Skip to main content
Log in

Optical Redox Imaging of Lonidamine Treatment Response of Melanoma Cells and Xenografts

  • Research Article
  • Published:
Molecular Imaging and Biology Aims and scope Submit manuscript

Abstract

Purpose

Fluorescence of co-enzyme reduced nicotinamide adenine dinucleotide (NADH) and oxidized flavoproteins (Fp) provides a sensitive measure of the mitochondrial redox state and cellular metabolism. By imaging NADH and Fp, we investigated the utility of optical redox imaging (ORI) to monitor cellular metabolism and detect early metabolic response to cancer drugs.

Procedures

We performed ORI of human melanoma DB-1 cells in culture and DB-1 mouse xenografts to detect the redox response to lonidamine (LND) treatment.

Results

For cultured cells, LND treatment for 45 min significantly lowered NADH levels with no significant change in Fp, resulting in a significant increase in the Fp redox ratio (Fp/(NADH+Fp)); 3-h prolonged treatment led to a decrease in NADH and an increase in Fp and a more oxidized redox state compared to control. Significant decrease in the mitochondrial redox capacity of LND-treated cells was observed for the first time. For xenografts, 45-min LND treatment resulted in a significant reduction of NADH content, no significant changes in Fp content, and a trend of increase in the Fp redox ratio. Intratumor redox heterogeneity was observed in both control and LND-treated groups.

Conclusion

Our results support the utility of ORI for evaluating cellular metabolism and monitoring early metabolic response to cancer drugs.

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

Similar content being viewed by others

References

  1. DeBerardinis RJ, Chandel NS (2016) Fundamentals of cancer metabolism. Sci Adv 2:e1600200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chance B, Cohen P, Jobsis F, Schoener B (1962) Intracellular oxidation-reduction states in vivo. Science 137:499–508

    Article  CAS  PubMed  Google Scholar 

  3. Chance B, Ernster L, Garland PB, Lee CP, Light PA, Ohnishi T, Ragan CI, Wong D (1967) Flavoproteins of the mitochondrial respiratory chain. Proc Natl Acad Sci U S A 57:1498–1505

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hassinen I, Chance B (1968) Oxidation-reduction properties of the mitochondrial flavoprotein chain. Biochem Biophys Res Commun 31:895–900

    Article  CAS  PubMed  Google Scholar 

  5. Garland PB, Chance B, Ernster L, Lee CP, Wong D (1967) Flavoproteins of mitochondrial fatty acid oxidation. Proc Natl Acad Sci U S A 58:1696–1702

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ragan CI, Garland PB (1969) The intra-mitochondrial localization of flavoproteins previously assigned to the respiratory chain. Eur J Biochem 10:399–410

    Article  CAS  PubMed  Google Scholar 

  7. Kunz WS (1986) Spectral properties of fluorescent flavoproteins of isolated rat liver mitochondria. FEBS Lett 195:92–96

    Article  CAS  PubMed  Google Scholar 

  8. Kunz WS (1988) Evaluation of electron-transfer flavoprotein and alpha-lipoamide dehydrogenase redox states by two-channel fluorimetry and its application to the investigation of beta-oxidation. Biochim Biophys Acta 932:8–16

    Article  CAS  PubMed  Google Scholar 

  9. Kunz WS, Kunz W (1985) Contribution of different enzymes to flavoprotein fluorescence of isolated rat liver mitochondria. Biochim Biophys Acta 841:237–246

    Article  CAS  PubMed  Google Scholar 

  10. Chance B, Schoener B, Oshino R, Itshak F, Nakase Y (1979) Oxidation-reduction ratio studies of mitochondria in freeze-trapped samples. NADH and flavoprotein fluorescence signals. J Biol Chem 254:4764–4771

    CAS  Google Scholar 

  11. Heikal AA (2010) Intracellular coenzymes as natural biomarkers for metabolic activities and mitochondrial anomalies. Biomark Med 4:241–263

