Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Metabolic enzyme LDHA activates Rac1 GTPase as a noncanonical mechanism to promote cancer

Abstract

The glycolytic enzyme lactate dehydrogenase A (LDHA) is frequently overexpressed in cancer, which promotes glycolysis and cancer. The oncogenic effect of LDHA has been attributed to its glycolytic enzyme activity. Here we report an unexpected noncanonical oncogenic mechanism of LDHA; LDHA activates small GTPase Rac1 to promote cancer independently of its glycolytic enzyme activity. Mechanistically, LDHA interacts with the active form of Rac1, Rac1–GTP, to inhibit Rac1–GTP interaction with its negative regulator, GTPase-activating proteins, leading to Rac1 activation in cancer cells and mouse tissues. In clinical breast cancer specimens, LDHA overexpression is associated with higher Rac1 activity. Rac1 inhibition suppresses the oncogenic effect of LDHA. Combination inhibition of LDHA enzyme activity and Rac1 activity by small-molecule inhibitors displays a synergistic inhibitory effect on breast cancers with LDHA overexpression. These results reveal a critical oncogenic mechanism of LDHA and suggest a promising therapeutic strategy for breast cancers with LDHA overexpression.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: LDHA directly interacts with Rac1.
Fig. 2: LDHA activates Rac1 in breast cancer cells and mouse mammary tissues.
Fig. 3: LDHA binds to Rac1–GTP and inhibits its binding with Rac1 GAPs to activate Rac1.
Fig. 4: LDHA activates Rac1 to promote colony formation, migration and invasion of breast cancer cells.
Fig. 5: LDHA activates Rac1 to promote growth and metastasis of breast tumors.
Fig. 6: LDHA and Rac1 small-molecule inhibitors display a synergistic inhibitory effect on colony formation, migration and invasion of breast cancer cells.
Fig. 7: LDHA and Rac1 small-molecule inhibitors display a synergistic inhibitory effect on primary and metastatic breast tumors.

Similar content being viewed by others

Data availability

Publicly available datasets used in this study are TGCA (https://portal.gdc.cancer.gov/), Oncomine (https://www.oncomine.com/) and Cancer Cell Line Encyclopedia (https://sites.broadinstitute.org/ccle/). All data supporting the present study are available within the article and supplementary information files. Source data are provided with this paper.

References

  1. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  2. Wolpaw, A. J. & Dang, C. V. Exploiting metabolic vulnerabilities of cancer with precision and accuracy. Trends Cell Biol. 28, 201–212 (2018).

    Article  CAS  Google Scholar 

  3. Zhu, J. & Thompson, C. B. Metabolic regulation of cell growth and proliferation. Nat. Rev. Mol. Cell Biol. 20, 436–450 (2019).

    Article  CAS  Google Scholar 

  4. Girgis, H. et al. Lactate dehydrogenase A is a potential prognostic marker in clear cell renal cell carcinoma. Mol. Cancer 13, 101 (2014).

    Article  Google Scholar 

  5. Huang, X. et al. High expressions of LDHA and AMPK as prognostic biomarkers for breast cancer. Breast 30, 39–46 (2016).

    Article  Google Scholar 

  6. Fantin, V. R., St-Pierre, J. & Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 9, 425–434 (2006).

    Article  CAS  Google Scholar 

  7. Cui, J. et al. FOXM1 promotes the Warburg effect and pancreatic cancer progression via transactivation of LDHA expression. Clin. Cancer Res. 20, 2595–2606 (2014).

    Article  CAS  Google Scholar 

  8. Zhao, D. et al. Lysine-5 acetylation negatively regulates lactate dehydrogenase A and is decreased in pancreatic cancer. Cancer Cell 23, 464–476 (2013).

    Article  CAS  Google Scholar 

  9. Koukourakis, M. I. et al. Lactate dehydrogenase 5 expression in operable colorectal cancer: strong association with survival and activated vascular endothelial growth factor pathway–a report of the Tumour Angiogenesis Research Group. J. Clin. Oncol. 24, 4301–4308 (2006).

    Article  CAS  Google Scholar 

  10. Lv, J. et al. Prognostic value of lactate dehydrogenase expression in different cancers: a meta-analysis. Am. J. Med. Sci. 358, 412–421 (2019).

    Article  Google Scholar 

  11. Shim, H. et al. c-Myc transactivation of LDH-A: implications for tumor metabolism and growth. Proc. Natl Acad. Sci. USA 94, 6658–6663 (1997).

    Article  CAS  Google Scholar 

  12. Le, A. et al. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc. Natl Acad. Sci. USA 107, 2037–2042 (2010).

    Article  CAS  Google Scholar 

  13. Wang, Z. Y. et al. LDH-A silencing suppresses breast cancer tumorigenicity through induction of oxidative stress mediated mitochondrial pathway apoptosis. Breast Cancer Res. Treat. 131, 791–800 (2012).

    Article  CAS  Google Scholar 

  14. Doherty, J. R. & Cleveland, J. L. Targeting lactate metabolism for cancer therapeutics. J. Clin. Investig. 123, 3685–3692 (2013).

    Article  CAS  Google Scholar 

  15. Yeung, C. et al. Targeting glycolysis through inhibition of lactate dehydrogenase impairs tumor growth in preclinical models of Ewing sarcoma. Cancer Res. 79, 5060–5073 (2019).

    Article  CAS  Google Scholar 

  16. Rizwan, A. et al. Relationships between LDH-A, lactate, and metastases in 4T1 breast tumors. Clin. Cancer Res. 19, 5158–5169 (2013).

    Article  CAS  Google Scholar 

  17. Xie, H. et al. Targeting lactate dehydrogenase-A inhibits tumorigenesis and tumor progression in mouse models of lung cancer and impacts tumor-initiating cells. Cell Metab. 19, 795–809 (2014).

    Article  CAS  Google Scholar 

  18. Martinez-Ordonez, A. et al. POU1F1 transcription factor induces metabolic reprogramming and breast cancer progression via LDHA regulation. Oncogene 40, 2725–2740 (2021).

    Article  CAS  Google Scholar 

  19. Semenza, G. L. et al. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J. Biol. Chem. 271, 32529–32537 (1996).

    Article  CAS  Google Scholar 

  20. Zhang, D. G., Zheng, J. N. & Pei, D. S. P53/microRNA-34-induced metabolic regulation: new opportunities in anticancer therapy. Mol. Cancer 13, 115 (2014).

    Article  Google Scholar 

  21. Valvona, C. J., Fillmore, H. L., Nunn, P. B. & Pilkington, G. J. The regulation and function of lactate dehydrogenase A: therapeutic potential in brain tumor. Brain Pathol. 26, 3–17 (2016).

    Article  CAS  Google Scholar 

  22. Feng, Y. et al. Lactate dehydrogenase A: a key player in carcinogenesis and potential target in cancer therapy. Cancer Med. 7, 6124–6136 (2018).

    Article  Google Scholar 

  23. Brand, A. et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24, 657–671 (2016).

    Article  CAS  Google Scholar 

  24. Woodford, M. R., Chen, V. Z., Backe, S. J., Bratslavsky, G. & Mollapour, M. Structural and functional regulation of lactate dehydrogenase-A in cancer. Future Med. Chem. 12, 439–455 (2020).

    Article  CAS  Google Scholar 

  25. Pan, C., Li, B. & Simon, M. C. Moonlighting functions of metabolic enzymes and metabolites in cancer. Mol. Cell 81, 3760–3774 (2021).

    Article  CAS  Google Scholar 

  26. Dasgupta, S. et al. Metabolic enzyme PFKFB4 activates transcriptional coactivator SRC-3 to drive breast cancer. Nature 556, 249–254 (2018).

    Article  CAS  Google Scholar 

  27. Enzo, E. et al. Aerobic glycolysis tunes YAP/TAZ transcriptional activity. EMBO J. 34, 1349–1370 (2015).

    Article  CAS  Google Scholar 

  28. Hodge, R. G. & Ridley, A. J. Regulating Rho GTPases and their regulators. Nat. Rev. Mol. Cell Biol. 17, 496–510 (2016).

    Article  CAS  Google Scholar 

  29. Kazanietz, M. G. & Caloca, M. J. The Rac GTPase in cancer: from old concepts to new paradigms. Cancer Res. 77, 5445–5451 (2017).

    Article  CAS  Google Scholar 

  30. Liang, J. et al. Rac1, a potential target for tumor therapy. Front Oncol. 11, 674426 (2021).

    Article  Google Scholar 

  31. Yue, X. et al. Gain-of-function mutant p53 activates small GTPase Rac1 through SUMOylation to promote tumor progression. Genes Dev. 31, 1641–1654 (2017).

    Article  CAS  Google Scholar 

  32. Rhodes, D. R. et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia 9, 166–180 (2007).

    Article  CAS  Google Scholar 

  33. Gyorffy, B. Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer. Comput Struct. Biotechnol. J. 19, 4101–4109 (2021).

    Article  CAS  Google Scholar 

  34. Soderberg, O. et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000 (2006).

    Article  Google Scholar 

  35. Zhang, C. et al. Glutaminase 2 is a novel negative regulator of small GTPase Rac1 and mediates p53 function in suppressing metastasis. eLife 5, e10727 (2016).

    Article  Google Scholar 

  36. Hayashi-Takagi, A. et al. Disrupted-in-schizophrenia 1 (DISC1) regulates spines of the glutamate synapse via Rac1. Nat. Neurosci. 13, 327–332 (2010).

    Article  CAS  Google Scholar 

  37. Cardama, G. A. et al. Relevance of small GTPase Rac1 pathway in drug and radio-resistance mechanisms: opportunities in cancer therapeutics. Crit. Rev. Oncol. Hematol. 124, 29–36 (2018).

    Article  CAS  Google Scholar 

  38. Fukata, M. et al. Rac1 and Cdc42 capture microtubules through IQGAP1 and CLIP-170. Cell 109, 873–885 (2002).

    Article  CAS  Google Scholar 

  39. Feig, L. A. Tools of the trade: use of dominant-inhibitory mutants of Ras-family GTPases. Nat. Cell Biol. 1, E25–E27 (1999).

    Article  CAS  Google Scholar 

  40. Joneson, T., White, M. A., Wigler, M. H. & Bar-Sagi, D. Stimulation of membrane ruffling and MAP kinase activation by distinct effectors of RAS. Science 271, 810–812 (1996).

    Article  CAS  Google Scholar 

  41. Keely, P. J., Westwick, J. K., Whitehead, I. P., Der, C. J. & Parise, L. V. Cdc42 and Rac1 induce integrin-mediated cell motility and invasiveness through PI(3)K. Nature 390, 632–636 (1997).

    Article  CAS  Google Scholar 

  42. Murphy, D. A. & Courtneidge, S. A. The ‘ins’ and ‘outs’ of podosomes and invadopodia: characteristics, formation and function. Nat. Rev. Mol. Cell Biol. 12, 413–426 (2011).

    Article  CAS  Google Scholar 

  43. Augoff, K., Hryniewicz-Jankowska, A. & Tabola, R. Invadopodia: clearing the way for cancer cell invasion. Ann. Transl. Med. 8, 902 (2020).

    Article  Google Scholar 

  44. Moshfegh, Y., Bravo-Cordero, J. J., Miskolci, V., Condeelis, J. & Hodgson, L. A trio-Rac1-Pak1 signalling axis drives invadopodia disassembly. Nat. Cell Biol. 16, 574–586 (2014).

    Article  CAS  Google Scholar 

  45. Donnelly, S. K. et al. Rac3 regulates breast cancer invasion and metastasis by controlling adhesion and matrix degradation. J. Cell Biol. 216, 4331–4349 (2017).

    Article  CAS  Google Scholar 

  46. Seals, D. F. et al. The adaptor protein Tks5/Fish is required for podosome formation and function, and for the protease-driven invasion of cancer cells. Cancer Cell 7, 155–165 (2005).

    Article  CAS  Google Scholar 

  47. Yamaguchi, H. et al. Lipid rafts and caveolin-1 are required for invadopodia formation and extracellular matrix degradation by human breast cancer cells. Cancer Res. 69, 8594–8602 (2009).

    Article  CAS  Google Scholar 

  48. Yamaguchi, H. et al. Phosphoinositide 3-kinase signaling pathway mediated by p110α regulates invadopodia formation. J. Cell Biol. 193, 1275–1288 (2011).

    Article  CAS  Google Scholar 

  49. Attanasio, F. et al. Novel invadopodia components revealed by differential proteomic analysis. Eur. J. Cell Biol. 90, 115–127 (2011).

    Article  CAS  Google Scholar 

  50. Gao, Y., Dickerson, J. B., Guo, F., Zheng, J. & Zheng, Y. Rational design and characterization of a Rac GTPase-specific small molecule inhibitor. Proc. Natl Acad. Sci. USA 101, 7618–7623 (2004).

    Article  CAS  Google Scholar 

  51. Pajak, B. et al. 2-Deoxy-d-glucose and its analogs: from diagnostic to therapeutic agents. Int. J. Mol. Sci. 21, 234 (2019).

    Article  Google Scholar 

  52. Weinberg, F. et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl Acad. Sci. USA 107, 8788–8793 (2010).

    Article  CAS  Google Scholar 

  53. Wang, F. et al. Glycolytic stimulation is not a requirement for M2 macrophage differentiation. Cell Metab. 28, 463–475 (2018).

    Article  CAS  Google Scholar 

  54. Zhang, C. et al. Tumour-associated mutant p53 drives the Warburg effect. Nat. Commun. 4, 2935 (2013).

    Article  Google Scholar 

  55. Pulaski, B. A. & Ostrand-Rosenberg, S. Mouse 4T1 breast tumor model. Curr. Protoc. Immunol. 20, Unit 20.2 (2001).

    Google Scholar 

  56. Wan, L., Pantel, K. & Kang, Y. Tumor metastasis: moving new biological insights into the clinic. Nat. Med. 19, 1450–1464 (2013).

    Article  CAS  Google Scholar 

  57. Guy, C. T., Cardiff, R. D. & Muller, W. J. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol. Cell. Biol. 12, 954–961 (1992).

    CAS  Google Scholar 

  58. Pioli, P. A., Hamilton, B. J., Connolly, J. E., Brewer, G. & Rigby, W. F. Lactate dehydrogenase is an AU-rich element-binding protein that directly interacts with AUF1. J. Biol. Chem. 277, 35738–35745 (2002).

    Article  CAS  Google Scholar 

  59. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  Google Scholar 

  60. Liu, J. et al. Parkin ubiquitinates phosphoglycerate dehydrogenase to suppress serine synthesis and tumor progression. J. Clin. Invest. 130, 3253–3269 (2020).

    Article  CAS  Google Scholar 

  61. Liu, J. et al. Parkin targets HIF-1α for ubiquitination and degradation to inhibit breast tumor progression. Nat. Commun. 8, 1823 (2017).

    Article  Google Scholar 

  62. Chang, C. Y. et al. Tumor suppressor p53 regulates intestinal type 2 immunity. Nat. Commun. 12, 3371 (2021).

    Article  CAS  Google Scholar 

  63. Pellegrin, S. & Mellor, H. Rho GTPase activation assays. Curr. Protoc. Cell Biol. 14, Unit 14.18 (2008).

    Google Scholar 

  64. Dawson, N. J., Bell, R. A. & Storey, K. B. Purification and properties of white muscle lactate dehydrogenase from the anoxia-tolerant turtle, the red-eared slider, trachemys scripta elegans. Enzym. Res. 2013, 784973 (2013).

    Article  Google Scholar 

  65. Javed, M. U., Yousuf, F. A., Hussain, A. N., Ishaq, M. & Waqar, M. A. Purification and properties of lactate dehydrogenase from liver of Uromastix hardwickii. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 111, 27–34 (1995).

    Article  CAS  Google Scholar 

  66. Chou, T. C. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 70, 440–446 (2010).

    Article  CAS  Google Scholar 

  67. Chou, T. C. & Talalay, P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv. Enzym. Regul. 22, 27–55 (1984).

    Article  CAS  Google Scholar 

  68. Baik, M. et al. Identification of invadopodia by TKS5 staining in human cancer lines and patient tumor samples. MethodsX 6, 718–726 (2019).

    Article  Google Scholar 

  69. Wang, Y. H. et al. Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis. Cell 158, 1309–1323 (2014).

    Article  CAS  Google Scholar 

  70. Zhen, C. et al. Gankyrin promotes breast cancer cell metastasis by regulating Rac1 activity. Oncogene 32, 3452–3460 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

LC–MS/MS proteomic analysis was performed at the Biological Mass Spectrometry facility of Rutgers University. This work was supported in part by grants from the National Institutes of Health (R01CA227912 and R01CA214746 to Z.F., as well as R01CA203965 and R01CA260837 to W.H.) and Congressionally Directed Medical Research Programs (CA214746 to Z.F.). T.Z. and C.C. were supported by the postdoctoral fellowship from New Jersey Commission on Cancer Research.

Author information

Authors and Affiliations

Authors

Contributions

J.L., C.Z., T.Z., C.C., J.W. and L.B. performed the experiments and analyzed data. L.Z. and B.G.H. analyzed data and contributed important materials. W.H. and Z.F. conceived and supervised the study. J.L., W.H. and Z.F. wrote the manuscript.

Corresponding authors

Correspondence to Wenwei Hu or Zhaohui Feng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks William J. Muller, Taro Hitosugi and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Alfredo Giménez-Cassina, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 LDHA overexpression and its association with clinical outcomes in different subtypes of breast cancers classified by the status of ER, PR or HER2.

a-c, The increased LDHA mRNA levels in different subtypes of breast cancers compared with matched adjacent non-tumor breast tissues. The data were obtained from TCGA and the P-value was analyzed by two-tailed paired Student’s t-test. d-f, High LDHA mRNA expression is associated with poor relapse-free survival in patients with different subtypes of breast cancers. The data were obtained from Kaplan-Meier plotter (http://kmplot.com) and analyzed by the log-rank (Mantel–Cox) test. ER: estrogen receptor; PR: progesterone receptor.

Extended Data Fig. 2 LDHA displayed a much weaker interaction with Rac1-T35S mutant compared with WT Rac1 in cells.

Hs578T cells with ectopic expression of LDHA-Flag and WT Myc-Rac1, Myc-Rac1-T17N, or Myc-Rac1-T35S were employed for co-IP assays followed by western-blot assays. Data represent three repeats with similar results.

Source data

Extended Data Fig. 3 The dominant negative Rac1-T17N mutant reduces the promoting effect of WT and L4 LDHA on colony formation, migration and invasion of breast cancer cells.

a-c, Hs578T and SK-BR3 cells with ectopic expression of WT or L4 LDHA were transduced with control or Rac1-T17N expression vectors for colony formation (a), migration (b) and invasion (c) assays. Data represent mean ± s.d. (n = 6 independent experiments), two-way ANOVA followed by Tukey’s or Bonferroni’s test. *: P < 0.0001.

Source data

Extended Data Fig. 4 LDHA but not Rac1 is localized in invadopodia in breast cancer cell lines.

HS578T and MDA-MB231 cells seeded on gelatin-coated coverslips were labeled with anti-LDHA or Rac1 in far-red, anti-Tks5 in green and phalloidin (to stain F-actin) in red. The co-localization of Tks5 and F-actin in a punctate manner in cells was used as an indication of invadopodia formation. Arrows indicate invadopodia in cells. Scale bars: 20 μm. Data represent three repeats with similar results.

Extended Data Fig. 5 The potential role of different Rho family proteins in mediating the oncogenic effect of LDHA in breast cancer cells.

a, The interaction between LDHA with different Rho family proteins analyzed by co-IP assays. Hs578T cells expressing LDHA-Flag and Myc-Rac1, Myc-Cdc42, Myc-Rac3, or Myc-RhoA were used for co-IP and western-blot assays. b, LDHA activated Rac3 and Cdc42, but at a much less extent compared with its effect on Rac1. Hs578T and SK-BR3 cells expressing LDHA-Flag were used for PAK-PBD pull-down assays to measure the levels of GTP-bound Rac1, Rac3 and Cdc42. Left panels: represented results. Right panels: relative Rac1, Rac3 and Cdc42 activities analyzed by comparing the levels of GTP-bound Rac1, Rac3 and Cdc42 to the levels of total Rac1, Rac3 and Cdc42 proteins, respectively, in cells. Data represent n = 3 independent experiments, two-tailed unpaired Student’s t-test. c, Knockdown of Rac3 or Cdc42 displayed a much less pronounced effect on colony formation, migration and invasion of breast cancer cells with WT or L4 LDHA expression compared with Rac1 knockdown. Data represent mean ± s.d. (n = 6 independent experiments), two-way ANOVA followed by Tukey’s test or Dunnett’s test. d, Knockdown of Rac3 and Cdc42 in cells was confirmed by Taqman real-time PCR assays. Their mRNA levels were normalized with Actin. Data represent n = 3 independent experiments, one-way ANOVA followed by Dunnett’s test. *: P < 0.0001.

Source data

Extended Data Fig. 6 The relative expression levels of Rac1, Rac3 and Cdc42 in breast cancer cells.

Rac1 has a much higher expression level than Rac3 and Cdc42 in majority of breast cancer cell lines, including cell lines used in this study (labeled in red) as shown by the RNA-Seq data from Cancer Cell Line Encyclopedia (CCLE; https://sites.broadinstitute.org/ccle/).

Extended Data Fig. 7 LDHA and Rac1 small-molecule inhibitors display a much more pronounced inhibitory effect on colony formation, migration and invasion of breast cancer cells.

a, The combination treatment displayed a much more pronounced inhibitory effect on colony formation of breast cancer cells. BT-549, MCF7 and ZR-75-1 cells were treated with indicated concentrations of FX11 and/or NSC23766 for 4 days before colony formation assays. Combo: FX11 + NSC23766. b, c, The combination treatment displayed a much more pronounced inhibitory effect on migration (b) and invasion (c) of breast cancer cells than the single inhibitor treatment as analyzed by Transwell assays. In b, c, BT-549, MCF7 and ZR-75-1 cells were treated with the indicated concentrations of FX11 and/or NSC23766 for 24 h. NSC: NSC23766. Data represent mean ± s.d. (n = 6 independent experiments), one-way ANOVA followed by Tukey’s test. *: P < 0.0001.

Source data

Extended Data Fig. 8 The effect of small-molecule inhibitor treatments on the body weights of tumor-bearing mice.

a, Small-molecule inhibitor treatments did not significantly affect the body weights of female nude mice bearing orthotopic tumors formed by WT Hs578T cells. b, Small-molecule inhibitor treatments did not significantly affect the body weights of female BALB/c mice bearing orthotopic tumors formed by 4T1 cells transduced with the control lentiviral shRNA vector. Mice were treated with FX11 (1 mg/kg/day; i.p.; once/day) and/or NSC23766 (1.5 mg/kg/day, i.p.; once/day), or vehicle (−). Mice with Hs578T tumors were treated for 3 weeks and mice with 4T1 tumors were treated for 2 weeks. The body weights of mice were measured and recorded at the days indicated. Data represent mean ± s.d. n=8 mice/group. Statistical differences were determined by two-way ANOVA followed by Tukey’s test.

Source data

Extended Data Fig. 9 The effect of FX11 and/or NSC23766 on the growth and lung metastasis of mammary tumors in MMTV-PyMT mice.

a, The effects of FX11 and/or NSC23766 on the growth and metastasis of mammary tumors in 10-week-old MMTV-PyMT mice. The 10-week-old female MMTV-PyMT mice that developed late-stage tumors were treated with FX11 (1 mg/kg/day; i.p.; once/day) and/or NSC23766 (1.5 mg/kg/day, i.p.; once/day) for 3 weeks before they were sacrificed for analysis. Data represent mean ± s.d.; n=6 mice/group. One-way ANOVA followed by Student’s t-test. Con: vehicle; NSC: NSC23766; combo: FX11 + NSC. b, The interaction between endogenous LDHA and Rac1 in both non-tumor and tumor mammary tissues of MMTV-PyMT mice detected by co-IP and western-blot assays. Non-tumor mammary tissues were obtained from 5-week-old MMTV-PyMT mice and mammary tumor tissues were obtained from 10-week-old MMTV-PyMT mice. Normal mammary tissues from 5-week-old LDHA-deficient mice (the R26-Cre-ERT2, LDHAflox/flox mice treated with Tamoxifen to delete LDHA as presented in Fig. 2h) were used as negative controls for assays. NC: negative control.

Source data

Extended Data Fig. 10 LDHA expression is not linked to any specific subtypes of breast cancer.

LDHA protein levels in two breast cancer TMAs, including TMA-RCINJ (a-c; n=200) and TMA-BR2082a (d-f; n=120), were analyzed by IHC staining and were compared in different cancer subtypes classified by the status of ER (a, d), PR (b, e), or HER2 (c, f). Statistical studies were performed by using χ2 test. The TMA-BR2161 does not have information on the status of ER, PR or HER2.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–8, Supplementary Tables 1 and 2 and unprocessed blots for Supplementary Figs.

Reporting Summary

Supplementary Data 1

Supplementary statistical source data.

Source data

Source Data Fig. 1

Unprocessed western blots for Fig. 1.

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Unprocessed western blots for Fig. 2.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Unprocessed western blots for Fig. 3.

Source Data Fig. 4

Unprocessed western blots for Fig. 4.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 5

Unprocessed western blots for Fig. 5.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Fig. 7

Statistical source data for Fig. 7.

Source Data Extended Data Fig. 2

Unprocessed western blots for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 5

Unprocessed western blots for Extended Data Fig. 5.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Unprocessed western blots for Extended Data Fig. 9.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Zhang, C., Zhang, T. et al. Metabolic enzyme LDHA activates Rac1 GTPase as a noncanonical mechanism to promote cancer. Nat Metab 4, 1830–1846 (2022). https://doi.org/10.1038/s42255-022-00708-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00708-4

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer