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.

  • Letter
  • Published:

An engineered enzyme that targets circulating lactate to alleviate intracellular NADH:NAD+ imbalance

Abstract

An elevated intracellular NADH:NAD+ ratio, or ‘reductive stress’, has been associated with multiple diseases, including disorders of the mitochondrial electron transport chain. As the intracellular NADH:NAD+ ratio can be in near equilibrium with the circulating lactate:pyruvate ratio, we hypothesized that reductive stress could be alleviated by oxidizing extracellular lactate to pyruvate. We engineered LOXCAT, a fusion of bacterial lactate oxidase (LOX) and catalase (CAT), which irreversibly converts lactate and oxygen to pyruvate and water. Addition of purified LOXCAT to the medium of cultured human cells with a defective electron transport chain decreased the extracellular lactate:pyruvate ratio, normalized the intracellular NADH:NAD+ ratio, upregulated glycolytic ATP production and restored cellular proliferation. In mice, tail-vein-injected LOXCAT lowered the circulating lactate:pyruvate ratio, blunted a metformin-induced rise in blood lactate:pyruvate ratio and improved NADH:NAD+ balance in the heart and brain. Our study lays the groundwork for a class of injectable therapeutic enzymes that alleviates intracellular redox imbalances by directly targeting circulating redox-coupled metabolites.

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: Effects of exogenous LOX and CAT, alone or in combination, on intracellular NADH:NAD+.
Fig. 2: Design and characterization of LOXCAT, a water-forming lactate oxidase.
Fig. 3: Effect of adding LOXCAT to the medium of cellular models of mitochondrial disease.
Fig. 4: Impact of intravenous LOXCAT on blood lactate:pyruvate and tissue NADH:NAD+ ratios.

Similar content being viewed by others

Data availability

All data are presented in the paper and supplementary files. Supporting data will be provided upon reasonable request.

References

  1. Vafai, S. B. & Mootha, V. K. Mitochondrial disorders as windows into an ancient organelle. Nature 491, 374–383 (2012).

    Article  CAS  Google Scholar 

  2. Thompson Legault, J. et al. A metabolic signature of mitochondrial dysfunction revealed through a monogenic form of Leigh syndrome. Cell Rep. 13, 981–989 (2015).

    Article  CAS  Google Scholar 

  3. Titov, D. V. et al. Complementation of mitochondrial electron transport chain by manipulation of the NAD+/NADH ratio. Science 352, 231–235 (2016).

    Article  CAS  Google Scholar 

  4. Xiao, W. & Loscalzo, J. Metabolic responses to reductive stress. Antioxid. Redox Signal https://doi.org/10.1089/ars.2019.7803 (2019).

  5. Gores, G. J. et al. Swelling, reductive stress, and cell death during chemical hypoxia in hepatocytes. Am. J. Physiol. 257, C347–C354 (1989).

    Article  CAS  Google Scholar 

  6. Tilton, W. M., Seaman, C., Carriero, D. & Piomelli, S. Regulation of glycolysis in the erythrocyte: role of the lactate/pyruvate and NAD/NADH ratios. J. Lab. Clin. Med. 118, 146–152 (1991).

    PubMed  CAS  Google Scholar 

  7. Williamson, J. R. et al. Hyperglycemic pseudohypoxia and diabetic complications. Diabetes 42, 801–813 (1993).

    Article  CAS  Google Scholar 

  8. Williamson, D. H., Lund, P. & Krebs, H. A. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem. J. 103, 514–527 (1967).

    Article  CAS  Google Scholar 

  9. Poole, R. C. & Halestrap, A. P. Transport of lactate and other monocarboxylates across mammalian plasma membranes. Am. J. Physiol. 264, C761–C782 (1993).

    Article  CAS  Google Scholar 

  10. Shaham, O. et al. A plasma signature of human mitochondrial disease revealed through metabolic profiling of spent media from cultured muscle cells. Proc. Natl Acad. Sci. USA 107, 1571–1575 (2010).

    Article  Google Scholar 

  11. Bucher, T. et al. State of oxidation-reduction and state of binding in the cytosolic NADH-system as disclosed by equilibration with extracellular lactate-pyruvate in hemoglobin-free perfused rat liver. Eur. J. Biochem. 27, 301–317 (1972).

    Article  CAS  Google Scholar 

  12. Hung, Y. P., Albeck, J. G., Tantama, M. & Yellen, G. Imaging cytosolic NADH-NAD(+) redox state with a genetically encoded fluorescent biosensor. Cell Metab. 14, 545–554 (2011).

    Article  CAS  Google Scholar 

  13. Fujii, T. et al. Efficacy of pyruvate therapy in patients with mitochondrial disease: a semi-quantitative clinical evaluation study. Mol. Genet. Metab. 112, 133–138 (2014).

    Article  CAS  Google Scholar 

  14. Saito, K. et al. Pyruvate therapy for mitochondrial DNA depletion syndrome. Biochim. Biophys. Acta 1820, 632–636 (2012).

    Article  CAS  Google Scholar 

  15. Li, S. J. et al. Crystallographic study on the interaction of l-lactate oxidase with pyruvate at 1.9 Angstrom resolution. Biochem. Biophys. Res. Comm. 358, 1002–1007 (2007).

    Article  CAS  Google Scholar 

  16. Melik-Adamyan, W. et al. Substrate flow in catalases deduced from the crystal structures of active site variants of HPII from Escherichia coli. Proteins 44, 270–281 (2001).

    Article  CAS  Google Scholar 

  17. Taurino, I. R. R. et al. Comparative study of three lactate oxidases from Aerococcus viridans for biosensing applications. Electrochim. Acta 93, 72–79 (2013).

    Article  CAS  Google Scholar 

  18. Andre, C., Kim, S. W., Yu, X. H. & Shanklin, J. Fusing catalase to an alkane-producing enzyme maintains enzymatic activity by converting the inhibitory byproduct H2O2 to the cosubstrate O2. Proc. Natl Acad. Sci. USA 110, 3191–3196 (2013).

    Article  Google Scholar 

  19. Martin, A., Baker, T. A. & Sauer, R. T. Rebuilt AAA+ motors reveal operating principles for ATP-fuelled machines. Nature 437, 1115–1120 (2005).

    Article  Google Scholar 

  20. Atkinson, D. E. & Walton, G. M. Adenosine triphosphate conservation in metabolic regulation. Rat liver citrate cleavage enzyme. J. Biol. Chem. 242, 3239–3241 (1967).

    PubMed  CAS  Google Scholar 

  21. De la Fuente, I. M. et al. On the dynamics of the adenylate energy system: homeorhesis vs homeostasis. PLoS ONE 9, e108676 (2014).

    Article  CAS  Google Scholar 

  22. King, M. P. & Attardi, G. Human cells lacking mtDNA: repopulation with exogenous mitochondria by complementation. Science 246, 500–503 (1989).

    Article  CAS  Google Scholar 

  23. Baertling, F. et al. NDUFA9 point mutations cause a variable mitochondrial complex I assembly defect. Clin. Genet. 93, 111–118 (2018).

    Article  CAS  Google Scholar 

  24. Haut, S. et al. A deletion in the human QP-C gene causes a complex III deficiency resulting in hypoglycaemia and lactic acidosis. Hum. Genet. 113, 118–122 (2003).

    Article  CAS  Google Scholar 

  25. Jonckheere, A. I. et al. A complex V ATP5A1 defect causes fatal neonatal mitochondrial encephalopathy. Brain 136, 1544–1554 (2013).

    Article  Google Scholar 

  26. Johnson, S. C. et al. mTOR inhibitors may benefit kidney transplant recipients with mitochondrial diseases. Kidney Int. 95, 455–466 (2019).

    Article  CAS  Google Scholar 

  27. Mukherjee, J. et al. Prolonged prophylactic protection from botulism with a single adenovirus treatment promoting serum expression of a VHH-based antitoxin protein. PLoS ONE 9, e106422 (2014).

    Article  Google Scholar 

  28. Nguyen, A. et al. The pharmacokinetics of an albumin-binding Fab (AB.Fab) can be modulated as a function of affinity for albumin. Protein Eng. Des. Sel. 19, 291–297 (2006).

    Article  CAS  Google Scholar 

  29. Gui, D. Y. et al. Environment dictates dependence on mitochondrial complex I for NAD+ and aspartate production and determines cancer cell sensitivity to metformin. Cell Metab. 24, 716–727 (2016).

    Article  CAS  Google Scholar 

  30. Nocito, L. et al. The extracellular redox state modulates mitochondrial function, gluconeogenesis, and glycogen synthesis in murine hepatocytes. PLoS ONE 10, e0122818 (2015).

    Article  CAS  Google Scholar 

  31. Ramirez, A. et al. Extracellular cysteine/cystine redox potential controls lung fibroblast proliferation and matrix expression through upregulation of transforming growth factor-β. Am. J. Physiol. Lung Cell. Mol. Physiol. 293, L972–L981 (2007).

    Article  CAS  Google Scholar 

  32. Muller, H. J. & Boos, J. Use of l-asparaginase in childhood ALL. Crit. Rev. Oncol. Hematol. 28, 97–113 (1998).

    Article  CAS  Google Scholar 

  33. Laffel, G. L. & Braunwald, E. Thrombolytic therapy. A new strategy for the treatment of acute myocardial infarction (2). N. Engl. J. Med. 311, 770–776 (1984).

    Article  CAS  Google Scholar 

  34. Arroyo, J. D. et al. A genome-wide CRISPR death screen identifies genes essential for oxidative phosphorylation. Cell Metab. 24, 875–885 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank O. Goldberger, K.J. Kamer and J.D. Arroyo for technical assistance. This work was supported in part by grants from the Marriott Foundation and the National Institutes of Health (R35GM122455). A.P. was supported by the Helen Hay Whitney Postdoctoral Fellowship and the Tosteson and Fund for Medical Discovery award. O.S.S. was supported by a F32 Fellowship from the National Institute of General Medical Sciences (1F32GM133047-01). V.K.M. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

A.P., X.R.B. and V.K.M. designed the study. A.P. performed and analyzed the biochemical and cell culture experiments. A.P., O.S., H.S. and R.S. performed and analyzed the LC–MS experiments. A.P., Y.M., G.S., E.M. and R.P.G. performed the mouse experiments and analyzed data. T.-L.T assisted with the K562 KO cellular experiments. F.I. and W.Z. supervised mouse studies. V.K.M. supervised the study. A.P. and V.K.M. wrote the manuscript.

Corresponding author

Correspondence to Vamsi K. Mootha.

Ethics declarations

Competing interests

Massachusetts General Hospital has filed a patent on the technology described in this paper listing V.K.M., A.P. and X.R.B as inventors. O.S.S. is a paid consultant for Proteinaceous Inc. V.K.M. is a paid advisor to Janssen Pharmaceuticals and 5AM Ventures and is a founder of Raze Therapeutics.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Effect of media supplemented with lactate oxidase (LOX), catalase (CAT), or both, on the growth of cells with electron transport chain dysfunction.

Growth curve for HeLa cells treated with antimycin A in the presence of LOX, CAT, a combination of LOX and CAT, or pyruvate. Data are mean ± S.D., n = 4 total wells from three biologically independent experiments.

Supplementary Figure 2 SDS-PAGE gels of purified LOXCAT, LOXCATmut, ABP-LOXCAT, and ABP-LOXCATmut.

SDS-PAGE gels of purified (a) LOXCAT and LOXCATmut, and (b) ABP-LOXCAT and ABP-LOXCATmut. The predicted molecular weight of these proteins is ~132 kDa. The data shown are representative of at least two gels from independent protein preparations with similar results.

Supplementary Figure 3 Oxygen consumption by LOX, LOXCAT, and LOXCATmut.

Representative measurements of oxygen consumption by LOX, LOXCAT, and LOXCATmut with lactate as a substrate. The bump in the LOX curve after catalase addition represents a temporary increase in the O2 concentrations due to H2O2 breakdown by catalase. Repeated in n = 2 biologically independent experiments with similar results.

Supplementary Figure 4 Effect of extracellular LOXCAT on the intracellular NADH/NAD+ ratio measured by LCMS.

Effect of media supplemented with LOXCAT, LOXCATmut, or pyruvate on the total cellular NADH/NAD+ ratios measured by LCMS in HeLa cells treated with antimycin A for 24 h. Data are mean ± S.D., n = 6 total wells from 3 biologically independent experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P = 0.99; and ****P < 0.0001.

Supplementary Figure 5 Effect of extracellular LOXCAT on media lactate and pyruvate.

Effect of extracellular LOXCAT, LOXCATmut, and pyruvate on the media levels of (a) lactate and (b) pyruvate in control, or antimycin A-treated HeLa cells at 24 h. Data are mean ± S.D., n = 16 total wells in antimycin-only treatment, n = 12 total wells in untreated and antimycin + pyruvate treatments, and n=10 total wells in antimycin + LOXCAT and antimycin + LOXCATmut treatments from 5 biologically independent experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P > 0.99; and P = 0.079; ***P = 0.0001 and 0.0006; and ****P < 0.0001.

Supplementary Figure 6 Effect of LOXCAT on glycolytic intermediates in antimycin A-treated HeLa cells.

Effect of media pyruvate, LOXCAT, or LOXCATmut on glycolytic intermediates in antimycin-treated HeLa cells at 24h. Data are mean ± S.D., n = 6 total wells from 3 biologically independent experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P = 0.99; **P = 0.0018, 0.0013, and 0.0027; ***P = 0.0001; and ****P < 0.0001.

Supplementary Figure 7 Morphology of antimycin A-treated HeLa cells with pyruvate, LOXCAT, or LOXCATmut.

Images of HeLa cells after 72h treatment with antimycin A with media containing pyruvate, LOXCAT or LOXCATmut. Shown are representative images of three biologically independent experiments with similar results.

Supplementary Figure 8 Effect of extracellular LOXCAT on the proliferation of cells with electron transport chain (ETC) deficiency.

Effect of extracellular LOXCAT on the proliferation of (a) HeLa cells treated with piericidin A or oligomycin A, (b) K562 UQCRB KO cells, and (c) K562 ATP5A1 KO cells at 72 h. Data are mean ± S.D., n = 6 total wells from 3 biologically independent experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P = 0.98, > 0.99 and 0.83; ***P = 0.0002, 0.0001, and 0.0001; and ****P < 0.0001.

Supplementary Figure 9 Effect of blood LOXCAT on the circulating [lactate]/[pyruvate] ratio in vivo.

Effect of intravenous LOXCAT on the circulating [lactate]/[pyruvate] ratios in wild-type C57BL/6J mice at 15 min. Data are mean ± S.D., n = 3 mice per treatment. Molar ratios of lactate and pyruvate are shown. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P = 0.14; ***P = 0.001 and 0.0002.

Supplementary Figure 10 Pharmacokinetics and pharmacodynamics of ABP-LOXCAT.

(a) Circulatory half-life of LOXCAT vs ABP-LOXCAT in wild-type C57BL/6J mice measured by western blot (data shown are representative of 2 blots from two mice with similar results) (b) Enzyme activity of ABP-LOXCAT vs ABP-LOXCATmut over 300 minutes in the circulation of wild-type C57BL/6J mice. Absolute concentrations of lactate and pyruvate were measured using LCMS. Data are mean ± S.D., n = 3 mice per timepoint. Two-way ANOVA followed by Sidak multiple comparisons test. ns, P = 0.59 and 0.99; **P = 0.0016 and 0.0079; and ****P < 0.0001.

Supplementary Figure 11 Full scans of all gels and blots used in the manuscript.

(a) Coomassie-stained SDS-PAGE gel of purified LOXCAT and LOXCATmut. (b) Coomassie-stained SDS-PAGE gel of purified ABP-LOXCAT and ABP-LOXCATmut. (c) Western blot detection of LOXCAT (anti-S-tag) from whole blood. (d) Western blot detection of mouse serum albumin (anti-albumin) from whole blood as loading control for LOXCAT. (e) Western blot detection of ABP-LOXCAT (anti-S-tag) from whole blood. (f) Western blot detection of mouse serum albumin (anti-albumin) from whole blood as loading control for ABP-LOXCAT. (g) Western blot detection of ATP5A1 (anti-ATP5A1) in K562 KO cells. (h) Western blot detection of actin (anti-actin) in K562 ATP5A1 KO cells. (i) Western blot detection of UQCRB (anti-UQCRB) in K562 KO cells. (j) Western blot detection of actin (anti-actin) in K562 UQCRB KO cells. All data shown are representative of two independent blots and at least two independent gels with similar results.

Supplementary Figure 12 Ability of LOXCAT and LOXCATmut to consume H2O2.

H2O2 measurements after (a) lactate and (b) H2O2 incubation with ABP-LOXCAT or ABP-LOXCATmut. Data are mean ± S.D., n = 4 biologically independent samples in panel (a) and n = 3 biologically independent samples in panel (b). One-way ANOVA followed by Tukey’s multiple comparisons test. **P = 0.0076 and 0.0077; and ****P < 0.0001.

Supplementary Figure 13 Effect of extracellular pyruvate, ABP-LOXCAT, or ABP-LOXCATmut on the proliferation of antimycin A-treated HeLa cells.

Effect of media supplemented with ABP-LOXCAT, ABP-LOXCATmut, and pyruvate on proliferative defects in HeLa cells treated with antimycin A at 72h. Data are mean ± S.D., n = 6 total wells from 3 biologically independent experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P = 0.99; and ****P < 0.0001.

Supplementary Figure 14 Effect of intravenous ABP-LOXCAT on tissue [NADH], [NAD+], and [NADH]/[NAD+] in metformin-treated mice.

Effect of circulating ABP-LOXCAT on (a) brain [NADH], (b) brain [NAD+], (c) heart [NADH], (d) heart [NAD+], (e) liver [NADH]/[NAD+] ratio, and (f) kidney [NADH]/[NAD+] ratio in metformin-treated mice. Molar ratios of NADH and NAD+ are shown. Data are mean ± S.D., n = 13 mice. One-way ANOVA followed by Tukey’s multiple comparisons test. ns, P = 0.82, 0.15, 0.17, 0.90, 0.47, 0.50, 0.99, 0.91, 0.97, 0.99, 0.23, 0.16, 0.083, 0.86, 0.69, 0.053, 0.98, 0.99; *P = 0.015; ***P = 0.0001, 0.0002 ; and ****P < 0.0001.

Supplementary Figure 15 Western blots for validation of K562 UQCRB KO and ATP5A1 KO cells.

Validation of K562 UQCRB KO and ATP5A1 KO cell lines by western blots with the following antibodies: anti-UQCRB (Abcam ab190360), anti-ATP5A1 (Abcam ab110411), and anti-β-Actin (A5441 Sigma). Shown are representative blots from two independent experiments with similar results.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Patgiri, A., Skinner, O.S., Miyazaki, Y. et al. An engineered enzyme that targets circulating lactate to alleviate intracellular NADH:NAD+ imbalance. Nat Biotechnol 38, 309–313 (2020). https://doi.org/10.1038/s41587-019-0377-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0377-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing