Skip to main content
Top
Published in: Experimental Hematology & Oncology 1/2015

Open Access 01-12-2015 | Research

Lactate, a putative survival factor for myeloma cells, is incorporated by myeloma cells through monocarboxylate transporters 1

Authors: Shiho Fujiwara, Naoko Wada, Yawara Kawano, Yutaka Okuno, Yoshitaka Kikukawa, Shinya Endo, Nao Nishimura, Nina Ueno, Hiroaki Mitsuya, Hiroyuki Hata

Published in: Experimental Hematology & Oncology | Issue 1/2015

Login to get access

Abstract

Background

Lactate levels within tumors are correlated with metastases, tumor recurrence, and radioresistance, thus apparently contributing to poor outcomes in patients with various cancers. We previously reported that high-level production of lactate by multiple myeloma (MM) cell lines is associated with high-level LDH activity within such MM cells. However, the kinetics of lactate remains to be studied. In the present study, we attempted to elucidate the mechanism of lactate incorporation into MM cells.

Methods

Six MM cell lines and stromal cells obtained through long-term culture of bone marrow samples from MM patients were employed. Incorporation of lactate was quantified using C14-labeled lactate. The role of MCT1, a member of the monocarboxylate transporters (MCTs), expressed on MM cells, was examined in the presence of its inhibitor (α-cyano-4-hydroxycinnamic acid: CHC) and by using gene-silencing technique.

Results

MM cell lines as well as stromal cells were found to produce lactate. Incorporation of C14-labeled lactate into MM cells occurred in all 6 MM cell lines analyzed. Inhibition of MCT1 by using CHC or MCT1-targeting siRNA reduced lactate incorporation and caused apoptosis in MM cells. This apoptosis was enhanced when the activity of pyruvate dehydrogenase kinase was blocked by dichroloacetate. Survival of normal peripheral blood mononuclear cells was not influenced by MCT1 inhibition.

Conclusions

The present data suggest that lactate is produced by MM cell lines and stromal cells, and contributes to the survival of such MM cells in autocrine or paracrine manners. Suppression of lactate incorporation by targeting MCT1 may provide a novel therapeutic strategy for MM which may be applicable for other B-cell neoplasms.
Literature
1.
go back to reference Kim JW, Dang CV. Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 2006;66:8927–30. doi:10.1158/0008-5472.CAN-06-1501.CrossRefPubMed Kim JW, Dang CV. Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 2006;66:8927–30. doi:10.1158/0008-5472.CAN-06-1501.CrossRefPubMed
2.
go back to reference Brizel DM, Schroeder T, Scher RL, Walenta S, Clough RW, Dewhirst MW, et al. Elevated tumor lactate concentrations predict for an increased risk of metastases in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2001;51:349–53.CrossRefPubMed Brizel DM, Schroeder T, Scher RL, Walenta S, Clough RW, Dewhirst MW, et al. Elevated tumor lactate concentrations predict for an increased risk of metastases in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2001;51:349–53.CrossRefPubMed
3.
go back to reference Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfor K, Rofstad EK, et al. High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res. 2000;60:916–21.PubMed Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfor K, Rofstad EK, et al. High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res. 2000;60:916–21.PubMed
4.
go back to reference Quennet V, Yaromina A, Zips D, Rosner A, Walenta S, Baumann M, et al. Tumor lactate content predicts for response to fractionated irradiation of human squamous cell carcinomas in nude mice. Radiother Oncol. 2006;81:130–5. doi:10.1016/j.radonc.2006.08.012.CrossRefPubMed Quennet V, Yaromina A, Zips D, Rosner A, Walenta S, Baumann M, et al. Tumor lactate content predicts for response to fractionated irradiation of human squamous cell carcinomas in nude mice. Radiother Oncol. 2006;81:130–5. doi:10.1016/j.radonc.2006.08.012.CrossRefPubMed
5.
go back to reference Kennedy KM, Dewhirst MW. Tumor metabolism of lactate: the influence and therapeutic potential for MCT and CD147 regulation. Future Oncol. 2010;6:127–48. doi:10.2217/fon.09.145.CrossRefPubMedCentralPubMed Kennedy KM, Dewhirst MW. Tumor metabolism of lactate: the influence and therapeutic potential for MCT and CD147 regulation. Future Oncol. 2010;6:127–48. doi:10.2217/fon.09.145.CrossRefPubMedCentralPubMed
6.
go back to reference Draoui N, Feron O. Lactate shuttles at a glance: from physiological paradigms to anti-cancer treatments. Dis Model Mech. 2011;4:727–32. doi:10.1242/dmm.007724.CrossRefPubMedCentralPubMed Draoui N, Feron O. Lactate shuttles at a glance: from physiological paradigms to anti-cancer treatments. Dis Model Mech. 2011;4:727–32. doi:10.1242/dmm.007724.CrossRefPubMedCentralPubMed
7.
go back to reference Halestrap AP. The monocarboxylate transporter family–Structure and functional characterization. IUBMB Life. 2012;64:1–9. doi:10.1002/iub.573.CrossRefPubMed Halestrap AP. The monocarboxylate transporter family–Structure and functional characterization. IUBMB Life. 2012;64:1–9. doi:10.1002/iub.573.CrossRefPubMed
8.
go back to reference Halestrap AP, Meredith D. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflugers Arch. 2004;447:619–28. doi:10.1007/s00424-003-1067-2.CrossRefPubMed Halestrap AP, Meredith D. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflugers Arch. 2004;447:619–28. doi:10.1007/s00424-003-1067-2.CrossRefPubMed
9.
go back to reference Kirk P, Wilson MC, Heddle C, Brown MH, Barclay AN, Halestrap AP. CD147 is tightly associated with lactate transporters MCT1 and MCT4 and facilitates their cell surface expression. EMBO J. 2000;19:3896–904. doi:10.1093/emboj/19.15.3896.CrossRefPubMedCentralPubMed Kirk P, Wilson MC, Heddle C, Brown MH, Barclay AN, Halestrap AP. CD147 is tightly associated with lactate transporters MCT1 and MCT4 and facilitates their cell surface expression. EMBO J. 2000;19:3896–904. doi:10.1093/emboj/19.15.3896.CrossRefPubMedCentralPubMed
10.
