Skip to main content

Advertisement

Log in

Tumor pH and metastasis: a malignant process beyond hypoxia

  • Published:
Cancer and Metastasis Reviews Aims and scope Submit manuscript

A Correction to this article was published on 15 June 2019

This article has been updated

Abstract

Tumors often show, compared to normal tissues, a markedly decreased extracellular pH resulting from anaerobic or aerobic glycolysis in combination with a reduced removal of acidic metabolites. Several studies indicate that acidosis induces (independently from hypoxia) hematogenous and lymphatic spread of tumor cells worsening the long-term prognosis of tumor patients. This review gives an overview on the impact of low pH on different steps of metastasis including (a) local tumor cell invasion and angiogenesis, (b) intravasation of tumor cells and detachment into the circulation, and (c) adherence of circulating tumor cells, transmigration and invasion in the new host tissue. The review describes pH-dependent cellular mechanisms fostering these steps such as endothelial-to-mesenchymal transition (EMT), activation of cell migration, degradation of the extracellular matrix, or angiogenesis. The review discusses mechanisms of tumor cells for proton sensing including acid-sensitive ion channels (ASICs, TRPs) or ion transporters (NHE1) and G protein coupled H+-sensors. Finally, the review describes several intracellular signaling cascades activated by H+ sensing mechanisms leading to transcriptional, post-transcriptional, or functional changes in the cell relevant for the metastatic spread. From these studies, different therapeutical approaches are described to overcome tumor acidosis or to interfere with the signaling cascades to reduce the metastatic potential of tumors.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Change history

  • 27 June 2019

    The authors have noticed a typographical error in the published article. The term “epithelial-to-mesenchymal transition” should have been used instead of the term “endothelial-to-mesenchymal transition” throughout the manuscript.

Abbreviations

CREB:

cAMP response element-binding protein

ASIC:

Acid-sensing ion channel

EMT:

Endothelial-to-mesenchymal transition

GPR:

G protein-coupled receptor

HIF:

Hypoxia-inducible factor

MAPK:

Mitogen-activated protein kinase

MMP:

Matrix-metalloproteinase

TRP:

Transient receptor potential channel

References

  1. Vander Heiden, M. G., Cantley, L. C., & Thompson, C. B. (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science, 324(5930), 1029–1033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Folkman, J. (1971). Tumor angiogenesis: therapeutic implications. The New England Journal of Medicine, 285(21), 1182–1186.

    Article  CAS  PubMed  Google Scholar 

  3. De Palma, M., Biziato, D., & Petrova, T. V. (2017). Microenvironmental regulation of tumour angiogenesis. Nature Reviews. Cancer, 17(8), 457–474. https://doi.org/10.1038/nrc.2017.51.

    Article  CAS  PubMed  Google Scholar 

  4. Konerding, M. A., Malkusch, W., Klapthor, B., van Ackern, C., Fait, E., Hill, S. A., et al. (1999). Evidence for characteristic vascular patterns in solid tumours: quantitative studies using corrosion casts. British Journal of Cancer, 80(5–6), 724–732.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bussink, J., Kaanders, J. H., Rijken, P. F., Martindale, C. A., & van der Kogel, A. J. (1998). Multiparameter analysis of vasculature, perfusion and proliferation in human tumour xenografts. British Journal of Cancer, 77(1), 57–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vaupel, P., Kallinowski, F., & Okunieff, P. (1989). Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review. Cancer Research, 49, 6449–6465.

    CAS  PubMed  Google Scholar 

  7. Gray, L. H., Conger, A. D., Ebert, M., Hornsey, S., & Scott, O. C. A. (1953). The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. The British Journal of Radiology, 26, 638–648.

    Article  CAS  PubMed  Google Scholar 

  8. Warburg, O. (1925). Über den Stoffwechsel der Carcinomzelle. Klinische Wochenschrift, 4, 534–536. https://doi.org/10.1007/BF01726151.

  9. Lindeman, L. R., Randtke, E. A., High, R. A., Jones, K. M., Howison, C. M., & Pagel, M. D. (2018). A comparison of exogenous and endogenous CEST MRI methods for evaluating in vivo pH. Magnetic Resonance in Medicine, 79(5), 2766–2772. https://doi.org/10.1002/mrm.26924.

    Article  PubMed  Google Scholar 

  10. Fidler, I. J. (2003). The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nature Reviews. Cancer, 3(6), 453–458. https://doi.org/10.1038/nrc1098.

    Article  CAS  PubMed  Google Scholar 

  11. Hunter, K. W., Crawford, N. P., & Alsarraj, J. (2008). Mechanisms of metastasis. Breast Cancer Research, 10(Suppl 1), S2. https://doi.org/10.1186/bcr1988.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lambert, A. W., Pattabiraman, D. R., & Weinberg, R. A. (2017). Emerging biological principles of metastasis. Cell, 168(4), 670–691. https://doi.org/10.1016/j.cell.2016.11.037.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. van Zijl, F., Krupitza, G., & Mikulits, W. (2011). Initial steps of metastasis: cell invasion and endothelial transmigration. Mutation Research, 728(1–2), 23–34. https://doi.org/10.1016/j.mrrev.2011.05.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ye, X., & Weinberg, R. A. (2015). Epithelial-mesenchymal plasticity: a central regulator of cancer progression. Trends in Cell Biology, 25(11), 675–686. https://doi.org/10.1016/j.tcb.2015.07.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lamouille, S., Xu, J., & Derynck, R. (2014). Molecular mechanisms of epithelial-mesenchymal transition. Nature Reviews. Molecular Cell Biology, 15(3), 178–196. https://doi.org/10.1038/nrm3758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Le Bras, G. F., Taubenslag, K. J., & Andl, C. D. (2012). The regulation of cell-cell adhesion during epithelial-mesenchymal transition, motility and tumor progression. Cell Adhesion & Migration, 6(4), 365–373. https://doi.org/10.4161/cam.21326.

    Article  Google Scholar 

  17. Chiang, A. C., & Massague, J. (2008). Molecular basis of metastasis. The New England Journal of Medicine, 359(26), 2814–2823. https://doi.org/10.1056/NEJMra0805239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Valastyan, S., & Weinberg, R. A. (2011). Tumor metastasis: molecular insights and evolving paradigms. Cell, 147(2), 275–292. https://doi.org/10.1016/j.cell.2011.09.024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Spiegel, A., Brooks, M. W., Houshyar, S., Reinhardt, F., Ardolino, M., Fessler, E., Chen, M. B., Krall, J. A., DeCock, J., Zervantonakis, I. K., Iannello, A., Iwamoto, Y., Cortez-Retamozo, V., Kamm, R. D., Pittet, M. J., Raulet, D. H., & Weinberg, R. A. (2016). Neutrophils suppress intraluminal NK cell-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discovery, 6(6), 630–649. https://doi.org/10.1158/2159-8290.CD-15-1157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Psaila, B., & Lyden, D. (2009). The metastatic niche: adapting the foreign soil. Nature Reviews Cancer, 9(4), 285–293. https://doi.org/10.1038/nrc2621.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Vaupel, P., & Mayer, A. (2007). Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Reviews, 26(2), 225–239. https://doi.org/10.1007/s10555-007-9055-1.

    Article  CAS  PubMed  Google Scholar 

  22. Nordsmark, M., Bentzen, S. M., Rudat, V., Brizel, D., Lartigau, E., Stadler, P., et al. (2005). Prognostic value of tumor oxygenation in 397 head and neck tumors after primary radiation therapy. An international multi-center study. Radiotherapy and Oncology, 77(1), 18–24. https://doi.org/10.1016/j.radonc.2005.06.038.

    Article  PubMed  Google Scholar 

  23. Höckel, M., Schlenger, K., Aral, B., Mitze, M., Schäffer, U., & Vaupel, P. (1996). Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Research, 56(19), 4509–4515.

    PubMed  Google Scholar 

  24. Semenza, G. L. (2016). The hypoxic tumor microenvironment: a driving force for breast cancer progression. Biochimica et Biophysica Acta, 1863(3), 382–391. https://doi.org/10.1016/j.bbamcr.2015.05.036.

    Article  CAS  PubMed  Google Scholar 

  25. Ackerstaff, E., Artemov, D., Gillies, R. J., & Bhujwalla, Z. M. (2007). Hypoxia and the presence of human vascular endothelial cells affect prostate cancer cell invasion and metabolism. Neoplasia, 9(12), 1138–1151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Laitala, A., & Erler, J. T. (2018). Hypoxic signalling in tumour stroma. Frontiers in Oncology, 8, 189. https://doi.org/10.3389/fonc.2018.00189.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Ishikawa, H., Sakurai, H., Hasegawa, M., Mitsuhashi, N., Takahashi, M., Masuda, N., Nakajima, M., Kitamoto, Y., Saitoh, J. I., & Nakano, T. (2004). Expression of hypoxic-inducible factor 1alpha predicts metastasis-free survival after radiation therapy alone in stage IIIB cervical squamous cell carcinoma. International Journal of Radiation Oncology, Biology, Physics, 60(2), 513–521. https://doi.org/10.1016/j.ijrobp.2004.03.025.

    Article  CAS  PubMed  Google Scholar 

  28. Chang, Q., Jurisica, I., Do, T., & Hedley, D. W. (2011). Hypoxia predicts aggressive growth and spontaneous metastasis formation from orthotopically grown primary xenografts of human pancreatic cancer. Cancer Research, 71(8), 3110–3120. https://doi.org/10.1158/0008-5472.CAN-10-4049.

    Article  CAS  PubMed  Google Scholar 

  29. Schito, L., & Rey, S. (2017). Hypoxic pathobiology of breast cancer metastasis. Biochimica et Biophysica Acta, Reviews on Cancer, 1868(1), 239–245. https://doi.org/10.1016/j.bbcan.2017.05.004.

    Article  CAS  PubMed  Google Scholar 

  30. Gillies, R. J., & Gatenby, R. A. (2007). Hypoxia and adaptive landscapes in the evolution of carcinogenesis. Cancer Metastasis Reviews, 26(2), 311–317. https://doi.org/10.1007/s10555-007-9065-z.

    Article  CAS  PubMed  Google Scholar 

  31. Gao, T., Li, J. Z., Lu, Y., Zhang, C. Y., Li, Q., Mao, J., & Li, L. H. (2016). The mechanism between epithelial mesenchymal transition in breast cancer and hypoxia microenvironment. Biomedicine & Pharmacotherapy, 80, 393–405. https://doi.org/10.1016/j.biopha.2016.02.044.

    Article  CAS  Google Scholar 

  32. Rankin, E. B., & Giaccia, A. J. (2016). Hypoxic control of metastasis. Science, 352(6282), 175–180. https://doi.org/10.1126/science.aaf4405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gilkes, D. M. (2016). Implications of hypoxia in breast cancer metastasis to bone. International Journal of Molecular Sciences, 17(10), https://doi.org/10.3390/ijms17101669.

  34. Nobre, A. R., Entenberg, D., Wang, Y., Condeelis, J., & Aguirre-Ghiso, J. A. (2018). The different routes to metastasis via hypoxia-regulated programs. Trends in Cell Biology, 28, 941–956. https://doi.org/10.1016/j.tcb.2018.06.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Walenta, S., Voelxen, N. F., & Mueller-Klieser, W. (2016). Lactate—an integrative mirror of cancer metabolism. Recent Results in Cancer Research, 207, 23–37. https://doi.org/10.1007/978-3-319-42118-6_2.

    Article  CAS  PubMed  Google Scholar 

  36. Walenta, S., Wetterling, M., Lehrke, M., Schwickert, G., Sundfor, K., Rofstad, E. K., et al. (2000). High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Research, 60(4), 916–921.

    CAS  PubMed  Google Scholar 

  37. Ellingsen, C., Walenta, S., Hompland, T., Mueller-Klieser, W., & Rofstad, E. K. (2013). The microenvironment of cervical carcinoma xenografts: associations with lymph node metastasis and its assessment by DCE-MRI. Translational Oncology, 6(5), 607–617.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Dhup, S., Dadhich, R. K., Porporato, P. E., & Sonveaux, P. (2012). Multiple biological activities of lactic acid in cancer: influences on tumor growth, angiogenesis and metastasis. Current Pharmaceutical Design, 18(10), 1319–1330.

    Article  CAS  PubMed  Google Scholar 

  39. Payen, V. L., Hsu, M. Y., Radecke, K. S., Wyart, E., Vazeille, T., Bouzin, C., et al. (2017). Monocarboxylate transporter MCT1 promotes tumor metastasis independently of its activity as a lactate transporter. Cancer Research, 77(20), 5591–5601. https://doi.org/10.1158/0008-5472.CAN-17-0764.

    Article  CAS  PubMed  Google Scholar 

  40. Shah, S. A., Jain, R. K., & Finney, P. L. (1983). Enhanced metastasis formation by combined hyperthermia and hyperglycemia in rats bearing Walker 256 carcinosarcoma. Cancer Letters, 19(3), 317–323.

    Article  CAS  PubMed  Google Scholar 

  41. Schlappack, O. K., Zimmermann, A., & Hill, R. P. (1991). Glucose starvation and acidosis: effect on experimental metastatic potential, DNA content and MTX resistance of murine tumour cells. British Journal of Cancer, 64(4), 663–670.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rofstad, E. K., Mathiesen, B., Kindem, K., & Galappathi, K. (2006). Acidic extracellular pH promotes experimental metastasis of human melanoma cells in athymic nude mice. Cancer Research, 66(13), 6699–6707. https://doi.org/10.1158/0008-5472.CAN-06-0983.

    Article  CAS  PubMed  Google Scholar 

  43. Riemann, A., Schneider, B., Gündel, D., Stock, C., Thews, O., & Gekle, M. (2014). Acidic priming enhances metastatic potential of cancer cells. Pflügers Archiv, 466(11), 2127–2138. https://doi.org/10.1007/s00424-014-1458-6.

    Article  CAS  PubMed  Google Scholar 

  44. Peppicelli, S., Bianchini, F., Torre, E., & Calorini, L. (2014). Contribution of acidic melanoma cells undergoing epithelial-to-mesenchymal transition to aggressiveness of non-acidic melanoma cells. Clinical & Experimental Metastasis, 31(4), 423–433. https://doi.org/10.1007/s10585-014-9637-6.

    Article  CAS  Google Scholar 

  45. Kalliomäki, T., & Hill, R. P. (2004). Effects of tumour acidification with glucose+MIBG on the spontaneous metastatic potential of two murine cell lines. British Journal of Cancer, 90(9), 1842–1849.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Lora-Michiels, M., Yu, D., Sanders, L., Poulson, J. M., Azuma, C., Case, B., Vujaskovic, Z., Thrall, D. E., Charles, H. C., & Dewhirst, M. W. (2006). Extracellular pH and P-31 magnetic resonance spectroscopic variables are related to outcome in canine soft tissue sarcomas treated with thermoradiotherapy. Clinical Cancer Research, 12(19), 5733–5740. https://doi.org/10.1158/1078-0432.CCR-05-2669.

    Article  CAS  PubMed  Google Scholar 

  47. Wang, L., Fan, Z., Zhang, J., Changyi, Y., Huang, C., Gu, Y., Xu, Z., Tang, Z., Lu, W., Wei, X., & Li, C. (2015). Evaluating tumor metastatic potential by imaging intratumoral acidosis via pH-activatable near-infrared fluorescent probe. International Journal of Cancer, 136(4), E107–E116. https://doi.org/10.1002/ijc.29153.

    Article  CAS  PubMed  Google Scholar 

  48. Estrella, V., Chen, T., Lloyd, M., Wojtkowiak, J., Cornnell, H. H., Ibrahim-Hashim, A., Bailey, K., Balagurunathan, Y., Rothberg, J. M., Sloane, B. F., Johnson, J., Gatenby, R. A., & Gillies, R. J. (2013). Acidity generated by the tumor microenvironment drives local invasion. Cancer Research, 73(5), 1524–1535. https://doi.org/10.1158/0008-5472.CAN-12-2796.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gatenby, R. A., Gawlinski, E. T., Gmitro, A. F., Kaylor, B., & Gillies, R. J. (2006). Acid-mediated tumor invasion: a multidisciplinary study. Cancer Research, 66(10), 5216–5223. https://doi.org/10.1158/0008-5472.CAN-05-4193.

    Article  CAS  PubMed  Google Scholar 

  50. Rozhin, J., Sameni, M., Ziegler, G., & Sloane, B. F. (1994). Pericellular pH affects distribution and secretion of cathepsin B in malignant cells. Cancer Research, 54(24), 6517–6525.

    CAS  PubMed  Google Scholar 

  51. DeClerck, K., & Elble, R. C. (2010). The role of hypoxia and acidosis in promoting metastasis and resistance to chemotherapy. Front Biosci (Landmark Ed), 15, 213–225.

    Article  CAS  Google Scholar 

  52. Gatenby, R. A., & Gillies, R. J. (2004). Why do cancers have high aerobic glycolysis? Nature Reviews. Cancer, 4(11), 891–899. https://doi.org/10.1038/nrc1478.

    Article  CAS  PubMed  Google Scholar 

  53. Lunt, S. J., Chaudary, N., & Hill, R. P. (2009). The tumor microenvironment and metastatic disease. Clinical & Experimental Metastasis, 26(1), 19–34. https://doi.org/10.1007/s10585-008-9182-2.

    Article  Google Scholar 

  54. Payen, V. L., Porporato, P. E., Baselet, B., & Sonveaux, P. (2016). Metabolic changes associated with tumor metastasis, part 1: tumor pH, glycolysis and the pentose phosphate pathway. Cellular and Molecular Life Sciences, 73(7), 1333–1348. https://doi.org/10.1007/s00018-015-2098-5.

    Article  CAS  PubMed  Google Scholar 

  55. Peppicelli, S., Bianchini, F., & Calorini, L. (2014). Extracellular acidity, a “reappreciated” trait of tumor environment driving malignancy: perspectives in diagnosis and therapy. Cancer Metastasis Reviews, 33(2–3), 823–832. https://doi.org/10.1007/s10555-014-9506-4.

    Article  CAS  PubMed  Google Scholar 

  56. Viklund, J., Avnet, S., & De Milito, A. (2017). Pathobiology and therapeutic implications of tumor acidosis. Current Medicinal Chemistry, 24(26), 2827–2845. https://doi.org/10.2174/0929867323666161228142849.

    Article  CAS  PubMed  Google Scholar 

  57. Redfern, A. D., Spalding, L. J., & Thompson, E. W. (2018). The kraken wakes: induced EMT as a driver of tumour aggression and poor outcome. Clinical & Experimental Metastasis, 35(4), 285–308. https://doi.org/10.1007/s10585-018-9906-x.

    Article  Google Scholar 

  58. Diepenbruck, M., & Christofori, G. (2016). Epithelial-mesenchymal transition (EMT) and metastasis: yes, no, maybe? Current Opinion in Cell Biology, 43, 7–13. https://doi.org/10.1016/j.ceb.2016.06.002.

    Article  CAS  PubMed  Google Scholar 

  59. Sciacovelli, M., & Frezza, C. (2017). Metabolic reprogramming and epithelial-to-mesenchymal transition in cancer. The FEBS Journal, 284(19), 3132–3144. https://doi.org/10.1111/febs.14090.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Coupland, L. A., & Parish, C. R. (2014). Platelets, selectins, and the control of tumor metastasis. Seminars in Oncology, 41(3), 422–434. https://doi.org/10.1053/j.seminoncol.2014.04.003.

    Article  CAS  PubMed  Google Scholar 

  61. Yuen, A., & Diaz, B. (2014). The impact of hypoxia in pancreatic cancer invasion and metastasis. Hypoxia (Auckl), 2, 91–106. https://doi.org/10.2147/HP.S52636.

    Article  Google Scholar 

  62. Jung, H. Y., Fattet, L., & Yang, J. (2015). Molecular pathways: linking tumor microenvironment to epithelial-mesenchymal transition in metastasis. Clinical Cancer Research, 21(5), 962–968. https://doi.org/10.1158/1078-0432.CCR-13-3173.

    Article  CAS  PubMed  Google Scholar 

  63. Catalano, V., Turdo, A., Di Franco, S., Dieli, F., Todaro, M., & Stassi, G. (2013). Tumor and its microenvironment: a synergistic interplay. Seminars in Cancer Biology, 23(6 Pt B), 522–532. https://doi.org/10.1016/j.semcancer.2013.08.007.

    Article  CAS  PubMed  Google Scholar 

  64. Riemann, A., Rauschner, M., Gießelmann, M., Reime, S., Haupt, V., & Thews, O. (2018). Extracellular acidosis modulates the expression of epithelial-mesenchymal transition (EMT) markers and adhesion of epithelial and tumor cells. Neoplasia, submitted.

  65. Cabalgante, M. J., Gadola, L., Luzardo, L., Marquez, M., Boggia, J., & Boim, M. A. (2012). Calcium citrate improves the epithelial-to-mesenchymal transition induced by acidosis in proximal tubular cells. Jornal Brasileiro de Nefrologia, 34(4), 343–348.

    Article  PubMed  Google Scholar 

  66. Peppicelli, S., Bianchini, F., Toti, A., Laurenzana, A., Fibbi, G., & Calorini, L. (2015). Extracellular acidity strengthens mesenchymal stem cells to promote melanoma progression. Cell Cycle, 14(19), 3088–3100. https://doi.org/10.1080/15384101.2015.1078032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Corbet, C., & Feron, O. (2017). Tumour acidosis: from the passenger to the driver’s seat. Nature Reviews. Cancer, 17(10), 577–593. https://doi.org/10.1038/nrc.2017.77.

    Article  CAS  PubMed  Google Scholar 

  68. Riemann, A., Reime, S., & Thews, O. (2018). Acidic extracellular environment affects miRNA expression in tumors in vitro and in vivo. International Journal of Cancer. https://doi.org/10.1002/ijc.31790.

  69. Chen, K. H., Tung, P. Y., Wu, J. C., Chen, Y., Chen, P. C., Huang, S. H., & Wang, S. M. (2008). An acidic extracellular pH induces Src kinase-dependent loss of ß-catenin from the adherens junction. Cancer Letters, 267(1), 37–48. https://doi.org/10.1016/j.canlet.2008.03.005.

    Article  CAS  PubMed  Google Scholar 

  70. Stock, C., Gassner, B., Hauck, C. R., Arnold, H., Mally, S., Eble, J. A., Dieterich, P., & Schwab, A. (2005). Migration of human melanoma cells depends on extracellular pH and Na+/H+ exchange. The Journal of Physiology, 567(Pt 1), 225–238. https://doi.org/10.1113/jphysiol.2005.088344.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li, S., Xiong, N., Peng, Y., Tang, K., Bai, H., Lv, X., Jiang, Y., Qin, X., Yang, H., Wu, C., Zhou, P., & Liu, Y. (2018). Acidic pHe regulates cytoskeletal dynamics through conformational integrin ß1 activation and promotes membrane protrusion. Biochimica et Biophysica Acta - Molecular Basis of Disease, 1864(7), 2395–2408. https://doi.org/10.1016/j.bbadis.2018.04.019.

    Article  CAS  PubMed  Google Scholar 

  72. Paradise, R. K., Lauffenburger, D. A., & Van Vliet, K. J. (2011). Acidic extracellular pH promotes activation of integrin ɑvß3. PLoS One, 6(1), e15746. https://doi.org/10.1371/journal.pone.0015746.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Chen, A., Beetham, H., Black, M. A., Priya, R., Telford, B. J., Guest, J., Wiggins, G. A. R., Godwin, T. D., Yap, A. S., & Guilford, P. J. (2014). E-cadherin loss alters cytoskeletal organization and adhesion in non-malignant breast cells but is insufficient to induce an epithelial-mesenchymal transition. BMC Cancer, 14, 552. https://doi.org/10.1186/1471-2407-14-552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hofschröer, V., Koch, K. A., Ludwig, F. T., Friedl, P., Oberleithner, H., Stock, C., & Schwab, A. (2017). Extracellular protonation modulates cell-cell interaction mechanics and tissue invasion in human melanoma cells. Scientific Reports, 7, 42369. https://doi.org/10.1038/srep42369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chen, C. H., Lin, H., Chuang, S. M., Lin, S. Y., & Chen, J. J. (2010). Acidic stress facilitates tyrosine phosphorylation of HLJ1 to associate with actin cytoskeleton in lung cancer cells. Experimental Cell Research, 316(17), 2910–2921. https://doi.org/10.1016/j.yexcr.2010.06.027.

    Article  CAS  PubMed  Google Scholar 

  76. Schwab, A., Fabian, A., Hanley, P. J., & Stock, C. (2012). Role of ion channels and transporters in cell migration. Physiological Reviews, 92(4), 1865–1913. https://doi.org/10.1152/physrev.00018.2011.

    Article  CAS  PubMed  Google Scholar 

  77. Wu, Y., Gao, B., Xiong, Q. J., Wang, Y. C., Huang, D. K., & Wu, W. N. (2017). Acid-sensing ion channels contribute to the effect of extracellular acidosis on proliferation and migration of A549 cells. Tumour Biology, 39(6), 1010428317705750. https://doi.org/10.1177/1010428317705750.

    Article  PubMed  Google Scholar 

  78. Sudhan, D. R., & Siemann, D. W. (2013). Cathepsin L inhibition by the small molecule KGP94 suppresses tumor microenvironment enhanced metastasis associated cell functions of prostate and breast cancer cells. Clinical & Experimental Metastasis, 30(7), 891–902. https://doi.org/10.1007/s10585-013-9590-9.

    Article  CAS  Google Scholar 

  79. Parks, S. K., & Pouysségur, J. (2015). The Na+/HCO3 co-transporter SLC4A4 plays a role in growth and migration of colon and breast cancer cells. Journal of Cellular Physiology, 230(8), 1954–1963. https://doi.org/10.1002/jcp.24930.

    Article  CAS  PubMed  Google Scholar 

  80. Moellering, R. E., Black, K. C., Krishnamurty, C., Baggett, B. K., Stafford, P., Rain, M., Gatenby, R. A., & Gillies, R. J. (2008). Acid treatment of melanoma cells selects for invasive phenotypes. Clinical & Experimental Metastasis, 25(4), 411–425. https://doi.org/10.1007/s10585-008-9145-7.

    Article  CAS  Google Scholar 

  81. Riemann, A., Schneider, B., Gündel, D., Stock, C., Gekle, M., & Thews, O. (2016). Acidosis promotes metastasis formation by enhancing tumor cell motility. Advances in Experimental Medicine and Biology, 876, 215–220. https://doi.org/10.1007/978-1-4939-3023-4_27.

    Article  CAS  PubMed  Google Scholar 

  82. Svastova, E., & Pastorekova, S. (2013). Carbonic anhydrase IX: a hypoxia-controlled “catalyst” of cell migration. Cell Adhesion & Migrationa, 7(2), 226–231. https://doi.org/10.4161/cam.23257.

    Article  Google Scholar 

  83. McDonald, P. C., Swayampakula, M., & Dedhar, S. (2018). Coordinated regulation of metabolic transporters and migration/invasion by carbonic anhydrase IX. Metabolites, 8(1), 20. https://doi.org/10.3390/metabo8010020.

  84. Csaderova, L., Debreova, M., Radvak, P., Stano, M., Vrestiakova, M., Kopacek, J., Pastorekova, S., & Svastova, E. (2013). The effect of carbonic anhydrase IX on focal contacts during cell spreading and migration. Frontiers in Physiology, 4, 271. https://doi.org/10.3389/fphys.2013.00271.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Zhou, Z. H., Song, J. W., Li, W., Liu, X., Cao, L., Wan, L. M., Tan, Y. X., Ji, S. P., Liang, Y. M., & Gong, F. (2017). The acid-sensing ion channel, ASIC2, promotes invasion and metastasis of colorectal cancer under acidosis by activating the calcineurin/NFAT1 axis. Journal of Experimental & Clinical Cancer Research, 36(1), 130. https://doi.org/10.1186/s13046-017-0599-9.

    Article  Google Scholar 

  86. Ahn, H., Kim, J. M., Lee, K., Kim, H., & Jeong, D. (2012). Extracellular acidosis accelerates bone resorption by enhancing osteoclast survival, adhesion, and migration. Biochemical and Biophysical Research Communications, 418(1), 144–148. https://doi.org/10.1016/j.bbrc.2011.12.149.

    Article  CAS  PubMed  Google Scholar 

  87. Srivastava, J., Barreiro, G., Groscurth, S., Gingras, A. R., Goult, B. T., Critchley, D. R., Kelly, M. J. S., Jacobson, M. P., & Barber, D. L. (2008). Structural model and functional significance of pH-dependent talin-actin binding for focal adhesion remodeling. Proceedings of the National Academy of Sciences of the United States of America, 105(38), 14436–14441. https://doi.org/10.1073/pnas.0805163105.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Pope, B. J., Zierler-Gould, K. M., Kühne, R., Weeds, A. G., & Ball, L. J. (2004). Solution structure of human cofilin: actin binding, pH sensitivity, and relationship to actin-depolymerizing factor. The Journal of Biological Chemistry, 279(6), 4840–4848. https://doi.org/10.1074/jbc.M310148200.

    Article  CAS  PubMed  Google Scholar 

  89. Sidani, M., Wessels, D., Mouneimne, G., Ghosh, M., Goswami, S., Sarmiento, C., Wang, W., Kuhl, S., el-Sibai, M., Backer, J. M., Eddy, R., Soll, D., & Condeelis, J. (2007). Cofilin determines the migration behavior and turning frequency of metastatic cancer cells. The Journal of Cell Biology, 179(4), 777–791. https://doi.org/10.1083/jcb.200707009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Huang, S., He, P., Xu, D., Li, J., Peng, X., & Tang, Y. (2017). Acidic stress induces apoptosis and inhibits angiogenesis in human bone marrow-derived endothelial progenitor cells. Oncology Letters, 14(5), 5695–5702. https://doi.org/10.3892/ol.2017.6947.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Kessenbrock, K., Plaks, V., & Werb, Z. (2010). Matrix metalloproteinases: regulators of the tumor microenvironment. Cell, 141(1), 52–67. https://doi.org/10.1016/j.cell.2010.03.015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Vandooren, J., Van den Steen, P. E., & Opdenakker, G. (2013). Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9): the next decade. Critical Reviews in Biochemistry and Molecular Biology, 48(3), 222–272. https://doi.org/10.3109/10409238.2013.770819.

    Article  CAS  PubMed  Google Scholar 

  93. Kato, Y., Ozawa, S., Tsukuda, M., Kubota, E., Miyazaki, K., St-Pierre, Y., & Hata, R. I. (2007). Acidic extracellular pH increases calcium influx-triggered phospholipase D activity along with acidic sphingomyelinase activation to induce matrix metalloproteinase-9 expression in mouse metastatic melanoma. The FEBS Journal, 274(12), 3171–3183. https://doi.org/10.1111/j.1742-4658.2007.05848.x.

    Article  CAS  PubMed  Google Scholar 

  94. Kato, Y., Lambert, C. A., Colige, A. C., Mineur, P., Noel, A., Frankenne, F., Foidart, J. M., Baba, M., Hata, R. I., Miyazaki, K., & Tsukuda, M. (2005). Acidic extracellular pH induces matrix metalloproteinase-9 expression in mouse metastatic melanoma cells through the phospholipase D-mitogen-activated protein kinase signaling. The Journal of Biological Chemistry, 280(12), 10938–10944. https://doi.org/10.1074/jbc.M411313200.

    Article  CAS  PubMed  Google Scholar 

  95. Matsubara, T., DiResta, G. R., Kakunaga, S., Li, D., & Healey, J. H. (2013). Additive influence of extracellular pH, oxygen tension, and pressure on invasiveness and survival of human osteosarcoma cells. Frontiers in Oncology, 3, 199. https://doi.org/10.3389/fonc.2013.00199.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Giusti, I., D'Ascenzo, S., Millimaggi, D., Taraboletti, G., Carta, G., Franceschini, N., et al. (2008). Cathepsin B mediates the pH-dependent proinvasive activity of tumor-shed microvesicles. Neoplasia, 10(5), 481–488.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Huang, S., Tang, Y., Peng, X., Cai, X., Wa, Q., Ren, D., Li, Q., Luo, J., Li, L., Zou, X., & Huang, S. (2016). Acidic extracellular pH promotes prostate cancer bone metastasis by enhancing PC-3 stem cell characteristics, cell invasiveness and VEGF-induced vasculogenesis of BM-EPCs. Oncology Reports, 36(4), 2025–2032. https://doi.org/10.3892/or.2016.4997.

    Article  CAS  PubMed  Google Scholar 

  98. Martínez-Zaguilán, R., Seftor, E. A., Seftor, R. E., Chu, Y. W., Gillies, R. J., & Hendrix, M. J. (1996). Acidic pH enhances the invasive behavior of human melanoma cells. Clinical & Experimental Metastasis, 14(2), 176–186.

    Article  Google Scholar 

  99. Roshy, S., Sloane, B. F., & Moin, K. (2003). Pericellular cathepsin B and malignant progression. Cancer Metastasis Reviews, 22(2–3), 271–286.

    Article  CAS  PubMed  Google Scholar 

  100. Jang, A., & Hill, R. P. (1997). An examination of the effects of hypoxia, acidosis, and glucose starvation on the expression of metastasis-associated genes in murine tumor cells. Clinical & Experimental Metastasis, 15(5), 469–483.

    Article  CAS  Google Scholar 

  101. Fukumura, D., Xu, L., Chen, Y., Gohongi, T., Seed, B., & Jain, R. K. (2001). Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Research, 61(16), 6020–6024.

    CAS  PubMed  Google Scholar 

  102. Shi, Q., Le, X., Wang, B., Abbruzzese, J. L., Xiong, Q., He, Y., et al. (2001). Regulation of vascular endothelial growth factor expression by acidosis in human cancer cells. Oncogene, 20(28), 3751–3756. https://doi.org/10.1038/sj.onc.1204500.

    Article  CAS  PubMed  Google Scholar 

  103. Xie, K., Wei, D., Shi, Q., & Huang, S. (2004). Constitutive and inducible expression and regulation of vascular endothelial growth factor. Cytokine & Growth Factor Reviews, 15(5), 297–324. https://doi.org/10.1016/j.cytogfr.2004.04.003.

    Article  CAS  Google Scholar 

  104. Elias, A. P., & Dias, S. (2008). Microenvironment changes (in pH) affect VEGF alternative splicing. Cancer Microenvironment, 1(1), 131–139. https://doi.org/10.1007/s12307-008-0013-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wyder, L., Suply, T., Ricoux, B., Billy, E., Schnell, C., Baumgarten, B. U., Maira, S. M., Koelbing, C., Ferretti, M., Kinzel, B., Müller, M., Seuwen, K., & Ludwig, M. G. (2011). Reduced pathological angiogenesis and tumor growth in mice lacking GPR4, a proton sensing receptor. Angiogenesis, 14(4), 533–544. https://doi.org/10.1007/s10456-011-9238-9.

    Article  CAS  PubMed  Google Scholar 

  106. Jing, Z., Xu, H., Chen, X., Zhong, Q., Huang, J., Zhang, Y., Guo, W., Yang, Z., Ding, S., Chen, P., & Huang, Z. (2016). The proton-sensing G-protein coupled receptor GPR4 promotes angiogenesis in head and neck cancer. PLoS One, 11(4), e0152789. https://doi.org/10.1371/journal.pone.0152789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Mena, H. A., Lokajczyk, A., Dizier, B., Strier, S. E., Voto, L. S., Boisson-Vidal, C., Schattner, M., & Negrotto, S. (2014). Acidic preconditioning improves the proangiogenic responses of endothelial colony forming cells. Angiogenesis, 17(4), 867–879. https://doi.org/10.1007/s10456-014-9434-5.

    Article  CAS  PubMed  Google Scholar 

  108. Peppicelli, S., Bianchini, F., Contena, C., Tombaccini, D., & Calorini, L. (2013). Acidic pH via NF-kappaB favours VEGF-C expression in human melanoma cells. Clinical & Experimental Metastasis, 30(8), 957–967. https://doi.org/10.1007/s10585-013-9595-4.

    Article  CAS  Google Scholar 

  109. Nakanishi, M., Morita, Y., Hata, K., & Muragaki, Y. (2016). Acidic microenvironments induce lymphangiogenesis and IL-8 production via TRPV1 activation in human lymphatic endothelial cells. Experimental Cell Research, 345(2), 180–189. https://doi.org/10.1016/j.yexcr.2016.06.006.

    Article  CAS  PubMed  Google Scholar 

  110. Schwab, A., & Stock, C. (2014). Ion channels and transporters in tumour cell migration and invasion. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 369(1638), 20130102. https://doi.org/10.1098/rstb.2013.0102.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Stock, C., & Schwab, A. (2015). Ion channels and transporters in metastasis. Biochim Biophys Acta, 1848(10 Pt B), 2638–2646. https://doi.org/10.1016/j.bbamem.2014.11.012.

    Article  CAS  PubMed  Google Scholar 

  112. Jin, C., Ye, Q. H., Yuan, F. L., Gu, Y. L., Li, J. P., Shi, Y. H., Shen, X. M., Bo-Liu, & Lin, Z. H. (2015). Involvement of acid-sensing ion channel 1ɑ in hepatic carcinoma cell migration and invasion. Tumour Biology, 36(6), 4309–4317. https://doi.org/10.1007/s13277-015-3070-6.

    Article  CAS  PubMed  Google Scholar 

  113. Li, X., Ye, J. X., Xu, M. H., Zhao, M. D., & Yuan, F. L. (2017). Evidence that activation of ASIC1a by acidosis increases osteoclast migration and adhesion by modulating integrin/Pyk2/Src signaling pathway. Osteoporosis International, 28(7), 2221–2231. https://doi.org/10.1007/s00198-017-4017-0.

    Article  CAS  PubMed  Google Scholar 

  114. Kato, Y., Ozawa, S., Miyamoto, C., Maehata, Y., Suzuki, A., Maeda, T., & Baba, Y. (2013). Acidic extracellular microenvironment and cancer. Cancer Cell International, 13(1), 89. https://doi.org/10.1186/1475-2867-13-89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Maeda, T., Suzuki, A., Koga, K., Miyamoto, C., Maehata, Y., Ozawa, S., Hata, R. I., Nagashima, Y., Nabeshima, K., Miyazaki, K., & Kato, Y. (2017). TRPM5 mediates acidic extracellular pH signaling and TRPM5 inhibition reduces spontaneous metastasis in mouse B16-BL6 melanoma cells. Oncotarget, 8(45), 78312–78326. https://doi.org/10.18632/oncotarget.20826.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Stock, C., Mueller, M., Kraehling, H., Mally, S., Noel, J., Eder, C., et al. (2007). pH nanoenvironment at the surface of single melanoma cells. Cellular Physiology and Biochemistry, 20(5), 679–686. https://doi.org/10.1159/000107550.

    Article  CAS  PubMed  Google Scholar 

  117. Martin, C., Pedersen, S. F., Schwab, A., & Stock, C. (2011). Intracellular pH gradients in migrating cells. American Journal of Physiology. Cell Physiology, 300(3), C490–C495. https://doi.org/10.1152/ajpcell.00280.2010.

    Article  CAS  PubMed  Google Scholar 

  118. Ludwig, F. T., Schwab, A., & Stock, C. (2013). The Na+ /H+ −exchanger (NHE1) generates pH nanodomains at focal adhesions. Journal of Cellular Physiology, 228(6), 1351–1358. https://doi.org/10.1002/jcp.24293.

    Article  CAS  PubMed  Google Scholar 

  119. Vahle, A. K., Domikowsky, B., Schwoppe, C., Krahling, H., Mally, S., Schafers, M., et al. (2014). Extracellular matrix composition and interstitial pH modulate NHE1-mediated melanoma cell motility. International Journal of Oncology, 44(1), 78–90. https://doi.org/10.3892/ijo.2013.2158.

    Article  CAS  PubMed  Google Scholar 

  120. Brackenbury, W. J. (2012). Voltage-gated sodium channels and metastatic disease. Channels (Austin, Tex.), 6(5), 352–361. https://doi.org/10.4161/chan.21910.

    Article  CAS  Google Scholar 

  121. Nassios, A., Wallner, S., Haferkamp, S., Klingelhoffer, C., Brochhausen, C., & Schreml, S. (2018). Expression of proton-sensing G-protein-coupled receptors in selected skin tumors. Experimental Dermatology. https://doi.org/10.1111/exd.13809.

  122. Damaghi, M., Wojtkowiak, J. W., & Gillies, R. J. (2013). pH sensing and regulation in cancer. Frontiers in Physiology, 4, 370. https://doi.org/10.3389/fphys.2013.00370.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Justus, C. R., Dong, L., & Yang, L. V. (2013). Acidic tumor microenvironment and pH-sensing G protein-coupled receptors. Frontiers in Physiology, 4, 354. https://doi.org/10.3389/fphys.2013.00354.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Justus, C. R., & Yang, L. V. (2015). GPR4 decreases B16F10 melanoma cell spreading and regulates focal adhesion dynamics through the G13/Rho signaling pathway. Experimental Cell Research, 334(1), 100–113. https://doi.org/10.1016/j.yexcr.2015.03.022.

    Article  CAS  PubMed  Google Scholar 

  125. Castellone, R. D., Leffler, N. R., Dong, L., & Yang, L. V. (2011). Inhibition of tumor cell migration and metastasis by the proton-sensing GPR4 receptor. Cancer Letters, 312(2), 197–208. https://doi.org/10.1016/j.canlet.2011.08.013.

    Article  CAS  PubMed  Google Scholar 

  126. de Valliere, C., Cosin-Roger, J., Simmen, S., Atrott, K., Melhem, H., Zeitz, J., et al. (2016). Hypoxia positively regulates the expression of pH-sensing G-protein-coupled receptor OGR1 (GPR68). Cellular and Molecular Gastroenterology and Hepatology, 2(6), 796–810. https://doi.org/10.1016/j.jcmgh.2016.06.003.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Singh, L. S., Berk, M., Oates, R., Zhao, Z., Tan, H., Jiang, Y., Zhou, A., Kirmani, K., Steinmetz, R., Lindner, D., & Xu, Y. (2007). Ovarian cancer G protein-coupled receptor 1, a new metastasis suppressor gene in prostate cancer. Journal of the National Cancer Institute, 99(17), 1313–1327. https://doi.org/10.1093/jnci/djm107.

    Article  CAS  PubMed  Google Scholar 

  128. Li, J., Guo, B., Wang, J., Cheng, X., Xu, Y., & Sang, J. (2013). Ovarian cancer G protein coupled receptor 1 suppresses cell migration of MCF7 breast cancer cells via a Galpha12/13-Rho-Rac1 pathway. Journal of Molecular Signaling, 8(1), 6. https://doi.org/10.1186/1750-2187-8-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Chen, P., Zuo, H., Xiong, H., Kolar, M. J., Chu, Q., Saghatelian, A., Siegwart, D. J., & Wan, Y. (2017). Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proceedings of the National Academy of Sciences of the United States of America, 114(3), 580–585. https://doi.org/10.1073/pnas.1614035114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Wu, W. S., Wu, J. R., & Hu, C. T. (2008). Signal cross talks for sustained MAPK activation and cell migration: the potential role of reactive oxygen species. Cancer Metastasis Reviews, 27(2), 303–314. https://doi.org/10.1007/s10555-008-9112-4.

    Article  CAS  PubMed  Google Scholar 

  131. Sauvant, C., Nowak, M., Wirth, C., Schneider, B., Riemann, A., Gekle, M., & Thews, O. (2008). Acidosis induces multi-drug resistance in rat prostate cancer cells (AT1) in vitro and in vivo by increasing the activity of the p-glycoprotein via activation of p38. International Journal of Cancer, 123(11), 2532–2542.

    Article  CAS  PubMed  Google Scholar 

  132. Riemann, A., Reime, S., & Thews, O. (2017). Tumor acidosis and hypoxia differently modulate the inflammatory program: measurements in vitro and in vivo. Neoplasia, 19(12), 1033–1042. https://doi.org/10.1016/j.neo.2017.09.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Sarosi, G. A., Jr., Jaiswal, K., Herndon, E., Lopez-Guzman, C., Spechler, S. J., & Souza, R. F. (2005). Acid increases MAPK-mediated proliferation in Barrett's esophageal adenocarcinoma cells via intracellular acidification through a Cl-/HCO3- exchanger. American Journal of Physiology. Gastrointestinal and Liver Physiology, 289(6), G991–G997. https://doi.org/10.1152/ajpgi.00215.2005.

    Article  CAS  PubMed  Google Scholar 

  134. Riemann, A., Schneider, B., Ihling, A., Nowak, M., Sauvant, C., Thews, O., & Gekle, M. (2011). Acidic environment leads to ROS-induced MAPK signaling in cancer cells. PLoS One, 6(7), e22445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Riemann, A., Ihling, A., Schneider, B., Gekle, M., & Thews, O. (2013). Impact of extracellular acidosis on intracellular pH control and cell signaling in tumor cells. Advances in Experimental Medicine and Biology, 789, 221–228.

    Article  CAS  PubMed  Google Scholar 

  136. Riemann, A., Ihling, A., Thomas, J., Schneider, B., Thews, O., & Gekle, M. (2015). Acidic environment activates inflammatory programs in fibroblasts via a cAMP-MAPK pathway. Biochim. Biophys. Acta, 1853(2), 299–307. https://doi.org/10.1016/j.bbamcr.2014.11.022.

    Article  CAS  PubMed  Google Scholar 

  137. Bischoff, D. S., Zhu, J. H., Makhijani, N. S., & Yamaguchi, D. T. (2008). Acidic pH stimulates the production of the angiogenic CXC chemokine, CXCL8 (interleukin-8), in human adult mesenchymal stem cells via the extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, and NF-κB pathways. J Cell Biochem, 104(4), 1378–1392. https://doi.org/10.1002/jcb.21714.

    Article  CAS  PubMed  Google Scholar 

  138. Chen, B., Liu, J., Ho, T. T., Ding, X., & Mo, Y. Y. (2016). ERK-mediated NF-κB activation through ASIC1 in response to acidosis. Oncogenesis, 5(12), e279. https://doi.org/10.1038/oncsis.2016.81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Xu, L., Fukumura, D., & Jain, R. K. (2002). Acidic extracellular pH induces vascular endothelial growth factor (VEGF) in human glioblastoma cells via ERK1/2 MAPK signaling pathway: mechanism of low pH-induced VEGF. The Journal of Biological Chemistry, 277(13), 11,368–11,374. https://doi.org/10.1074/jbc.M108347200.

    Article  CAS  Google Scholar 

  140. Wu, W. S. (2006). The signaling mechanism of ROS in tumor progression. Cancer Metastasis Rev, 25(4), 695–705.

    Article  CAS  PubMed  Google Scholar 

  141. Tochhawng, L., Deng, S., Pervaiz, S., & Yap, C. T. (2013). Redox regulation of cancer cell migration and invasion. Mitochondrion, 13(3), 246–253. https://doi.org/10.1016/j.mito.2012.08.002.

    Article  CAS  PubMed  Google Scholar 

  142. Payne, S. L., Fogelgren, B., Hess, A. R., Seftor, E. A., Wiley, E. L., Fong, S. F., et al. (2005). Lysyl oxidase regulates breast cancer cell migration and adhesion through a hydrogen peroxide-mediated mechanism. Cancer Res, 65(24), 11,429–11,436. https://doi.org/10.1158/0008-5472.CAN-05-1274.

    Article  CAS  Google Scholar 

  143. Tobar, N., Guerrero, J., Smith, P. C., & Martinez, J. (2010). NOX4-dependent ROS production by stromal mammary cells modulates epithelial MCF-7 cell migration. British Journal of Cancer, 103(7), 1040–1047. https://doi.org/10.1038/sj.bjc.6605847.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Mori, K., Shibanuma, M., & Nose, K. (2004). Invasive potential induced under long-term oxidative stress in mammary epithelial cells. Cancer Res, 64(20), 7464–7472.

    Article  CAS  PubMed  Google Scholar 

  145. Riemann, A., Ihling, A., Reime, S., Gekle, M., & Thews, O. (2016). Impact of the tumor microenvironment on the expression of inflammatory mediators in cancer cells. Advances in Experimental Medicine and Biology, 923, 105–111. https://doi.org/10.1007/978-3-319-38,810-6_14.

    Article  CAS  PubMed  Google Scholar 

  146. Pan, B., Ren, H., Lv, X., Zhao, Y., Yu, B., He, Y., et al. (2012). Hypochlorite-induced oxidative stress elevates the capability of HDL in promoting breast cancer metastasis. Journal of Translational Medicine, 10, 65. https://doi.org/10.1186/1479-5876-10-65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Xu, J., Peng, Z., Li, R., Dou, T., Xu, W., Gu, G., et al. (2009). Normoxic induction of cerebral HIF-1ɑ by acetazolamide in rats: role of acidosis. Neuroscience Letters, 451(3), 274–278. https://doi.org/10.1016/j.neulet.2009.01.008.

    Article  CAS  PubMed  Google Scholar 

  148. Nadtochiy, S. M., Schafer, X., Fu, D., Nehrke, K., Munger, J., & Brookes, P. S. (2016). Acidic pH is a metabolic switch for 2-hydroxyglutarate generation and signaling. The Journal of Biological Chemistry, 291(38), 20,188–20,197. https://doi.org/10.1074/jbc.M116.738799.

    Article  CAS  Google Scholar 

  149. Melchionna, R., Romani, M., Ambrosino, V., D’Arcangelo, D., Cencioni, C., Porcelli, D., et al. (2010). Role of HIF-1α in proton-mediated CXCR4 down-regulation in endothelial cells. Cardiovascular Research, 86(2), 293–301. https://doi.org/10.1093/cvr/cvp393.

    Article  CAS  PubMed  Google Scholar 

  150. Mekhail, K., Khacho, M., Gunaratnam, L., & Lee, S. (2004). Oxygen sensing by H+: implications for HIF and hypoxic cell memory. Cell Cycle, 3(8), 1027–1029.

    Article  CAS  PubMed  Google Scholar 

  151. Mekhail, K., Gunaratnam, L., Bonicalzi, M. E., & Lee, S. (2004). HIF activation by pH-dependent nucleolar sequestration of VHL. Nature Cell Biology, 6(7), 642–647. https://doi.org/10.1038/ncb1144.

    Article  CAS  PubMed  Google Scholar 

  152. Filatova, A., Seidel, S., Bogurcu, N., Graf, S., Garvalov, B. K., & Acker, T. (2016). Acidosis acts through HSP90 in a PHD/VHL-independent manner to promote HIF function and stem cell maintenance in glioma. Cancer Research, 76(19), 5845–5856. https://doi.org/10.1158/0008-5472.CAN-15-2630.

    Article  CAS  PubMed  Google Scholar 

  153. Willam, C., Warnecke, C., Schefold, J. C., Kügler, J., Koehne, P., Frei, U., et al. (2006). Inconsistent effects of acidosis on HIF-alpha protein and its target genes. Pflugers Archiv, 451(4), 534–543. https://doi.org/10.1007/s00424-005-1486-3.

    Article  CAS  PubMed  Google Scholar 

  154. Tang, X., Lucas, J. E., Chen, J. L., LaMonte, G., Wu, J., Wang, M. C., et al. (2012). Functional interaction between responses to lactic acidosis and hypoxia regulates genomic transcriptional outputs. Cancer Research, 72(2), 491–502. https://doi.org/10.1158/0008-5472.CAN-11-2076.

    Article  CAS  PubMed  Google Scholar 

  155. Braeuer, R. R., Zigler, M., Villares, G. J., Dobroff, A. S., & Bar-Eli, M. (2011). Transcriptional control of melanoma metastasis: the importance of the tumor microenvironment. Seminars in Cancer Biology, 21(2), 83–88. https://doi.org/10.1016/j.semcancer.2010.12.007.

    Article  CAS  PubMed  Google Scholar 

  156. Oehlke, O., Schlosshardt, C., Feuerstein, M., & Roussa, E. (2012). Acidosis-induced V-ATPase trafficking in salivary ducts is initiated by cAMP/PKA/CREB pathway via regulation of Rab11b expression. The International Journal of Biochemistry & Cell Biology, 44(8), 1254–1265. https://doi.org/10.1016/j.biocel.2012.04.018.

    Article  CAS  Google Scholar 

  157. Nakamura, K., Kamouchi, M., Arimura, K., Nishimura, A., Kuroda, J., Ishitsuka, K., et al. (2012). Extracellular acidification activates cAMP responsive element binding protein via Na+/H+ exchanger isoform 1-mediated Ca2+ oscillation in central nervous system pericytes. Arteriosclerosis, Thrombosis, and Vascular Biology, 32(11), 2670–2677. https://doi.org/10.1161/ATVBAHA.112.254946.

    Article  CAS  PubMed  Google Scholar 

  158. Sin, W. C., Zhang, Y., Zhong, W., Adhikarakunnathu, S., Powers, S., Hoey, T., et al. (2004). G protein-coupled receptors GPR4 and TDAG8 are oncogenic and overexpressed in human cancers. Oncogene, 23(37), 6299–6303. https://doi.org/10.1038/sj.onc.1207838.

    Article  CAS  PubMed  Google Scholar 

  159. Wang, J. Q., & Wu, K. J. (2015). Epigenetic regulation of epithelial-mesenchymal transition by hypoxia in cancer: targets and therapy. Current Pharmaceutical Design, 21(10), 1272–1278.

    Article  CAS  PubMed  Google Scholar 

  160. Bavelloni, A., Ramazzotti, G., Poli, A., Piazzi, M., Focaccia, E., Blalock, W., et al. (2017). MiRNA-210: A Current Overview. Anticancer Research, 37(12), 6511–6521. https://doi.org/10.21873/anticanres.12107.

    Article  CAS  PubMed  Google Scholar 

  161. Choudhry, H., Harris, A. L., & McIntyre, A. (2016). The tumour hypoxia induced non-coding transcriptome. Molecular Aspects of Medicine, 47–48, 35–53. https://doi.org/10.1016/j.mam.2016.01.003.

    Article  CAS  PubMed  Google Scholar 

  162. Rupaimoole, R., Calin, G. A., Lopez-Berestein, G., & Sood, A. K. (2016). miRNA Deregulation in cancer cells and the tumor microenvironment. Cancer Discovery, 6(3), 235–246. https://doi.org/10.1158/2159-8290.CD-15-0893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Huang, X., Ding, L., Bennewith, K. L., Tong, R. T., Welford, S. M., Ang, K. K., et al. (2009). Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Molecular Cell, 35(6), 856–867. https://doi.org/10.1016/j.molcel.2009.09.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Wentz-Hunter, K. K., & Potashkin, J. A. (2011). The role of miRNAs as key regulators in the neoplastic microenvironment. Molecular Biology International, 839, 872. https://doi.org/10.4061/2011/839872.

    Article  CAS  Google Scholar 

  165. Schanza, L. M., Seles, M., Stotz, M., Fosselteder, J., Hutterer, G. C., Pichler, M., et al. (2017). MicroRNAs associated with Von Hippel-Lindau pathway in renal cell carcinoma: a comprehensive review. International Journal of Molecular Sciences, 18(11), E2495. https://doi.org/10.3390/ijms18112495.

    Article  CAS  PubMed  Google Scholar 

  166. Qin, Q., Furong, W., & Baosheng, L. (2014). Multiple functions of hypoxia-regulated miR-210 in cancer. Journal of Experimental & Clinical Cancer Research, 33, 50. https://doi.org/10.1186/1756-9966-33-50.

    Article  CAS  Google Scholar 

  167. Zhang, S., Wang, W., Liu, G., Xie, S., Li, Q., Li, Y., et al. (2017). Long non-coding RNA HOTTIP promotes hypoxia-induced epithelial-mesenchymal transition of malignant glioma by regulating the miR-101/ZEB1 axis. Biomedicine & Pharmacotherapy, 95, 711–720. https://doi.org/10.1016/j.biopha.2017.08.133.

    Article  CAS  Google Scholar 

  168. Riemann, A., Reime, S., & Thews, O. (2017). Hypoxia-related tumor acidosis affects microRNA expression pattern in prostate and breast tumor cells. Advances in Experimental Medicine and Biology, 977, 119–124. https://doi.org/10.1007/978-3-319-55,231-6_16.

    Article  CAS  PubMed  Google Scholar 

  169. Miao, L., Xiong, X., Lin, Y., Cheng, Y., Lu, J., Zhang, J., et al. (2014). miR-203 inhibits tumor cell migration and invasion via caveolin-1 in pancreatic cancer cells. Oncology Letters, 7(3), 658–662. https://doi.org/10.3892/ol.2014.1807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Wang, N., Liang, H., Zhou, Y., Wang, C., Zhang, S., Pan, Y., et al. (2014). miR-203 suppresses the proliferation and migration and promotes the apoptosis of lung cancer cells by targeting SRC. PLoS One, 9(8), e105570. https://doi.org/10.1371/journal.pone.0105570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Ding, X., Park, S. I., McCauley, L. K., & Wang, C. Y. (2013). Signaling between transforming growth factor ß (TGF-ß) and transcription factor SNAI2 represses expression of microRNA miR-203 to promote epithelial-mesenchymal transition and tumor metastasis. The Journal of Biological Chemistry, 288(15), 10,241–10,253. https://doi.org/10.1074/jbc.M112.443655.

    Article  CAS  Google Scholar 

  172. Vohwinkel, C. U., Lecuona, E., Sun, H., Sommer, N., Vadasz, I., Chandel, N. S., et al. (2011). Elevated CO2 levels cause mitochondrial dysfunction and impair cell proliferation. The Journal of Biological Chemistry, 286(43), 37,067–37,076. https://doi.org/10.1074/jbc.M111.290056.

    Article  CAS  Google Scholar 

  173. Lu, Y. Y., Zheng, J. Y., Liu, J., Huang, C. L., Zhang, W., & Zeng, Y. (2015). miR-183 induces cell proliferation, migration, and invasion by regulating PDCD4 expression in the SW1990 pancreatic cancer cell line. Biomedicine & Pharmacotherapy, 70, 151–157. https://doi.org/10.1016/j.biopha.2015.01.016.

    Article  CAS  Google Scholar 

  174. Fan, D., Wang, Y., Qi, P., Chen, Y., Xu, P., Yang, X., et al. (2016). MicroRNA-183 functions as the tumor suppressor via inhibiting cellular invasion and metastasis by targeting MMP-9 in cervical cancer. Gynecologic Oncology, 141(1), 166–174. https://doi.org/10.1016/j.ygyno.2016.02.006.

    Article  CAS  PubMed  Google Scholar 

  175. Wang, J., Wang, X., Li, Z., Liu, H., & Teng, Y. (2014). MicroRNA-183 suppresses retinoblastoma cell growth, invasion and migration by targeting LRP6. FEBS Journal, 281(5), 1355–1365. https://doi.org/10.1111/febs.12659.

    Article  CAS  PubMed  Google Scholar 

  176. Cai, X., Peng, D., Wei, H., Yang, X., Huang, Q., Lin, Z., et al. (2017). miR-215 suppresses proliferation and migration of non-small cell lung cancer cells. Oncology Letters, 13(4), 2349–2353. https://doi.org/10.3892/ol.2017.5692.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Khella, H. W., Bakhet, M., Allo, G., Jewett, M. A., Girgis, A. H., Latif, A., et al. (2013). miR-192, miR-194 and miR-215: a convergent microRNA network suppressing tumor progression in renal cell carcinoma. Carcinogenesis, 34(10), 2231–2239. https://doi.org/10.1093/carcin/bgt184.

    Article  CAS  PubMed  Google Scholar 

  178. Koltai, T. (2016). Cancer: fundamentals behind pH targeting and the double-edged approach. Onco Targets and Therapy, 9, 6343–6360. https://doi.org/10.2147/OTT.S115438.

    Article  CAS  Google Scholar 

  179. Silva, A. S., Yunes, J. A., Gillies, R. J., & Gatenby, R. A. (2009). The potential role of systemic buffers in reducing intratumoral extracellular pH and acid-mediated invasion. Cancer Research, 69(6), 2677–2684. https://doi.org/10.1158/0008-5472.CAN-08-2394.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Fais, S., Venturi, G., & Gatenby, B. (2014). Microenvironmental acidosis in carcinogenesis and metastases: new strategies in prevention and therapy. Cancer Metastasis Reviews, 33(4), 1095–1108. https://doi.org/10.1007/s10555-014-9531-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Robey, I. F., Baggett, B. K., Kirkpatrick, N. D., Roe, D. J., Dosescu, J., Sloane, B. F., et al. (2009). Bicarbonate increases tumor pH and inhibits spontaneous metastases. Cancer Research, 69(6), 2260–2268. https://doi.org/10.1158/0008-5472.CAN-07-5575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Ibrahim-Hashim, A., Abrahams, D., Enriquez-Navas, P. M., Luddy, K., Gatenby, R. A., & Gillies, R. J. (2017). Tris-base buffer: a promising new inhibitor for cancer progression and metastasis. Cancer Medicine, 6(7), 1720–1729. https://doi.org/10.1002/cam4.1032.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Ibrahim Hashim, A., Cornnell, H. H., Coelho Ribeiro Mde, L., Abrahams, D., Cunningham, J., Lloyd, M., et al. (2011). Reduction of metastasis using a non-volatile buffer. Clinical & Experimental Metastasis, 28(8), 841–849. https://doi.org/10.1007/s10585-011-9415-7.

    Article  CAS  Google Scholar 

  184. Chao, M., Wu, H., Jin, K., Li, B., Wu, J., Zhang, G., et al. (2016). A nonrandomized cohort and a randomized study of local control of large hepatocarcinoma by targeting intratumoral lactic acidosis. Elife, 5, e15691. https://doi.org/10.7554/eLife.15691.

  185. Jin, U. H., Lee, S. O., Pfent, C., & Safe, S. (2014). The aryl hydrocarbon receptor ligand omeprazole inhibits breast cancer cell invasion and metastasis. BMC Cancer, 14, 498. https://doi.org/10.1186/1471-2407-14-498.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This study was supported by the Deutsche Forschungsgemeinschaft DFG (grant TH 482/6-1).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Oliver Thews.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thews, O., Riemann, A. Tumor pH and metastasis: a malignant process beyond hypoxia. Cancer Metastasis Rev 38, 113–129 (2019). https://doi.org/10.1007/s10555-018-09777-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10555-018-09777-y

Keywords

Navigation