell Physiol. 2016, 310, C260–C269. [Google Scholar] [CrossRef]
- Masson, N.; Ratcliffe, P.J. Hypoxia signaling pathways in cancer metabolism: The importance of co-selecting interconnected physiological pathways. Cancer Metab. 2014, 2, 3. [Google Scholar] [CrossRef]
- Saxena, K.; Jolly, M.K. Acute vs. Chronic vs. Cyclic Hypoxia: Their Differential Dynamics, Molecular Mechanisms, and Effects on Tumor Progression. Biomolecules 2019, 9, 339. [Google Scholar] [CrossRef]
- Hu, C.J.; Sataur, A.; Wang, L.; Chen, H.; Simon, M.C. The N-terminal transactivation domain confers target gene specificity of hypoxia-inducible factors HIF-1alpha and HIF-2alpha. Mol. Biol. Cell 2007, 18, 4528–4542. [Google Scholar] [CrossRef]
- Keith, B.; Simon, M.C. Hypoxia-inducible factors, stem cells, and cancer. Cell 2007, 129, 465–472. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Q.; Qiao, L.; Liu, Q.; Kong, X.; Hu, J.; Hu, W.; Wu, Z.; Li, M.; Liu, L. Hypoxia associated multi-omics molecular landscape of tumor tissue in patients with hepatocellular carcinoma. Aging 2021, 13, 6525–6553. [Google Scholar] [CrossRef]
- Holmquist-Mengelbier, L.; Fredlund, E.; Löfstedt, T.; Noguera, R.; Navarro, S.; Nilsson, H.; Pietras, A.; Vallon-Christersson, J.; Borg, A.; Gradin, K.; et al. Recruitment of HIF-1alpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype. Cancer Cell 2006, 10, 413–423. [Google Scholar] [CrossRef] [PubMed]
- Koivunen, P.; Hirsilä, M.; Günzler, V.; Kivirikko, K.I.; Myllyharju, J. Catalytic properties of the asparaginyl hydroxylase (FIH) in the oxygen sensing pathway are distinct from those of its prolyl 4-hydroxylases. J. Biol. Chem. 2004, 279, 9899–9904. [Google Scholar] [CrossRef] [PubMed]
- Koh, M.Y.; Darnay, B.G.; Powis, G. Hypoxia-associated factor, a novel E3-ubiquitin ligase, binds and ubiquitinates hypoxia-inducible factor 1alpha, leading to its oxygen-independent degradation. Mol. Cell. Biol. 2008, 28, 7081–7095. [Google Scholar] [CrossRef] [PubMed]
- de Heer, E.C.; Jalving, M.; Harris, A.L. HIFs, angiogenesis, and metabolism: Elusive enemies in breast cancer. J. Clin. Investig. 2020, 130, 5074–5087. [Google Scholar] [CrossRef]
- Maxwell, P.H.; Pugh, C.W.; Ratcliffe, P.J. Activation of the HIF pathway in cancer. Curr. Opin. Genet. Dev. 2001, 11, 293–299. [Google Scholar] [CrossRef]
- Courtnay, R.; Ngo, D.C.; Malik, N.; Ververis, K.; Tortorella, S.M.; Karagiannis, T.C. Cancer metabolism and the Warburg effect: The role of HIF-1 and PI3K. Mol. Biol. Rep. 2015, 42, 841–851. [Google Scholar] [CrossRef] [PubMed]
- Kronblad, A.; Jirström, K.; Rydén, L.; Nordenskjöld, B.; Landberg, G. Hypoxia-inducible factor-1alpha is a prognostic marker in premenopausal patients with intermediate to highly differentiated breast cancer but not a predictive marker for tamoxifen response. Int. J. Cancer 2006, 118, 2609–2616. [Google Scholar] [CrossRef]
- Choi, W.; Boland, J.; Lin, J. Hypoxia-Inducible Factor-2α as a Novel Target in Renal Cell Carcinoma. J. Kidney Cancer VHL 2021, 8, 1–7. [Google Scholar] [CrossRef]
- Lee, P.; Chandel, N.S.; Simon, M.C. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat. Rev. Mol. Cell Biol. 2020, 21, 268–283. [Google Scholar] [CrossRef]
- Iommarini, L.; Porcelli, A.M.; Gasparre, G.; Kurelac, I. Non-Canonical Mechanisms Regulating Hypoxia-Inducible Factor 1 Alpha in Cancer. Front. Oncol. 2017, 7, 286. [Google Scholar] [CrossRef] [PubMed]
- Jarman, E.J.; Ward, C.; Turnbull, A.K.; Martinez-Perez, C.; Meehan, J.; Xintaropoulou, C.; Sims, A.H.; Langdon, S.P. HER2 regulates HIF-2α and drives an increased hypoxic response in breast cancer. Breast Cancer Res. 2019, 21, 10. [Google Scholar] [CrossRef] [PubMed]
- Semenza, G.L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J. Clin. Investig. 2013, 123, 3664–3671. [Google Scholar] [CrossRef] [PubMed]
- Fuady, J.H.; Gutsche, K.; Santambrogio, S.; Varga, Z.; Hoogewijs, D.; Wenger, R.H. Estrogen-dependent downregulation of hypoxia-inducible factor (HIF)-2α in invasive breast cancer cells. Oncotarget 2016, 7, 31153–31165. [Google Scholar] [CrossRef] [PubMed]
- Bialesova, L.; Xu, L.; Gustafsson, J.Å.; Haldosen, L.A.; Zhao, C.; Dahlman-Wright, K. Estrogen receptor β2 induces proliferation and invasiveness of triple negative breast cancer cells: Association with regulation of PHD3 and HIF-1α. Oncotarget 2017, 8, 76622–76633. [Google Scholar] [CrossRef] [PubMed]