go back to reference Wilson MC, Meredith D, Halestrap AP. Fluorescence resonance energy transfer studies on the interaction between the lactate transporter MCT1 and CD147 provide information on the topology and stoichiometry of the complex in situ. J Biol Chem. 2002;277:3666–72. doi:10.1074/jbc.M109658200.CrossRefPubMed Wilson MC, Meredith D, Halestrap AP. Fluorescence resonance energy transfer studies on the interaction between the lactate transporter MCT1 and CD147 provide information on the topology and stoichiometry of the complex in situ. J Biol Chem. 2002;277:3666–72. doi:10.1074/jbc.M109658200.CrossRefPubMed
11.
go back to reference Nabeshima K, Iwasaki H, Koga K, Hojo H, Suzumiya J, Kikuchi M. Emmprin (basigin/CD147): matrix metalloproteinase modulator and multifunctional cell recognition molecule that plays a critical role in cancer progression. Pathol Int. 2006;56:359–67. doi:10.1111/j.1440-1827.2006.01972.x.CrossRefPubMed Nabeshima K, Iwasaki H, Koga K, Hojo H, Suzumiya J, Kikuchi M. Emmprin (basigin/CD147): matrix metalloproteinase modulator and multifunctional cell recognition molecule that plays a critical role in cancer progression. Pathol Int. 2006;56:359–67. doi:10.1111/j.1440-1827.2006.01972.x.CrossRefPubMed
12.
go back to reference Pinheiro C, Longatto-Filho A, Azevedo-Silva J, Casal M, Schmitt FC, Baltazar F. Role of monocarboxylate transporters in human cancers: state of the art. J Bioenerg Biomembr. 2012;44:127–39. doi:10.1007/s10863-012-9428-1.CrossRefPubMed Pinheiro C, Longatto-Filho A, Azevedo-Silva J, Casal M, Schmitt FC, Baltazar F. Role of monocarboxylate transporters in human cancers: state of the art. J Bioenerg Biomembr. 2012;44:127–39. doi:10.1007/s10863-012-9428-1.CrossRefPubMed
13.
go back to reference Walters DK, Arendt BK, Jelinek DF. CD147 regulates the expression of MCT1 and lactate export in multiple myeloma cells. Cell Cycle. 2013;12:3175–83. doi:10.4161/cc.26193.CrossRefPubMedCentralPubMed Walters DK, Arendt BK, Jelinek DF. CD147 regulates the expression of MCT1 and lactate export in multiple myeloma cells. Cell Cycle. 2013;12:3175–83. doi:10.4161/cc.26193.CrossRefPubMedCentralPubMed
14.
go back to reference Fujiwara S, Kawano Y, Yuki H, Okuno Y, Nosaka K, Mitsuya H, et al. PDK1 inhibition is a novel therapeutic target in multiple myeloma. Br J Cancer. 2013;108:170–8. doi:10.1038/bjc.2012.527.CrossRefPubMedCentralPubMed Fujiwara S, Kawano Y, Yuki H, Okuno Y, Nosaka K, Mitsuya H, et al. PDK1 inhibition is a novel therapeutic target in multiple myeloma. Br J Cancer. 2013;108:170–8. doi:10.1038/bjc.2012.527.CrossRefPubMedCentralPubMed
15.
go back to reference Sonveaux P, Vegran F, Schroeder T, Wergin MC, Verrax J, Rabbani ZN, et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J Clin Invest. 2008;118:3930–42. doi:10.1172/Jci36843.PubMedCentralPubMed Sonveaux P, Vegran F, Schroeder T, Wergin MC, Verrax J, Rabbani ZN, et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J Clin Invest. 2008;118:3930–42. doi:10.1172/Jci36843.PubMedCentralPubMed
17.
go back to reference Martinez-Outschoorn UE, Whitaker-Menezes D, Valsecchi M, Martinez-Cantarin MP, Dulau-Florea A, Gong J, et al. Reverse Warburg effect in a patient with aggressive B-cell lymphoma: is lactic acidosis a paraneoplastic syndrome? Semin Oncol. 2013;40:403–18. doi:10.1053/j.seminoncol.2013.04.016.CrossRefPubMed Martinez-Outschoorn UE, Whitaker-Menezes D, Valsecchi M, Martinez-Cantarin MP, Dulau-Florea A, Gong J, et al. Reverse Warburg effect in a patient with aggressive B-cell lymphoma: is lactic acidosis a paraneoplastic syndrome? Semin Oncol. 2013;40:403–18. doi:10.1053/j.seminoncol.2013.04.016.CrossRefPubMed
18.
go back to reference Busk M, Walenta S, Mueller-Klieser W, Steiniche T, Jakobsen S, Horsman MR, et al. Inhibition of tumor lactate oxidation: consequences for the tumor microenvironment. Radiother Oncol. 2011;99:404–11. doi:10.1016/j.radonc.2011.05.053.CrossRefPubMed Busk M, Walenta S, Mueller-Klieser W, Steiniche T, Jakobsen S, Horsman MR, et al. Inhibition of tumor lactate oxidation: consequences for the tumor microenvironment. Radiother Oncol. 2011;99:404–11. doi:10.1016/j.radonc.2011.05.053.CrossRefPubMed
19.
go back to reference Hirschhaeuser F, Sattler UG, Mueller-Klieser W. Lactate: a metabolic key player in cancer. Cancer Res. 2011;71:6921–5. doi:10.1158/0008-5472.CAN-11-1457.CrossRefPubMed Hirschhaeuser F, Sattler UG, Mueller-Klieser W. Lactate: a metabolic key player in cancer. Cancer Res. 2011;71:6921–5. doi:10.1158/0008-5472.CAN-11-1457.CrossRefPubMed