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Varone A, Xylas J, Quinn KP, Pouli D, Sridharan G, McLaughlin-Drubin ME, Alonzo C, Lee K, Munger K, Georgakoudi I (2014) Endogenous two-photon fluorescence imaging elucidates metabolic changes related to enhanced glycolysis and glutamine consumption in precancerous epithelial tissues. Cancer Res 74:3067–3075

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ozawa K, Chance B, Tanaka A, Iwata S, Kitai T, Ikai I (1992) Linear correlation between acetoacetate/beta-hydroxybutyrate in arterial blood and oxidized flavoprotein/reduced pyridine nucleotide in freeze-trapped human liver tissue. Biochim Biophys Acta 1138:350–352

    Article  CAS  PubMed  Google Scholar 

  14. Chance B, Baltscheffsky H (1958) Respiratory enzymes in oxidative phosphorylation. VII Binding of intramitochondrial reduced pyridine nucleotide. J Biol Chem 233:736–739

    CAS  PubMed  Google Scholar 

  15. Chance B, Schoener B (1966) Fluorometric studies of flavin component of the respiratory chain. Flavins Flavoproteins 81:510–519

    Google Scholar 

  16. Xu HN, Tchou J, Feng M, Zhao H, Li LZ (2016) Optical redox imaging indices discriminate human breast cancer from normal tissues. J Biomed Opt 21:114003

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kirkpatrick ND, Brewer MA, Utzinger U (2007) Endogenous optical biomarkers of ovarian cancer evaluated with multiphoton microscopy. Cancer Epidemiol Biomark Prev 16:2048–2057

    Article  CAS  Google Scholar 

  18. Li LZ, Zhou R, Xu HN, Moon L, Zhong T, Kim EJ, Qiao H, Reddy R, Leeper D, Chance B, Glickson JD (2009) Quantitative magnetic resonance and optical imaging biomarkers of melanoma metastatic potential. Proc Natl Acad Sci U S A 106:6608–6613

    Article  PubMed  PubMed Central  Google Scholar 

  19. Xu HN, Nioka S, Glickson JD, Chance B, Li LZ (2010) Quantitative mitochondrial redox imaging of breast cancer metastatic potential. J Biomed Opt 15:036010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sun N, Xu HN, Luo Q, Li LZ (2016) Potential indexing of the invasiveness of breast cancer cells by mitochondrial redox ratios. Adv Exp Med Biol 923:121–127

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Alhallak K, Rebello LG, Muldoon TJ, Quinn KP, Rajaram N (2016) Optical redox ratio identifies metastatic potential-dependent changes in breast cancer cell metabolism. Biomed Opt Express 7:4364–4374

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ostrander JH, McMahon CM, Lem S et al (2010) Optical redox ratio differentiates breast cancer cell lines based on estrogen receptor status. Cancer Res 70:4759–4766

    Article  CAS  Google Scholar 

  23. Xu HN, Feng M, Moon L, Dolloff N, El-Deiry W, Li LZ (2013) Redox imaging of the p53-dependent mitochondrial redox state in colon cancer ex vivo. J Innov Opt Health Sci 06:1350016

    Article  CAS  Google Scholar 

  24. Xu HN, Nioka S, Li LZ (2013) Imaging heterogeneity in the mitochondrial redox state of premalignant pancreas in the pancreas-specific PTEN-null transgenic mouse model. Biomarker Res 1:6

    Article  Google Scholar 

  25. Xu HN, Zhao H, Mir TA et al (2013) Chop therapy induced mitochondrial redox state alteration in non-Hodgkin’s lymphoma xenografts. J Innov Opt Health Sci 6:1350011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shah AT, Beckler MD, Walsh AJ, Jones WP, Pohlmann PR, Skala MC (2014) Optical metabolic imaging of treatment response in human head and neck squamous cell carcinoma. PLoS One 9:e90746

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Walsh AJ, Cook RS, Sanders ME, Aurisicchio L, Ciliberto G, Arteaga CL (2014) Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res 74:5184–5194

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Walsh AJ, Cook RS, Manning HC, Hicks DJ, Lafontant A, Arteaga CL, Skala MC (2013) Optical metabolic imaging identifies glycolytic levels, subtypes, and early-treatment response in breast cancer. Cancer Res 73:6164–6174

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Alam SR, Wallrabe H, Svindrych Z, Chaudhary AK, Christopher KG, Chandra D, Periasamy A (2017) Investigation of mitochondrial metabolic response to doxorubicin in prostate cancer cells: an NADH, FAD and tryptophan FLIM assay. Sci Rep 7:10451

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kirkpatrick ND, Zou C, Brewer MA, Brands WR, Drezek RA, Utzinger U (2005) Endogenous fluorescence spectroscopy of cell suspensions for chemopreventive drug monitoring. Photochem Photobiol 81:125–134

    Article  CAS  PubMed  Google Scholar 

  31. Cannon TM, Shah AT, Walsh AJ, Skala MC (2015) High-throughput measurements of the optical redox ratio using a commercial microplate reader. J Biomed Opt 20:010503

  32. Cervantes-Madrid D, Romero Y, Due, et al. (2015) Reviving lonidamine and 6-Diazo-5-oxo-L-norleucine to be used in combination for metabolic cancer therapy. BioMed Research International 2015:690492

  33. Floridi A, Lehninger AL (1983) Action of the antitumor and antispermatogenic agent lonidamine on electron transport in ehrlich ascites tumor mitochondria. Arch Biochem Biophys 226:73–83

    Article  CAS  PubMed  Google Scholar 

  34. Nath K, Nelson DS, Ho A et al (2013) 31P and 1H MRS of DB-1 melanoma xenografts: lonidamine selectively decreases tumor intracellular pH and energy status and sensitizes tumors to melphalan. NMR Biomed 26:98–105

    Article  CAS  PubMed  Google Scholar 

  35. Nath K, Nelson DS, Heitjan DF, Zhou R, Leeper DB, Glickson JD (2015) Effects of hyperglycemia on lonidamine-induced acidification and de-energization of human melanoma xenografts and sensitization to melphalan. NMR Biomed 28:395–403

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nath K, Nelson DS, Putt ME, Leeper DB, Garman B, Nathanson KL, Glickson JD (2016) Comparison of the lonidamine potentiated effect of nitrogen mustard alkylating agents on the systemic treatment of DB-1 human melanoma xenografts in mice. PLoS One 11:e0157125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Quistorff B, Haselgrove JC, Chance B (1985) High spatial resolution readout of 3-D metabolic organ structure: an automated, low-temperature redox ratio-scanning instrument. Anal Biochem 148:389–400

    Article  CAS  PubMed  Google Scholar 

  38. Li LZ, Xu HN, Ranji M et al (2009) Mitochondrial redox imaging for cancer diagnostic and therapeutic studies. J Innov Optical Health Sci 2:325–341

    Article  Google Scholar 

  39. Xu HN, Zheng G, Tchou J, Nioka S, Li LZ (2013) Characterizing the metabolic heterogeneity in human breast cancer xenografts by 3D high resolution fluorescence imaging. SpringerPlus 2:73

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xu HN, Zhou R, Moon L, Feng M, Li LZ (2014) 3D imaging of the mitochondrial redox state of rat hearts under normal and fasting conditions. J Innov Opt Health Sci 7:1350045

    Article  CAS  PubMed  Google Scholar 

  41. Huang S, Heikal AA, Webb WW (2002) Two-photon fluorescence spectroscopy and microscopy of NAD(P)H and flavoprotein. Biophys J 82:2811–2825

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bartolome F, Abramov AY (2015) Measurement of mitochondrial NADH and FAD autofluorescence in live cells. Methods Mol Biol 1264:263–270

    Article  CAS  PubMed  Google Scholar 

  43. Yu Q, Heikal AA (2009) Two-photon autofluorescence dynamics imaging reveals sensitivity of intracellular NADH concentration and conformation to cell physiology at the single-cell level. J Photochem Photobiol B Biol 95:46–57

    Article  CAS  Google Scholar 

  44. Blinova K, Levine RL, Boja ES, Griffiths GL, Shi ZD, Ruddy B, Balaban RS (2008) Mitochondrial NADH fluorescence is enhanced by complex I binding. Biochemistry 47:9636–9645

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Luo X, Li R, Yan LJ (2015) Roles of pyruvate, NADH, and mitochondrial complex I in redox balance and imbalance in beta cell function and dysfunction. J Diabetes Res 2015:512618

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Berg JM, Tymoczko JL, Stryer L (2002) Many shuttles allow movement across the mitochondrial membranes. In: Biochemistry. 5th edition. New York: W H freeman; section 18.5, Available from: https://www.ncbi.nlm.nih.gov/books/NBK22470/

  47. Nath K, Guo L, Nancolas B et al (2016) Mechanism of antineoplastic activity of lonidamine. Biochim Biophys Acta Rev Cancer 1866:151–162

    Article  CAS  Google Scholar 

  48. Guo L, Shestov AA, Worth AJ, Nath K, Nelson DS, Leeper DB, Glickson JD, Blair IA (2016) Inhibition of mitochondrial complex II by the anticancer agent lonidamine. J Biol Chem 291:42–57

    Article  CAS  PubMed  Google Scholar 

  49. Nancolas B, Guo L, Zhou R, Nath K, Nelson DS, Leeper DB, Blair IA, Glickson JD, Halestrap AP (2016) The anti-tumour agent lonidamine is a potent inhibitor of the mitochondrial pyruvate carrier and plasma membrane monocarboxylate transporters. Biochem J 473:929–936

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kunz WS, Gellerich FN (1993) Quantification of the content of fluorescent flavoproteins in mitochondria from liver, kidney cortex, skeletal muscle, and brain. Biochem Med Metab Biol 50:103–110

    Article  CAS  Google Scholar 

  51. Heaster TM, Walsh AJ, Zhao Y, et al. (2018) Autofluorescence imaging identifies tumor cell-cycle status on a single-cell level. J Biophotonics 11:e201600276.

  52. Pal R, Mamidi MK, Das AK, Bhonde R (2012) Diverse effects of dimethyl sulfoxide (DMSO) on the differentiation potential of human embryonic stem cells. Arch Toxicol 86:651–661

    Article  CAS  PubMed  Google Scholar 

  53. Yuan C, Gao J, Guo J, Bai L, Marshall C, Cai Z, Wang L, Xiao M (2014) Dimethyl sulfoxide damages mitochondrial integrity and membrane potential in cultured astrocytes. PLoS One 9:e107447

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wang C-C, Lin S-Y, Lai Y-H, Liu YJ, Hsu YL, Chen JJW (2012) Dimethyl sulfoxide promotes the multiple functions of the tumor suppressor HLJ1 through activator Protein-1 activation in NSCLC cells. PLoS One 7:e33772

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Saunders NA, Simpson F, Thompson EW, Hill MM, Endo-Munoz L, Leggatt G, Minchin RF, Guminski A (2012) Role of intratumoural heterogeneity in cancer drug resistance: molecular and clinical perspectives. EMBO Mol Med 4:675–684

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lopes-Rodrigues V, Di Luca A, Mleczko J et al (2017) Identification of the metabolic alterations associated with the multidrug resistant phenotype in cancer and their intercellular transfer mediated by extracellular vesicles. Sci Rep 7:44541

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the NIH Grants R01CA155348 (L.Z. Li), R01CA191207 (L.Z. Li), R01-CA129544 (J. Glickson), and R01CA172820 (J. Glickson). We would also like to thank Ms. Lily Moon for technical assistance with mouse xenografts.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to He N. Xu or Lin Z. Li.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Electronic supplementary material

ESM 1

(PDF 230 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, H.N., Feng, M., Nath, K. et al. Optical Redox Imaging of Lonidamine Treatment Response of Melanoma Cells and Xenografts. Mol Imaging Biol 21, 426–435 (2019). https://doi.org/10.1007/s11307-018-1258-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11307-018-1258-z

Keywords

Navigation