- Jögi, A.; Ehinger, A.; Hartman, L.; Alkner, S. Expression of HIF-1α is related to a poor prognosis and tamoxifen resistance in contralateral breast cancer. PLoS ONE 2019, 14, e0226150. [Google Scholar] [CrossRef]
- Abu-Jamous, B.; Buffa, F.M.; Harris, A.L.; Nandi, A.K. In vitro downregulated hypoxia transcriptome is associated with poor prognosis in breast cancer. Mol. Cancer 2017, 16, 105. [Google Scholar] [CrossRef]
- Tasharrofi, B.; Soudyab, M.; Nikpayam, E.; Iranpour, M.; Mirfakhraie, R.; Sarrafzadeh, S.; Geranpayeh, L.; Azargashb, E.; Sayad, A.; Ghafouri-Fard, S. Comparative expression analysis of hypoxia-inducible factor-alpha and its natural occurring antisense in breast cancer tissues and adjacent noncancerous tissues. Cell Biochem. Funct. 2016, 34, 572–578. [Google Scholar] [CrossRef]
- Chen, Y.; Wang, Y.; Luo, W. ZMYND8 is a primary HIF coactivator that mediates breast cancer progression. Mol. Cell. Oncol. 2018, 5, e1479619. [Google Scholar] [CrossRef]
- Shi, Y.H.; Wang, Y.X.; Bingle, L.; Gong, L.H.; Heng, W.J.; Li, Y.; Fang, W.G. In vitro study of HIF-1 activation and VEGF release by bFGF in the T47D breast cancer cell line under normoxic conditions: Involvement of PI-3K/Akt and MEK1/ERK pathways. J. Pathol. 2005, 205, 530–536. [Google Scholar] [CrossRef]
- Fu, D.; He, C.; Wei, J.; Zhang, Z.; Luo, Y.; Tan, H.; Ren, C. PGK1 is a potential survival biomarker and invasion promoter by regulating the HIF-1α-mediated epithelial-mesenchymal transition process in breast cancer. Cell. Physiol. Biochem. 2018, 51, 2434–2444. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Geng, Y.H.; Wang, P.; Zhou, Y.T.; Yang, H.; Huo, Y.F.; Zhang, H.Q.; Li, Y.; He, H.Y.; Tian, X.X.; et al. Extracellular ATP promotes breast cancer invasion and epithelial-mesenchymal transition via hypoxia-inducible factor 2α signaling. Cancer Sci. 2019, 110, 2456–2470. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Chen, Y.; Bao, L.; Zhang, B.; Wang, J.E.; Kumar, A.; Xing, C.; Wang, Y.; Luo, W. CHD4 Promotes Breast Cancer Progression as a Coactivator of Hypoxia-Inducible Factors. Cancer Res. 2020, 80, 3880–3891. [Google Scholar] [CrossRef]
- Chen, F.; Chen, J.; Yang, L.; Liu, J.; Zhang, X.; Zhang, Y.; Tu, Q.; Yin, D.; Lin, D.; Wong, P.P.; et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumor-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat. Cell Biol. 2019, 21, 498–510. [Google Scholar] [CrossRef] [PubMed]
- Gómez, V.; Eykyn, T.R.; Mustapha, R.; Flores-Borja, F.; Male, V.; Barber, P.R.; Patsialou, A.; Green, R.; Panagaki, F.; Li, C.W.; et al. Breast cancer-associated macrophages promote tumorigenesis by suppressing succinate dehydrogenase in tumor cells. Sci. Signal. 2020, 13, eaax4585. [Google Scholar] [CrossRef]
- You, D.; Du, D.; Zhao, X.; Li, X.; Ying, M.; Hu, X. Mitochondrial malic enzyme 2 promotes breast cancer metastasis via stabilizing HIF-1α under hypoxia. Chin. J. Cancer Res.—Chung Kuo Yen Cheng Yen Chiu 2021, 33, 308–322. [Google Scholar] [CrossRef]
- Bao, X.; Zhang, J.; Huang, G.; Yan, J.; Xu, C.; Dou, Z.; Sun, C.; Zhang, H. The crosstalk between HIFs and mitochondrial dysfunctions in cancer development. Cell Death Dis. 2021, 12, 215. [Google Scholar] [CrossRef]
- Wang, G.L.; Semenza, G.L. General involvement of hypoxia-inducible factor 1 in transcriptional response to hypoxia. Proc. Natl. Acad. Sci. USA 1993, 90, 4304–4308. [Google Scholar] [CrossRef]
- Papandreou, I.; Cairns, R.A.; Fontana, L.; Lim, A.L.; Denko, N.C. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006, 3, 187–197. [Google Scholar] [CrossRef]
- Castelli, S.; Ciccarone, F.; Tavian, D.; Ciriolo, M.R. ROS-dependent HIF1α activation under forced lipid catabolism entails glycolysis and mitophagy as mediators of higher proliferation rate in cervical cancer cells. J. Exp. Clin. Cancer Res. 2021, 40, 94. [Google Scholar] [CrossRef]
- Schito, L.; Semenza, G.L. Hypoxia-Inducible Factors: Master Regulators of Cancer Progression. Trends Cancer 2016, 2, 758–770. [Google Scholar] [CrossRef] [PubMed]
- Soni, S.; Padwad, Y.S. HIF-1 in cancer therapy: Two-decade-long story of a transcription factor. Acta Oncol. 2017, 56, 503–515. [Google Scholar] [CrossRef] [PubMed]
- Keith, B.; Johnson, R.S.; Simon, M.C. HIF1α and HIF2α: Sibling rivalry in hypoxic tumor growth and progression. Nat. Rev. Cancer 2011, 12, 9–22. [Google Scholar] [CrossRef] [PubMed]
- Mimeault, M.; Batra, S.K. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J. Cell. Mol. Med. 2013, 17, 30–54. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Fan, J.; Yan, C.Y.; Ling, R.; Yun, J. Activation of hypoxia-inducible factor-1α by prolonged in vivo hyperinsulinemia treatment potentiates cancerous progression in estrogen receptor-positive breast cancer cells. Biochem. Biophys. Res. Commun. 2017, 491, 545–551. [Google Scholar] [CrossRef] [PubMed]
- Bharti, S.K.; Mironchik, Y.; Wildes, F.; Penet, M.F.; Goggins, E.; Krishnamachary, B.; Bhujwalla, Z.M. Metabolic consequences of HIF silencing in a triple negative human breast cancer xenograft. Oncotarget 2018, 9, 15326–15339. [Google Scholar] [CrossRef] [PubMed]
- Samanta, D.; Park, Y.; Ni, X.; Li, H.; Zahnow, C.A.; Gabrielson, E.; Pan, F.; Semenza, G.L. Chemotherapy induces enrichment of CD47+/CD73+/PDL1+ immune evasive triple-negative breast cancer cells. Proc. Natl. Acad. Sci. USA 2018, 115, E1239–E1248. [Google Scholar] [CrossRef]
- Tanaka, T.; Kutomi, G.; Kajiwara, T.; Kukita, K.; Kochin, V.; Kanaseki, T.; Tsukahara, T.; Hirohashi, Y.; Torigoe, T.; Okamoto, Y.; et al. Cancer-associated oxidoreductase ERO1-α promotes immune escape through up-regulation of PD-L1 in human breast cancer. Oncotarget 2017, 8, 24706–24718. [Google Scholar] [CrossRef]
- Lock, F.E.; McDonald, P.C.; Lou, Y.; Serrano, I.; Chafe, S.C.; Ostlund, C.; Aparicio, S.; Winum, J.Y.; Supuran, C.T.; Dedhar, S. Targeting carbonic anhydrase IX depletes breast cancer stem cells within the hypoxic niche. Oncogene 2013, 32, 5210–5219. [Google Scholar] [CrossRef]
- Moreno Roig, E.; Groot, A.J.; Yaromina, A.; Hendrickx, T.C.; Barbeau, L.; Giuranno, L.; Dams, G.; Ient, J.; Olivo Pimentel, V.; van Gisbergen, M.W.; et al. HIF-1α and HIF-2α Differently Regulate the Radiation Sensitivity of NSCLC Cells. Cells 2019, 8, 45. [Google Scholar] [CrossRef] [Green Version]
- Yan, Y.; Liu, F.; Han, L.; Zhao, L.; Chen, J.; Olopade, O.I.; He, M.; Wei, M. HIF-2α promotes conversion to a stem cell phenotype and induces chemoresistance in breast cancer cells by activating Wnt and Notch pathways. J. Exp. Clin. Cancer Res. 2018, 37, 256. [Google Scholar] [CrossRef] [PubMed]
- Menrad, H.; Werno, C.; Schmid, T.; Copanaki, E.; Deller, T.; Dehne, N.; Brüne, B. Roles of hypoxia-inducible factor-1alpha (HIF-1alpha) versus HIF-2alpha in the survival of hepatocellular tumor spheroids. Hepatology 2010, 51, 2183–2192. [Google Scholar] [CrossRef]
- Walter, K.M.; Schönenberger, M.J.; Trötzmüller, M.; Horn, M.; Elsässer, H.P.; Moser, A.B.; Lucas, M.S.; Schwarz, T.; Gerber, P.A.; Faust, P.L.; et al. Hif-2α promotes degradation of mammalian peroxisomes by selective autophagy. Cell Metab. 2014, 20, 882–897. [Google Scholar] [CrossRef] [PubMed]
- Zhong, R.; Xu, H.; Chen, G.; Zhao, G.; Gao, Y.; Liu, X.; Ma, S.; Dong, L. The role of hypoxia-inducible factor-1α in radiation-induced autophagic cell death in breast cancer cells. Tumor Biol. J. Int. Soc. Oncodev. Biol. Med. 2015, 36, 7077–7083. [Google Scholar] [CrossRef] [PubMed]
- Flamant, L.; Notte, A.; Ninane, N.; Raes, M.; Michiels, C. Anti-apoptotic role of HIF-1 and AP-1 in paclitaxel exposed breast cancer cells under hypoxia. Mol. Cancer 2010, 9, 191. [Google Scholar] [CrossRef]
- Michels, J.; Johnson, P.W.; Packham, G. Mcl-1. Int. J. Biochem. Cell Biol. 2005, 37, 267–271. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Bosch-Marce, M.; Shimoda, L.A.; Tan, Y.S.; Baek, J.H.; Wesley, J.B.; Gonzalez, F.J.; Semenza, G.L. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J. Biol. Chem. 2008, 283, 10892–10903. [Google Scholar] [CrossRef]
- Romero, M.A.; Bayraktar Ekmekcigil, O.; Bagca, B.G.; Avci, C.B.; Sabitaliyevich, U.Y.; Zhenisovna, T.G.; Aras, A.; Farooqi, A.A. Role of Autophagy in Breast Cancer Development and Progression: Opposite Sides of the Same Coin. Adv. Exp. Med. Biol. 2019, 1152, 65–73. [Google Scholar] [CrossRef]
- Lan, J.; Lu, H.; Samanta, D.; Salman, S.; Lu, Y.; Semenza, G.L. Hypoxia-inducible factor 1-dependent expression of adenosine receptor 2B promotes breast cancer stem cell enrichment. Proc. Natl. Acad. Sci. USA 2018, 115, E9640–E9648. [Google Scholar] [CrossRef]
- Wang, R.; Godet, I.; Yang, Y.; Salman, S.; Lu, H.; Lyu, Y.; Zuo, Q.; Wang, Y.; Zhu, Y.; Chen, C.; et al. Hypoxia-inducible factor-dependent ADAM12 expression mediates breast cancer invasion and metastasis. Proc. Natl. Acad. Sci. USA 2021, 118, e2020490118. [Google Scholar] [CrossRef]
- Funasaka, T.; Yanagawa, T.; Hogan, V.; Raz, A. Regulation of phosphoglucose isomerase/autocrine motility factor expression by hypoxia. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2005, 19, 1422–1430. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Wong, C.C.; Wei, H.; Gilkes, D.M.; Korangath, P.; Chaturvedi, P.; Schito, L.; Chen, J.; Krishnamachary, B.; Winnard, P.T., Jr.; et al. HIF-1-dependent expression of angiopoietin-like 4 and L1CAM mediates vascular metastasis of hypoxic breast cancer cells to the lungs. Oncogene 2012, 31, 1757–1770. [Google Scholar] [CrossRef] [PubMed]
- Regan Anderson, T.M.; Peacock, D.L.; Daniel, A.R.; Hubbard, G.K.; Lofgren, K.A.; Girard, B.J.; Schörg, A.; Hoogewijs, D.; Wenger, R.H.; Seagroves, T.N.; et al. Breast tumor kinase (Brk/PTK6) is a mediator of hypoxia-associated breast cancer progression. Cancer Res. 2013, 73, 5810–5820. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Xie, P.; Hao, N.; Zhang, M.; Liu, Y.; Liu, P.; Semenza, G.L.; He, J.; Zhang, H. HIF-1-regulated expression of calreticulin promotes breast tumorigenesis and progression through Wnt/β-catenin pathway activation. Proc. Natl. Acad. Sci. USA 2021, 118, e2109144118. [Google Scholar] [CrossRef]
- Ye, I.C.; Fertig, E.J.; DiGiacomo, J.W.; Considine, M.; Godet, I.; Gilkes, D.M. Molecular Portrait of Hypoxia in Breast Cancer: A Prognostic Signature and Novel HIF-Regulated Genes. Mol. Cancer Res. 2018, 16, 1889–1901. [Google Scholar] [CrossRef] [PubMed]
- Krutilina, R.I.; Playa, H.; Brooks, D.L.; Schwab, L.P.; Parke, D.N.; Oluwalana, D.; Layman, D.R.; Fan, M.; Johnson, D.L.; Yue, J.; et al. HIF-Dependent CKB Expression Promotes Breast Cancer Metastasis, Whereas Cyclocreatine Therapy Impairs Cellular Invasion and Improves Chemotherapy Efficacy. Cancers 2021, 14, 27. [Google Scholar] [CrossRef]
- Chaturvedi, P.; Gilkes, D.M.; Wong, C.C.; Kshitiz; Luo, W.; Zhang, H.; Wei, H.; Takano, N.; Schito, L.; Levchenko, A.; et al. Hypoxia-inducible factor-dependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. J. Clin. Investig. 2013, 123, 189–205. [Google Scholar] [CrossRef]
- Lin, Q.; Fang, X.; Liang, G.; Luo, Q.; Cen, Y.; Shi, Y.; Jia, S.; Li, J.; Yang, W.; Sanders, A.J.; et al. Silencing CTNND1 Mediates Triple-Negative Breast Cancer Bone Metastasis via Upregulating CXCR4/CXCL12 Axis and Neutrophils Infiltration in Bone. Cancers 2021, 13, 5703. [Google Scholar] [CrossRef]
- Todd, V.M.; Vecchi, L.A., 3rd; Clements, M.E.; Snow, K.P.; Ontko, C.D.; Himmel, L.; Pinelli, C.; Rafat, M.; Johnson, R.W. Hypoxia-inducible factor signaling in breast tumors controls spontaneous tumor dissemination in a site-specific manner. Commun. Biol. 2021, 4, 1122. [Google Scholar] [CrossRef]
- Liu, X.; Qiao, K.; Zhu, K.; Li, X.; Zhao, C.; Li, J.; Feng, D.; Fang, Y.; Wang, P.; Qian, C.; et al. Long Noncoding RNA HCG18 Promotes Malignant Phenotypes of Breast Cancer Cells via the HCG18/miR-103a-3p/UBE2O/mTORC1/HIF-1α-Positive Feedback Loop. Front. Cell Dev. Biol. 2021, 9, 675082. [Google Scholar] [CrossRef]
- Ju, J.A.; Godet, I.; Ye, I.C.; Byun, J.; Jayatilaka, H.; Lee, S.J.; Xiang, L.; Samanta, D.; Lee, M.H.; Wu, P.H.; et al. Hypoxia Selectively Enhances Integrin α5β1 Receptor Expression in Breast Cancer to Promote Metastasis. Mol. Cancer Res. 2017, 15, 723–734. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.; Song, X.; Li, Y.; Chen, B.; Zhao, W.; Wang, L.; Zhang, H.; Liu, Y.; Han, D.; Zhang, N.; et al. LncRNA BCRT1 promotes breast cancer progression by targeting the miR-1303/PTBP3 axis. Mol. Cancer 2020, 19, 85. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Maldonado, L.; Tiana, M.; Roche, O.; Prado-Cabrero, A.; Jensen, L.; Fernandez-Barral, A.; Guijarro-Muñoz, I.; Favaro, E.; Moreno-Bueno, G.; Sanz, L.; et al. EFNA3 long noncoding RNAs induced by hypoxia promote metastatic dissemination. Oncogene 2015, 34, 2609–2620. [Google Scholar] [CrossRef] [PubMed]
- Niu, Y.; Bao, L.; Chen, Y.; Wang, C.; Luo, M.; Zhang, B.; Zhou, M.; Wang, J.E.; Fang, Y.V.; Kumar, A.; et al. HIF2-Induced Long Noncoding RNA RAB11B-AS1 Promotes Hypoxia-Mediated Angiogenesis and Breast Cancer Metastasis. Cancer Res. 2020, 80, 964–975. [Google Scholar] [CrossRef]
- Ko, Y.S.; Rugira, T.; Jin, H.; Joo, Y.N.; Kim, H.J. Radiotherapy-Resistant Breast Cancer Cells Enhance Tumor Progression by Enhancing Premetastatic Niche Formation through the HIF-1α-LOX Axis. Int. J. Mol. Sci. 2020, 21, 8027. [Google Scholar] [CrossRef]
- Saatci, O.; Kaymak, A.; Raza, U.; Ersan, P.G.; Akbulut, O.; Banister, C.E.; Sikirzhytski, V.; Tokat, U.M.; Aykut, G.; Ansari, S.A.; et al. Targeting lysyl oxidase (LOX) overcomes chemotherapy resistance in triple negative breast cancer. Nat. Commun. 2020, 11, 2416. [Google Scholar] [CrossRef]
- Wong, C.C.; Zhang, H.; Gilkes, D.M.; Chen, J.; Wei, H.; Chaturvedi, P.; Hubbi, M.E.; Semenza, G.L. Inhibitors of hypoxia-inducible factor 1 block breast cancer metastatic niche formation and lung metastasis. J. Mol. Med. 2012, 90, 803–815. [Google Scholar] [CrossRef]
- Moon, E.J.; Mello, S.S.; Li, C.G.; Chi, J.T.; Thakkar, K.; Kirkland, J.G.; Lagory, E.L.; Lee, I.J.; Diep, A.N.; Miao, Y.; et al. The HIF target MAFF promotes tumor invasion and metastasis through IL11 and STAT3 signaling. Nat. Commun. 2021, 12, 4308. [Google Scholar] [CrossRef]
- Goggins, E.; Kakkad, S.; Mironchik, Y.; Jacob, D.; Wildes, F.; Krishnamachary, B.; Bhujwalla, Z.M. Hypoxia-Inducible Factors Modify Collagen I Fibers in MDA-MB-231 Triple Negative Breast Cancer Xenografts. Neoplasia 2018, 20, 131–139. [Google Scholar] [CrossRef]
- Li, N.A.; Wang, H.; Zhang, J.; Zhao, E. Knockdown of hypoxia inducible factor-2α inhibits cell invasion via the downregulation of MMP-2 expression in breast cancer cells. Oncol. Lett. 2016, 11, 3743–3748. [Google Scholar] [CrossRef] [Green Version]
- Hamad, H.A.; Enezei, H.H.; Alrawas, A.; Zakuan, N.M.; Abdullah, N.A.; Cheah, Y.K.; Hashim, N. Identification of Potential Chemical Substrates as Fuel for Hypoxic Tumors That May Be Linked to Invadopodium Formation in Hypoxia-Induced MDA-MB-231 Breast-Cancer Cell Line. Molecules 2020, 25, 3876. [Google Scholar] [CrossRef] [PubMed]
- Muñoz-Nájar, U.M.; Neurath, K.M.; Vumbaca, F.; Claffey, K.P. Hypoxia stimulates breast carcinoma cell invasion through MT1-MMP and MMP-2 activation. Oncogene 2006, 25, 2379–2392. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.Y.; Jang, Y.S.; Min, S.Y.; Song, J.Y. Overexpression of MMP-9 and HIF-1α in Breast Cancer Cells under Hypoxic Conditions. J. Breast Cancer 2011, 14, 88–95. [Google Scholar] [CrossRef] [PubMed]
- Schito, L.; Rey, S.; Tafani, M.; Zhang, H.; Wong, C.C.; Russo, A.; Russo, M.A.; Semenza, G.L. Hypoxia-inducible factor 1-dependent expression of platelet-derived growth factor B promotes lymphatic metastasis of hypoxic breast cancer cells. Proc. Natl. Acad. Sci. USA 2012, 109, E2707–E2716. [Google Scholar] [CrossRef]
- Storci, G.; Sansone, P.; Mari, S.; D’Uva, G.; Tavolari, S.; Guarnieri, T.; Taffurelli, M.; Ceccarelli, C.; Santini, D.; Chieco, P.; et al. TNFalpha up-regulates SLUG via the NF-kappaB/HIF1alpha axis, which imparts breast cancer cells with a stem cell-like phenotype. J. Cell. Physiol. 2010, 225, 682–691. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.H.; Wu, M.Z.; Chiou, S.H.; Chen, P.M.; Chang, S.Y.; Liu, C.J.; Teng, S.C.; Wu, K.J. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat. Cell Biol. 2008, 10, 295–305. [Google Scholar] [CrossRef]
- Semenza, G.L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 2003, 3, 721–732. [Google Scholar] [CrossRef]
- Xiang, L.; Gilkes, D.M.; Chaturvedi, P.; Luo, W.; Hu, H.; Takano, N.; Liang, H.; Semenza, G.L. Ganetespib blocks HIF-1 activity and inhibits tumor growth, vascularization, stem cell maintenance, invasion, and metastasis in orthotopic mouse models of triple-negative breast cancer. J. Mol. Med. 2014, 92, 151–164. [Google Scholar] [CrossRef]
- Befani, C.; Liakos, P. The role of hypoxia-inducible factor-2 alpha in angiogenesis. J. Cell. Physiol. 2018, 233, 9087–9098. [Google Scholar] [CrossRef]
- Dillekås, H.; Rogers, M.S.; Straume, O. Are 90% of deaths from cancer caused by metastases? Cancer Med. 2019, 8, 5574–5576. [Google Scholar] [CrossRef] [Green Version]
- Gilkes, D.M.; Semenza, G.L. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol. 2013, 9, 1623–1636. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Imanaka, N.; Chen, J.; Griffin, J.D. Hypoxia potentiates Notch signaling in breast cancer leading to decreased E-cadherin expression and increased cell migration and invasion. Br. J. Cancer 2010, 102, 351–360. [Google Scholar] [CrossRef] [PubMed]
- Mathieu, J.; Zhang, Z.; Zhou, W.; Wang, A.J.; Heddleston, J.M.; Pinna, C.M.; Hubaud, A.; Stadler, B.; Choi, M.; Bar, M.; et al. HIF induces human embryonic stem cell markers in cancer cells. Cancer Res. 2011, 71, 4640–4652. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Lu, H.; Xiang, L.; Bullen, J.W.; Zhang, C.; Samanta, D.; Gilkes, D.M.; He, J.; Semenza, G.L. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc. Natl. Acad. Sci. USA 2015, 112, E6215–E6223. [Google Scholar] [CrossRef]
- Ricardo, S.; Vieira, A.F.; Gerhard, R.; Leitão, D.; Pinto, R.; Cameselle-Teijeiro, J.F.; Milanezi, F.; Schmitt, F.; Paredes, J. Breast cancer stem cell markers CD44, CD24 and ALDH1: Expression distribution within intrinsic molecular subtype. J. Clin. Pathol. 2011, 64, 937–946. [Google Scholar] [CrossRef]
- Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef]
- Shiraishi, A.; Tachi, K.; Essid, N.; Tsuboi, I.; Nagano, M.; Kato, T.; Yamashita, T.; Bando, H.; Hara, H.; Ohneda, O. Hypoxia promotes the phenotypic change of aldehyde dehydrogenase activity of breast cancer stem cells. Cancer Sci. 2017, 108, 362–372. [Google Scholar] [CrossRef]
- Kim, R.J.; Park, J.R.; Roh, K.J.; Choi, A.R.; Kim, S.R.; Kim, P.H.; Yu, J.H.; Lee, J.W.; Ahn, S.H.; Gong, G.; et al. High aldehyde dehydrogenase activity enhances stem cell features in breast cancer cells by activating hypoxia-inducible factor-2α. Cancer Lett. 2013, 333, 18–31. [Google Scholar] [CrossRef]
- Brooks, D.L.; Schwab, L.P.; Krutilina, R.; Parke, D.N.; Sethuraman, A.; Hoogewijs, D.; Schörg, A.; Gotwald, L.; Fan, M.; Wenger, R.H.; et al. ITGA6 is directly regulated by hypoxia-inducible factors and enriches cancer stem cell activity and invasion in metastatic breast cancer models. Mol. Cancer 2016, 15, 26. [Google Scholar] [CrossRef]
- Xiang, L.; Gilkes, D.M.; Hu, H.; Luo, W.; Bullen, J.W.; Liang, H.; Semenza, G.L. HIF-1α and TAZ serve as reciprocal co-activators in human breast cancer cells. Oncotarget 2015, 6, 11768–11778. [Google Scholar] [CrossRef] [Green Version]
- Samanta, D.; Gilkes, D.M.; Chaturvedi, P.; Xiang, L.; Semenza, G.L. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, E5429–E5438. [Google Scholar] [CrossRef] [PubMed]
- Lu, H.; Tran, L.; Park, Y.; Chen, I.; Lan, J.; Xie, Y.; Semenza, G.L. Reciprocal Regulation of DUSP9 and DUSP16 Expression by HIF1 Controls ERK and p38 MAP Kinase Activity and Mediates Chemotherapy-Induced Breast Cancer Stem Cell Enrichment. Cancer Res. 2018, 78, 4191–4202. [Google Scholar] [CrossRef]
- Wolff, M.; Kosyna, F.K.; Dunst, J.; Jelkmann, W.; Depping, R. Impact of hypoxia inducible factors on estrogen receptor expression in breast cancer cells. Arch. Biochem. Biophys. 2017, 613, 23–30. [Google Scholar] [CrossRef] [PubMed]
- Nie, C.; Lv, H.; Bie, L.; Hou, H.; Chen, X. Hypoxia-inducible factor 1-alpha expression correlates with response to neoadjuvant chemotherapy in women with breast cancer. Medicine 2018, 97, e13551. [Google Scholar] [CrossRef] [PubMed]
- Cui, J.; Jiang, H. Prediction of postoperative survival of triple-negative breast cancer based on nomogram model combined with expression of HIF-1α and c-myc. Medicine 2019, 98, e17370. [Google Scholar] [CrossRef] [PubMed]
- Shamis, S.; McMillan, D.C.; Edwards, J. The relationship between hypoxia-inducible factor 1α (HIF-1α) and patient survival in breast cancer: Systematic review and meta-analysis. Crit. Rev. Oncol./Hematol. 2021, 159, 103231. [Google Scholar] [CrossRef]
- Zhang, J.; Zhang, S.; Gao, S.; Ma, Y.; Tan, X.; Kang, Y.; Ren, W. HIF-1α, TWIST-1, and ITGB-1, associated with Tumor Stiffness, as Novel Predictive Markers for the Pathological Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancer Manag. Res. 2020, 12, 2209–2222. [Google Scholar] [CrossRef]
- Liu, L.; Liu, W.; Wang, L.; Zhu, T.; Zhong, J.; Xie, N. Hypoxia-inducible factor 1 mediates intermittent hypoxia-induced migration of human breast cancer MDA-MB-231 cells. Oncol. Lett. 2017, 14, 7715–7722. [Google Scholar] [CrossRef]
- Soleymani Abyaneh, H.; Gupta, N.; Radziwon-Balicka, A.; Jurasz, P.; Seubert, J.; Lai, R.; Lavasanifar, A. STAT3 but Not HIF-1α Is Important in Mediating Hypoxia-Induced Chemoresistance in MDA-MB-231, a Triple Negative Breast Cancer Cell Line. Cancers 2017, 9, 137. [Google Scholar] [CrossRef]
- Koh, M.Y.; Spivak-Kroizman, T.; Venturini, S.; Welsh, S.; Williams, R.R.; Kirkpatrick, D.L.; Powis, G. Molecular mechanisms for the activity of PX-478, an antitumor inhibitor of the hypoxia-inducible factor-1alpha. Mol. Cancer Ther. 2008, 7, 90–100. [Google Scholar] [CrossRef] [Green Version]
- Bai, J.; Chen, W.B.; Zhang, X.Y.; Kang, X.N.; Jin, L.J.; Zhang, H.; Wang, Z.Y. HIF-2α regulates CD44 to promote cancer stem cell activation in triple-negative breast cancer via PI3K/AKT/mTOR signaling. World J. Stem Cells 2020, 12, 87–99. [Google Scholar] [CrossRef] [PubMed]
- Kachamakova-Trojanowska, N.; Podkalicka, P.; Bogacz, T.; Barwacz, S.; Józkowicz, A.; Dulak, J.; Łoboda, A. HIF-1 stabilization exerts anticancer effects in breast cancer cells in vitro and in vivo. Biochem. Pharmacol. 2020, 175, 113922. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Hill, H.; Christie, A.; Kim, M.S.; Holloman, E.; Pavia-Jimenez, A.; Homayoun, F.; Ma, Y.; Patel, N.; Yell, P.; et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature 2016, 539, 112–117. [Google Scholar] [CrossRef]
- Xu, R.; Wang, K.; Rizzi, J.P.; Huang, H.; Grina, J.A.; Schlachter, S.T.; Wang, B.; Wehn, P.M.; Yang, H.; Dixon, D.D.; et al. 3-[(1S,2S,3R)-2,3-Difluoro-1-hydroxy-7-methylsulfonylindan-4-yl]oxy-5-fluorobenzonitrile (PT2977), a Hypoxia-Inducible Factor 2α (HIF-2α) Inhibitor for the Treatment of Clear Cell Renal Cell Carcinoma. J. Med. Chem. 2019, 62, 6876–6893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Target Gene | HIF Isoform Regulation | Protein/RNA Role | References |
---|---|---|---|
A2BAR | HIF-1α | adenosine receptor 2A signaling in breast cancer cells promotes filopodia formation, invasion, and metastasis | [59] |
ADAM12 | HIF-1α and HIF-2α | disintegrin and metalloproteinase 12 (ADAM12); trim off the extracellular domain of the membrane-bound heparin-binding epidermal growth factor-like growth factor (HB-EGF), which binds to EGFR and triggers a signal transduction pathway involved in metastasis | [60] |
AMF | HIF-1α | autocrine motility factor; stimulates motility of cells under hypoxia | [61] |
ANGPTL4 | HIF-1α | angiopoietin-like 4; a secreted factor that inhibits EC-EC interaction | [62] |
BRK | HIF-1α and HIF-2α | Breast tumor kinase, increases migration via modulation of EMT -associated molecules | [63] |
CALR | HIF-1α | Calreticulin; facilitates invasion and metastasis by promoting the breast cancer stem cell phenotype through Wnt/β-catenin signaling | [64] |
CASP14 | HIF-1α | Caspase 14; breast cancer stem cell marker; has also been implicated as a gene found in brain-metastatic compared to non-brain metastatic breast cancer | [65] |
CKB | HIF-1α | Creatine kinase brain isoform, is a major effector of the HIF-1α-mediated promotion of metastatic phenotypes in ER-negative breast cancer | [66] |
CXCR3 | HIF-1α | CXCR3 is a Gαi G-protein coupled receptor (GPCR), a seven transmembrane spanning receptor; is the receptor for CXCL10 chemokine, increases migration | [67] |
CXCR4 | HIF-1α | CXCR-4 is an alpha-chemokine receptor specific for stromal-derived-factor-1 (SDF-1 also called CXCL12); tumor cells overexpressing CXCR4, have higher potential to survive the circulation stage | [68,69] |
HCG18 | HIF-1α | Long Noncoding RNA HLA complex group 18; positively regulates the expression of BC-related ubiquitin-conjugating enzyme E2O (UBE2O) by sponging miR-103a-3p, thus promoting the malignant phenotypes of BC cells through the UBE2O/AMPKα2/mTORC1 axis. | [70] |
ITGA5 | HIF-1α and HIF-2α | integrin that binds to fibronectin, promotes lung metastasis in orthotopic transplantation models of triple negative breast cancer | [71] |
L1CAM | HIF-1α | L1 cell adhesion molecule; integral membrane glycoprotein belonging to a large class of immunoglobulin superfamily cell adhesion molecules (CAMs) that mediate cell-to-cell adhesion at the cell surface | [62] |
LncRNA BCRT1 | HIF-1α | Long Noncoding RNA (breast cancer-related transcript 1; promotes cell mobility and tumor metastasis in breast cancer; regulate the EMT process | [72] |
LncRNA EFNA3 | HIF-1α | Long Noncoding RNA ephrin A3, increases protein ephrin 3 levels by sequestering miRNAs away from EFNA3 mRNA allowing for its translation; | [73] |
LncRNA RAB11B-AS1 | HIF-2α | Long Noncoding RNA RAB11B-AS1; increases the expression of angiogenic factors including VEGFA and ANGPTL4 in hypoxic breast cancer cells through the recruitment of RNA Pol II | [74] |
LOX | HIF-1α | Lysyl Oxidase, post-translationally modifies collagen molecules in the extracellular matrix (ECM), regulates cell adhesion, motility and invasion; increases metastasis by enhancing premetastatic niche formation | [75,76,77] |
LOXL2 | HIF-1α and HIF-2α | lysyl oxidase-like 2; post-translationally modifies collagen which is a component of the extracellular matrix (ECM); promotes lung metastasis by facilitating the formation of the pre-metastatic niche | [32,77] |
MAFF | HIF-1α | v-maf musculoaponeurotic fibrosarcoma oncogene homolog F; transcription factor regulating tumor invasion and metastasis via IL11/STAT3 pathways activation; | [78] |
MMP1 | HIF-2α | degradation of the extracellular matrix MMP-1, mostly type I collagen, promotes tumor growth and metastasis particularly to the brain | [79] |
MMP2 | HIF-1α and HIF-2α | matrix metalloproteinase 2; degradation of extracellular matrix; degrades type IV collagen | [80,81,82] |
MMP9 | HIF-1α and HIF-2α | matrix metallopeptidase 9; degradation of the extracellular matrix | [32,83] |
MMP14/MT1-MMP | HIF-1α and HIF-2α | matrix metalloproteinase-14 or MT1-MMP, is a member of the membrane-type MMP subfamily; degradation of the extracellular matrix | [79] |
P4HA1 | HIF-1α and HIF-2α | prolyl 4-hydroxylase-α1; essential for collagen biogenesis; 4-hydroxyproline residues are necessary for the proper folding of collagen polypeptide chains into stable triple helical molecule; HIF-1α-dependent ECM remodeling increases cell motility and promote invasion and metastasis. | [79] |
P4HA2 | HIF-1α | prolyl 4-hydroxylase-α2; essential for collagen biogenesis; 4-hydroxyproline residues are necessary for the proper folding of collagen polypeptide chains into a stable triple helical molecule; HIF-1α-dependent ECM remodeling increases cell motility and promotes invasion and metastasis. | [79] |
PDGFB | HIF-1α | platelet-derived growth factor B; promotes, metastasis of hypoxic breast cancer cells via lymphatic dissemination | [84] |
PGF | HIF-1α | placental growth factor, promotes the metastasis of breast cancer cells; promotes recruitment of mesenchymal stem cell, to the primary tumor site and, stimulates them to the expression of CXCL10 | [67] |
SNAIL1 | HIF-2α | transcriptional factor; plays a key role in the control of epithelial to mesenchymal transition | [32] |
SLUG | HIF-1α | transcriptional factor; plays a key role in the control of epithelial to mesenchymal transition | [85], |
TWIST | HIF-1α | transcriptional factor; plays a key role in the control of epithelial to mesenchymal transition | [86] |
VEGF | HIF-1α | vascular endothelial growth factor; stimulates the formation of blood vessels | [87,88] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kozal, K.; Krześlak, A. The Role of Hypoxia-Inducible Factor Isoforms in Breast Cancer and Perspectives on Their Inhibition in Therapy. Cancers 2022, 14, 4518. https://doi.org/10.3390/cancers14184518
Kozal K, Krześlak A. The Role of Hypoxia-Inducible Factor Isoforms in Breast Cancer and Perspectives on Their Inhibition in Therapy. Cancers. 2022; 14(18):4518. https://doi.org/10.3390/cancers14184518
Chicago/Turabian StyleKozal, Karolina, and Anna Krześlak. 2022. "The Role of Hypoxia-Inducible Factor Isoforms in Breast Cancer and Perspectives on Their Inhibition in Therapy" Cancers 14, no. 18: 4518. https://doi.org/10.3390/cancers14184518
APA StyleKozal, K., & Krześlak, A. (2022). The Role of Hypoxia-Inducible Factor Isoforms in Breast Cancer and Perspectives on Their Inhibition in Therapy. Cancers, 14(18), 4518. https://doi.org/10.3390/cancers14184518