20.
go back to reference Walenta S, Chau TV, Schroeder T, Lehr HA, Kunz-Schughart LA, Fuerst A, et al. Metabolic classification of human rectal adenocarcinomas: a novel guideline for clinical oncologists? J Cancer Res Clin Oncol. 2003;129:321–6. doi:10.1007/s00432-003-0450-x.CrossRefPubMed Walenta S, Chau TV, Schroeder T, Lehr HA, Kunz-Schughart LA, Fuerst A, et al. Metabolic classification of human rectal adenocarcinomas: a novel guideline for clinical oncologists? J Cancer Res Clin Oncol. 2003;129:321–6. doi:10.1007/s00432-003-0450-x.CrossRefPubMed
21.
go back to reference Yokota H, Guo J, Matoba M, Higashi K, Tonami H, Nagao Y. Lactate, choline, and creatine levels measured by vitro 1H-MRS as prognostic parameters in patients with non-small-cell lung cancer. J Magn Reson Imaging. 2007;25:992–9. doi:10.1002/jmri.20902.CrossRefPubMed Yokota H, Guo J, Matoba M, Higashi K, Tonami H, Nagao Y. Lactate, choline, and creatine levels measured by vitro 1H-MRS as prognostic parameters in patients with non-small-cell lung cancer. J Magn Reson Imaging. 2007;25:992–9. doi:10.1002/jmri.20902.CrossRefPubMed
22.
go back to reference Kang KW, Im YB, Go WJ, Han HK. C-myc amplification altered the gene expression of ABC- and SLC-transporters in human breast epithelial cells. Mol Pharm. 2009;6:627–33. doi:10.1021/mp800116f.CrossRefPubMed Kang KW, Im YB, Go WJ, Han HK. C-myc amplification altered the gene expression of ABC- and SLC-transporters in human breast epithelial cells. Mol Pharm. 2009;6:627–33. doi:10.1021/mp800116f.CrossRefPubMed
23.
go back to reference Coller HA, Grandori C, Tamayo P, Colbert T, Lander ES, Eisenman RN, et al. Expression analysis with oligonucleotide microarrays reveals that MYC regulates genes involved in growth, cell cycle, signaling, and adhesion. Proc Natl Acad Sci U S A. 2000;97:3260–5.CrossRefPubMedCentralPubMed Coller HA, Grandori C, Tamayo P, Colbert T, Lander ES, Eisenman RN, et al. Expression analysis with oligonucleotide microarrays reveals that MYC regulates genes involved in growth, cell cycle, signaling, and adhesion. Proc Natl Acad Sci U S A. 2000;97:3260–5.CrossRefPubMedCentralPubMed
24.
go back to reference Schuhmacher M, Kohlhuber F, Holzel M, Kaiser C, Burtscher H, Jarsch M, et al. The transcriptional program of a human B cell line in response to Myc. Nucleic Acids Res. 2001;29:397–406.CrossRefPubMedCentralPubMed Schuhmacher M, Kohlhuber F, Holzel M, Kaiser C, Burtscher H, Jarsch M, et al. The transcriptional program of a human B cell line in response to Myc. Nucleic Acids Res. 2001;29:397–406.CrossRefPubMedCentralPubMed
25.
go back to reference Doherty JR, Yang C, Scott KE, Cameron MD, Fallahi M, Li W, et al. Blocking lactate export by inhibiting the Myc target MCT1 Disables glycolysis and glutathione synthesis. Cancer Res. 2014;74:908–20. doi:10.1158/0008-5472.CAN-13-2034.CrossRefPubMedCentralPubMed Doherty JR, Yang C, Scott KE, Cameron MD, Fallahi M, Li W, et al. Blocking lactate export by inhibiting the Myc target MCT1 Disables glycolysis and glutathione synthesis. Cancer Res. 2014;74:908–20. doi:10.1158/0008-5472.CAN-13-2034.CrossRefPubMedCentralPubMed
26.
go back to reference Zhao Z, Wu MS, Zou C, Tang Q, Lu J, Liu D, et al. Downregulation of MCT1 inhibits tumor growth, metastasis and enhances chemotherapeutic efficacy in osteosarcoma through regulation of the NF-kappaB pathway. Cancer Lett. 2014;342:150–8. doi:10.1016/j.canlet.2013.08.042.CrossRefPubMed Zhao Z, Wu MS, Zou C, Tang Q, Lu J, Liu D, et al. Downregulation of MCT1 inhibits tumor growth, metastasis and enhances chemotherapeutic efficacy in osteosarcoma through regulation of the NF-kappaB pathway. Cancer Lett. 2014;342:150–8. doi:10.1016/j.canlet.2013.08.042.CrossRefPubMed
27.
go back to reference Miranda-Goncalves V, Honavar M, Pinheiro C, Martinho O, Pires MM, Pinheiro C, et al. Monocarboxylate transporters (MCTs) in gliomas: expression and exploitation as therapeutic targets. Neuro Oncol. 2013;15:172–88. doi:10.1093/neuonc/nos298.CrossRefPubMedCentralPubMed Miranda-Goncalves V, Honavar M, Pinheiro C, Martinho O, Pires MM, Pinheiro C, et al. Monocarboxylate transporters (MCTs) in gliomas: expression and exploitation as therapeutic targets. Neuro Oncol. 2013;15:172–88. doi:10.1093/neuonc/nos298.CrossRefPubMedCentralPubMed
28.
go back to reference Kumar A, Kant S, Singh SM. Targeting monocarboxylate transporter by alpha-cyano-4-hydroxycinnamate modulates apoptosis and cisplatin resistance of Colo205 cells: implication of altered cell survival regulation. Apoptosis. 2013;18:1574–85. doi:10.1007/s10495-013-0894-7.CrossRefPubMed Kumar A, Kant S, Singh SM. Targeting monocarboxylate transporter by alpha-cyano-4-hydroxycinnamate modulates apoptosis and cisplatin resistance of Colo205 cells: implication of altered cell survival regulation. Apoptosis. 2013;18:1574–85. doi:10.1007/s10495-013-0894-7.CrossRefPubMed
29.
go back to reference Diers AR, Broniowska KA, Chang CF, Hogg N. Pyruvate fuels mitochondrial respiration and proliferation of breast cancer cells: effect of monocarboxylate transporter inhibition. Biochem J. 2012;444:561–71. doi:10.1042/BJ20120294.CrossRefPubMed Diers AR, Broniowska KA, Chang CF, Hogg N. Pyruvate fuels mitochondrial respiration and proliferation of breast cancer cells: effect of monocarboxylate transporter inhibition. Biochem J. 2012;444:561–71. doi:10.1042/BJ20120294.CrossRefPubMed
30.
go back to reference Ohtsuki T, Yawata Y, Wada H, Sugihara T, Mori M, Namba M. Two human myeloma cell lines, amylase-producing KMS-12-PE and amylase-non-producing KMS-12-BM, were established from a patient, having the same chromosome marker, t(11;14)(q13;q32). Br J Haematol. 1989;73:199–204.CrossRefPubMed Ohtsuki T, Yawata Y, Wada H, Sugihara T, Mori M, Namba M. Two human myeloma cell lines, amylase-producing KMS-12-PE and amylase-non-producing KMS-12-BM, were established from a patient, having the same chromosome marker, t(11;14)(q13;q32). Br J Haematol. 1989;73:199–204.CrossRefPubMed
31.
go back to reference Ikeyama S, Nakagawa S, Arakawa M, Sugino H, Kakinuma A. Purification and characterization of IgE produced by human myeloma cell line, U266. Mol Immunol. 1986;23:159–67.CrossRefPubMed Ikeyama S, Nakagawa S, Arakawa M, Sugino H, Kakinuma A. Purification and characterization of IgE produced by human myeloma cell line, U266. Mol Immunol. 1986;23:159–67.CrossRefPubMed
32.
go back to reference Matsuoka Y, Moore GE, Yagi Y, Pressman D. Production of free light chains of immunoglobulin by a hematopoietic cell line derived from a patient with multiple myeloma. Proc Soc Exp Biol Med. 1967;125:1246–50.CrossRefPubMed Matsuoka Y, Moore GE, Yagi Y, Pressman D. Production of free light chains of immunoglobulin by a hematopoietic cell line derived from a patient with multiple myeloma. Proc Soc Exp Biol Med. 1967;125:1246–50.CrossRefPubMed
33.
go back to reference Togawa A, Inoue N, Miyamoto K, Hyodo H, Namba M. Establishment and characterization of a human myeloma cell line (KMM-1). Int J Cancer. 1982;29:495–500.CrossRefPubMed Togawa A, Inoue N, Miyamoto K, Hyodo H, Namba M. Establishment and characterization of a human myeloma cell line (KMM-1). Int J Cancer. 1982;29:495–500.CrossRefPubMed
34.
go back to reference Hata H, Matsuzaki H, Sonoki T, Takemoto S, Kuribayashi N, Nagasaki A, et al. Establishment of a CD45-positive immature plasma cell line from an aggressive multiple myeloma with high serum lactate dehydrogenase. Leukemia. 1994;8:1768–73.PubMed Hata H, Matsuzaki H, Sonoki T, Takemoto S, Kuribayashi N, Nagasaki A, et al. Establishment of a CD45-positive immature plasma cell line from an aggressive multiple myeloma with high serum lactate dehydrogenase. Leukemia. 1994;8:1768–73.PubMed
35.
go back to reference Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001;25:402–8. doi:10.1006/meth.2001.1262.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001;25:402–8. doi:10.1006/meth.2001.1262.CrossRefPubMed
Metadata
Title
Lactate, a putative survival factor for myeloma cells, is incorporated by myeloma cells through monocarboxylate transporters 1
Authors
Shiho Fujiwara
Naoko Wada
Yawara Kawano
Yutaka Okuno
Yoshitaka Kikukawa
Shinya Endo
Nao Nishimura
Nina Ueno
Hiroaki Mitsuya
Hiroyuki Hata
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Experimental Hematology & Oncology / Issue 1/2015
Electronic ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-015-0008-z

Other articles of this Issue 1/2015

Experimental Hematology & Oncology 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine