Skip to main content
Log in

Glutamine deficiency promotes stemness and chemoresistance in tumor cells through DRP1-induced mitochondrial fragmentation

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Glutamine is essential for maintaining the TCA cycle in cancer cells yet they undergo glutamine starvation in the core of tumors. Cancer stem cells (CSCs), responsible for tumor recurrence are often found in the nutrient limiting cores. Our study uncovers the molecular basis and cellular links between glutamine deprivation and stemness in the cancer cells. We showed that glutamine is dispensable for the survival of ovarian and colon cancer cells while it is required for their proliferation. Glutamine starvation leads to the metabolic reprogramming in tumor cells with enhanced glycolysis and unaltered oxidative phosphorylation. Production of reactive oxygen species (ROS) in glutamine limiting condition induces MAPK-ERK1/2 signaling pathway to phosphorylate dynamin-related protein-1(DRP1) at Ser616. Moreover, p-DRP1 promotes mitochondrial fragmentation and enhances numbers of CD44 and CD117/CD45 positive CSCs. Besides the established features of cancer stem cells, glutamine deprivation induces perinuclear localization of fragmented mitochondria and reduction in proliferation rate which are usually observed in CSCs. Treatment with glutaminase inhibitor (L-DON) mimics the effects of glutamine starvation without altering cell survival in in vitro as well as in in vivo model. Interestingly, the combinatorial treatment of L-DON with DRP1 inhibitor (MDiVi-1) reduces the stem cell population in tumor tissue in mouse model. Collectively our data suggest that glutamine deficiency in the core of tumors can increase the cancer stem cell population and the combination therapy with MDiVi-1 and L-DON is a useful approach to reduce CSCs population in tumor.

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
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Availability of data

Data are available on request from the corresponding author.

References

  1. Mitra T, Prasad P, Mukherjee P et al (2018) Stemness and chemoresistance are imparted to the OC cells through TGFβ1 driven EMT. J Cell Biochem 119:5775–5787. https://doi.org/10.1002/jcb.26753

    Article  CAS  PubMed  Google Scholar 

  2. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:646–674. https://doi.org/10.1016/j.cell.2011.02.013

    Article  CAS  PubMed  Google Scholar 

  3. Pavlova NN, Thompson CB (2016) The Emerging hallmarks of cancer metabolism. Cell Metab 23:27–47. https://doi.org/10.1016/j.cmet.2015.12.006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Souba WW (1993) Glutamine and cancer. Ann Surg 218:715–728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yang L, Achreja A, Yeung TL et al (2016) Targeting stromal glutamine synthetase in tumors disrupts tumor microenvironment-regulated cancer cell growth. Cell Metab 24:685–700. https://doi.org/10.1016/j.cmet.2016.10.011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cluntun AA, Lukey MJ, Cerione RA, Locasale JW (2017) Glutamine metabolism in cancer: understanding the heterogeneity. Trends in Cancer 3:169–180. https://doi.org/10.1016/j.trecan.2017.01.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Scalise M, Pochini L, Galluccio M et al (2017) Glutamine transport and mitochondrial metabolism in cancer cell growth. Front Oncol 7:1–9. https://doi.org/10.3389/fonc.2017.00306

    Article  Google Scholar 

  8. Huang W, Choi W, Chen Y et al (2013) A proposed role for glutamine in cancer cell growth through acid resistance. Cell Res 23:724–727. https://doi.org/10.1038/cr.2013.15

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sabharwal SS, Schumacker PT (2014) Mitochondrial ROS in cancer: initiators, amplifiers or an Achilles’ heel? Nat Rev Cancer 14:709–721. https://doi.org/10.1038/nrc3803

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhao J, Zhang J, Yu M et al (2013) Mitochondrial dynamics regulates migration and invasion of breast cancer cells. Oncogene 32:4814–4824. https://doi.org/10.1038/onc.2012.494

    Article  CAS  PubMed  Google Scholar 

  11. Al-Mehdi AB, Pastukh VM, Swiger BM et al (2012) Perinuclear mitochondrial clustering creates an oxidant-rich nuclear domain required for hypoxia-induced transcription. Sci Signal 5:1–10. https://doi.org/10.1126/scisignal.2002712

    Article  CAS  Google Scholar 

  12. Cacace A, Sboarina M, Vazeille T, Sonveaux P (2017) Glutamine activates STAT3 to control cancer cell proliferation independently of glutamine metabolism. Oncogene 36:2074–2084. https://doi.org/10.1038/onc.2016.364

    Article  CAS  PubMed  Google Scholar 

  13. Yang L, Moss T, Mangala LS et al (2014) Metabolic shifts toward glutamine regulate tumor growth, invasion and bioenergetics in ovarian cancer. Mol Syst Biol 10:1–23. https://doi.org/10.1002/msb.20134892

    Article  CAS  Google Scholar 

  14. Yuan L, Sheng X, Willson AK et al (2015) Glutamine promotes ovarian cancer cell proliferation through the mTOR/S6 pathway. Endocr Relat Cancer 22:577–591. https://doi.org/10.1530/ERC-15-0192

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Choi Y, Park K (2018) Targeting glutamine metabolism for cancer treatment. Biomol Ther 26:19–28

    Article  CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  17. Das N, Mandala A, Naaz S et al (2017) Melatonin protects against lipid-induced mitochondrial dysfunction in hepatocytes and inhibits stellate cell activation during hepatic fibrosis in mice. J Pineal Res 62:1–21. https://doi.org/10.1111/jpi.12404

    Article  CAS  Google Scholar 

  18. Chowdhury SR, Ray U, Chatterjee BP, Roy SS (2017) Targeted apoptosis in ovarian cancer cells through mitochondrial dysfunction in response to Sambucus nigra agglutinin. Cell Death Dis 8:1–12. https://doi.org/10.1038/cddis.2017.77

    Article  CAS  Google Scholar 

  19. Khan M, Biswas D, Ghosh M et al (2015) mTORC2 controls cancer cell survival by modulating gluconeogenesis. Cell Death Discov 1:1–12. https://doi.org/10.1038/cddiscovery.2015.16

    Article  CAS  Google Scholar 

  20. Maftah A, Petit JM, Ratinaud MH, Julien R (1989) 10-N nonyl-acridine orange: a fluorescent probe which stains mitochondria independently of their energetic state. Biochem Biophys Res Commun 164:185–190. https://doi.org/10.1016/0006-291X(89)91700-2

    Article  CAS  PubMed  Google Scholar 

  21. De R, Sarkar S, Mazumder S et al (2018) Macrophage migration inhibitory factor regulates mitochondrial dynamics and cell growth of human cancer cell lines through CD74-NF-κB signaling. J Biol Chem 293:19740–19760. https://doi.org/10.1074/jbc.RA118.003935

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bordi M, Nazio F, Campello S (2017) The close interconnection between mitochondrial dynamics and mitophagy in cancer. Front Oncol 7:1–9. https://doi.org/10.3389/fonc.2017.00081

    Article  Google Scholar 

  23. Xiao B, Deng X, Zhou W, Tan EK (2016) Flow cytometry-based assessment of mitophagy using mitotracker. Front Cell Neurosci 10:1–4. https://doi.org/10.3389/fncel.2016.00076

    Article  CAS  Google Scholar 

  24. Song IS (2015) Mitochondria as therapeutic targets for cancer stem cells. World J Stem Cells 7:418. https://doi.org/10.4252/wjsc.v7.i2.418

    Article  PubMed  PubMed Central  Google Scholar 

  25. Parte SC, Batra SK, Kakar SS (2018) Characterization of stem cell and cancer stem cell populations in ovary and ovarian tumors. J Ovarian Res 11:1–16. https://doi.org/10.1186/s13048-018-0439-3

    Article  CAS  Google Scholar 

  26. Akhter Z, Sharawat SK, Kumar V et al (2018) Aggressive serous epithelial ovarian cancer is potentially propagated by EpCAM + CD45 + phenotype. Oncogene. https://doi.org/10.1038/s41388-017-0106-y

    Article  PubMed  Google Scholar 

  27. Bhattacharya R, Mitra T, Ray Chaudhuri S, Roy SS (2018) Mesenchymal splice isoform of CD44 (CD44s) promotes EMT/invasion and imparts stem-like properties to ovarian cancer cells. J Cell Biochem 119:3373–3383. https://doi.org/10.1002/jcb.26504

    Article  CAS  PubMed  Google Scholar 

  28. Chen H, Chan DC (2017) Mitochondrial dynamics in regulating the unique phenotypes of cancer and stem cells. Cell Metab 26:39–48. https://doi.org/10.1016/j.cmet.2017.05.016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cetinbas NM, Sudderth J, Harris RC et al (2016) Glucose-dependent anaplerosis in cancer cells is required for cellular redox balance in the absence of glutamine. Sci Rep 6:1–12. https://doi.org/10.1038/srep32606

    Article  CAS  Google Scholar 

  30. Venneti S, Dunphy MP, Zhang H et al (2015) Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo. Sci Transl Med 7:1–10. https://doi.org/10.1126/scitranslmed.aaa1009

    Article  CAS  Google Scholar 

  31. Pan M, Reid MA, Lowman XH et al (2016) Regional glutamine deficiency in tumours promotes dedifferentiation through inhibition of histone demethylation. Nat Cell Biol 18:1090–1101. https://doi.org/10.1038/ncb3410

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wise DR, Thompson CB (2010) Glutamine addiction: a new therapeutic target in cancer. Trends Biochem Sci 35:427–433. https://doi.org/10.1016/j.tibs.2010.05.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zacharias NM, McCullough C, Shanmugavelandy S et al (2017) Metabolic differences in glutamine utilization lead to metabolic vulnerabilities in prostate cancer. Sci Rep 7:1–11. https://doi.org/10.1038/s41598-017-16327-z

    Article  CAS  Google Scholar 

  34. Zhu L, Ploessl K, Zhou R et al (2017) Metabolic imaging of glutamine in cancer. J Nucl Med 58:533–537. https://doi.org/10.2967/jnumed.116.182345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sonveaux P, Végran F, Schroeder T et al (2008) Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J Clin Invest 118:3930–3942. https://doi.org/10.1172/JCI36843

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Trotta AP, Chipuk JE (2017) Mitochondrial dynamics as regulators of cancer biology. Cell Mol Life Sci 74:1999–2017. https://doi.org/10.1007/s00018-016-2451-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Campello S, Scorrano L (2010) Mitochondrial shape changes: Orchestrating cell pathophysiology. EMBO Rep 11:678–684. https://doi.org/10.1038/embor.2010.115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhao RZ, Jiang S, Zhang L, Bin YuZ (2019) Mitochondrial electron transport chain, ROS generation and uncoupling (review). Int J Mol Med 44:3–15. https://doi.org/10.3892/ijmm.2019.4188

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kashatus JA, Nascimento A, Myers LJ et al (2015) Erk2 phosphorylation of Drp1 promotes mitochondrial fission and MAPK-driven tumor growth. Mol Cell 57:537–551. https://doi.org/10.1016/j.molcel.2015.01.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Afanas’ev I (2011) Reactive oxygen species signaling in cancer: comparison with aging. Aging Dis 2:219–230

    PubMed  Google Scholar 

  41. Yu T, Robotham JL, Yoon Y (2006) Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc Natl Acad Sci U S A 103:2653–2658. https://doi.org/10.1073/pnas.0511154103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kong B, Tsuyoshi H, Orisaka M et al (2015) Mitochondrial dynamics regulating chemoresistance in gynecological cancers. Ann N Y Acad Sci 1350:1–16. https://doi.org/10.1111/nyas.12883

    Article  CAS  PubMed  Google Scholar 

  43. Kingnate C, Charoenkwan K, Kumfu S et al (2018) Possible roles of mitochondrial dynamics and the effects of pharmacological interventions in chemoresistant ovarian cancer. EBioMedicine 34:256–266. https://doi.org/10.1016/j.ebiom.2018.07.026

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lonergan T, Bavister B, Brenner C (2007) Mitochondria in stem cells. Mitochondrion 7:289–296. https://doi.org/10.1016/j.mito.2007.05.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Liao J, Liu PP, Hou G et al (2017) Regulation of stem-like cancer cells by glutamine through β-catenin pathway mediated by redox signaling. Mol Cancer 16:1–13. https://doi.org/10.1186/s12943-017-0623-x

    Article  CAS  Google Scholar 

  46. Li B, Cao Y, Meng G et al (2019) Targeting glutaminase 1 attenuates stemness properties in hepatocellular carcinoma by increasing reactive oxygen species and suppressing Wnt/beta-catenin pathway. EBioMedicine 39:239–254. https://doi.org/10.1016/j.ebiom.2018.11.063

    Article  PubMed  Google Scholar 

  47. Tran TQ, Hanse EA, Habowski AN et al (2020) α-Ketoglutarate attenuates Wnt signaling and drives differentiation in colorectal cancer. Nat Cancer 1:345–358. https://doi.org/10.1038/s43018-020-0035-5

    Article  PubMed  PubMed Central  Google Scholar 

  48. Ju HQ, Lu YX, Chen DL et al (2016) Redox regulation of stem-like cells though the CD44v-xCT axis in colorectal cancer: mechanisms and therapeutic implications. Theranostics 6:1160–1175. https://doi.org/10.7150/thno.14848

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jin L, Alesi GN, Kang S (2016) Glutaminolysis as a target for cancer therapy. Oncogene 35:3619–3625. https://doi.org/10.1038/onc.2015.447

    Article  CAS  PubMed  Google Scholar 

  50. Leone RD, Zhao L, Englert JM et al (2019) Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 366:1013–1021. https://doi.org/10.1126/science.aav2588

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Matés JM, Di Paola FJ, Campos-Sandoval JA et al (2020) Therapeutic targeting of glutaminolysis as an essential strategy to combat cancer. Semin Cell Dev Biol 98:34–43. https://doi.org/10.1016/j.semcdb.2019.05.012

    Article  CAS  PubMed  Google Scholar 

  52. Peiris-Pagès M, Bonuccelli G, Sotgia F, Lisanti MP (2018) Mitochondrial fission as a driver of stemness in tumor cells: mDIVI1 inhibits mitochondrial function, cell migration and cancer stem cell (CSC) signalling. Oncotarget 9(17):13254–13275

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thankfully acknowledge Mr. Sounak Bhattacharya (confocal microscopy), Mr. Tanmoy Dalui, & Mrs. Debalina Chakraborty (flow cytometry), Dr. E. Padmanaban (NMR spectroscopy), and Mr. T. Muruganandan (AFM) of Central Instrumentation Facility of IICB. Prof. Pijush K. Das (CSIR-IICB) and Dr. Partha Chakrabarti (CSIR-IICB) are gratefully acknowledged for their valuable suggestions in preparing the manuscript. We are also thankful to Prof. Susanta Roychoudhury and Dr. Damayanti Das Ghosh (both from Saroj Gupta Cancer Centre & Research Institute, Kolkata, India) for providing human patient tissue samples and also for their valuable suggestions. Technical assistance of Mr. Prabir Kumar Dey is acknowledged. Other members of SSR laboratory are acknowledged for their co-operation.

Funding

This work was supported by grants from Science and Engineering Research Board (SERB) project GAP-360 (EMR/2016/002578) and the Council of Scientific and Industrial Research (CSIR) in house projects.

Author information

Authors and Affiliations

Authors

Contributions

PP, SG and SSR conceptualized the study; PP and SG investigated, performed analysis and validation of the data and wrote the original draft. SSR provided the financial support, supervised the work, reviewed, and approved the final manuscript.

Corresponding author

Correspondence to Sib Sankar Roy.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval for animal studies

All animal experiments were approved by the institutional animal ethics committee (IAEC), CSIR-Indian Institute of Chemical Biology, India, (Registration no. 147/GO/ReBi/S/99/CPSCEA) following the guidelines of Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), Govt. of India.

Ethical approval for human studies

The human patients' tissue samples were collected from Saroj Gupta Cancer Centre and Research Institute (SGCCRI), Kolkata, India with proper human ethics clearance from the Institutional Ethics Committee (approval number -IEC SGCCRI REF NO.- 16/2/2018/Non-Reg/SSR/3).

Consent to participate

Informed written consent was taken from the patients in accordance with the 1964 Helsinki declaration.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PPTX 57972 KB)

Supplementary file1 (MP4 7862 KB)

Supplementary file1 (MP4 8746 KB)

Supplementary file1 (MP4 6095 KB)

Supplementary file1 (MP4 6764 KB)

Supplementary file1 (MP4 2160 KB)

Supplementary file1 (MP4 2042 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Prasad, P., Ghosh, S. & Roy, S.S. Glutamine deficiency promotes stemness and chemoresistance in tumor cells through DRP1-induced mitochondrial fragmentation. Cell. Mol. Life Sci. 78, 4821–4845 (2021). https://doi.org/10.1007/s00018-021-03818-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-021-03818-6

Keywords

Navigation