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.

  • Protocol
  • Published:

High-throughput screening of tissue-nonspecific alkaline phosphatase for identification of effectors with diverse modes of action

Abstract

Here we describe a protocol for the identification of effectors of tissue-nonspecific alkaline phosphatase (TNAP). It is based on a highly sensitive method for detecting TNAP activity. After dephosphorylation by TNAP, a dioxetane-based substrate undergoes a series of chemical transformations resulting in light production. Light intensity serves as a quantitative measure of the velocity of the TNAP-catalyzed reaction in the steady state. This protocol includes guidelines for optimizing the assay and for high-throughput screening in multiwell plates. The assay is sensitive to the influence of diverse effectors of TNAP as long as the assay optimization steps are repeated for each new batch of the enzyme; full optimization is accomplished in under 2 d. Depending on the available equipment, 10,000–100,000 compounds can be screened in an 8-h period. This protocol provides a method of screening TNAP that is 1,000-fold more sensitive and 10-fold faster than a conventional colorimetric assay with p-nitrophenyl phosphate.

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

Figure 1: Catalytic mechanism of alkaline phosphatase reaction11.
Figure 2: Structures of two commonly used alkaline phosphatase substrates.
Figure 3: Luminescence-based assay for TNAP.
Figure 4: Optimization of CDP-Star concentration.
Figure 5: Optimization of DEA concentration.
Figure 6: Optimization of TNAP concentration in the luminescence assay.
Figure 7: Inhibition of TNAP activity by levamisole.
Figure 8: Z′-factor study of the optimized TNAP luminescence assay.

Similar content being viewed by others

References

  1. McComb, R.B., Bowers, G.N., Jr. & Posen, S. Alkaline Phosphatase (Plenum Press, New York, USA, 1979).

  2. Millán, J.L. Mammalian Alkaline Phosphatases. From Biology to Applications in Medicine and Biotechnology (Wiley-VCH Verlag, Weinheim, Germany, 2006) pp. 1–322.

  3. Millán, J.L. et al. Enzyme replacement therapy for murine hypophosphatasia. J. Bone Miner. Res. 23, 777–787 (2008).

    Article  Google Scholar 

  4. Fedde, K.N. et al. Alkaline phosphatase knock-out mice recapitulate the metabolic and skeletal defects of infantile hypophosphatasia. J. Bone Miner. Res. 14, 2015–2026 (1999).

    Article  CAS  Google Scholar 

  5. Whyte, M.P. et al. Perinatal hypophosphatasia: tissue levels of vitamin B6 are unremarkable despite markedly increased circulating concentrations of pyridoxal-5/-phosphate. Evidence for an ectoenzyme role for tissue-nonspecific alkaline phosphatase. J. Clin. Invest. 81, 1234–1239 (1988).

    Article  CAS  Google Scholar 

  6. Holtz, K.M., Stec, B. & Kantrowitz, E.R. A model of the transition state in the alkaline phosphatase reaction. J. Biol. Chem. 274, 8351–8354 (1999).

    Article  CAS  Google Scholar 

  7. Stinson, R.A. Kinetic parameters for the cleaved substrate, and enzyme and substrate stability, vary with the phosphoacceptor in alkaline phosphatase catalysis. Clin. Chem. 39, 2293–2297 (1993).

    CAS  PubMed  Google Scholar 

  8. Fishman, W.H. & Sie, H.-G. Organ-specific inhibition of human alkaline phosphatase isoenzymes of liver, bone, intestine and placenta; L-phenylalanine, L-tryptophan and L-homoarginine. Enzymologia 41, 140–167 (1971).

    Google Scholar 

  9. Van Belle, H. Alkaline phosphatase. I. Kinetics and inhibition by levamisole of purified isoenzymes from humans. Clin. Chem. 22, 972–976 (1976).

    CAS  PubMed  Google Scholar 

  10. Farley, J.R., Ivey, J.L. & Baylink, D.J. Human skeletal alkaline phosphatase—kinetic studies including pH dependence and inhibition by theophylline. J. Biol. Chem. 255, 4680–4686 (1980).

    CAS  PubMed  Google Scholar 

  11. Sergienko, E. et al. Identification and characterization of novel tissue-nonspecific alkaline phosphatase inhibitors with diverse modes of action. J. Biomol. Screen 14, 824–837 (2009).

    Article  CAS  Google Scholar 

  12. Ardecky, R. et al. Design and synthesis of pyrazole derivatives as potent and selective inhibitors of tissue-nonspecific alkaline phosphatase (TNAP). Bioorg. Med. Chem. Lett. 19, 222–225 (2009).

    Article  Google Scholar 

  13. Brooks, E. & Voyta, J.C. Purification of stable water-soluble dioxetanes. US patent 4,931,569, filed 14 September 1988, and issued 5 June 1990 (1990).

  14. Bronstein, I., Brooks, E. & Rouh-Rong, Juo Chemiluminescent 3-(substituted adamant-2′-ylidene) 1,2-dioxetanes. US patent 5,326,882, filed 30 April, 1992, and issued 5 July 1994 (1994).

  15. Zhang, J.H., Chung, T.D. & Oldenburg, K.R. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J. Biomol. Screen 4, 67–73 (1999).

    Article  CAS  Google Scholar 

  16. Narisawa, S. et al. High throughput screening and identification of novel alkaline phosphatase inhibitors—a druggable target for the treatment of vascular calcification. J. Bone Miner. Res. 22, 1700–1710 (2007).

    Article  CAS  Google Scholar 

  17. Dahl, R. et al. Discovery and validation of a series of aryl sulfonamides as selective inhibitors of tissue-nonspecific alkaline phosphatase (TNAP). J. Med. Chem. 52, 6919–6925 (2009).

    Article  CAS  Google Scholar 

  18. Fujifilm Life Sciences Fundamentals of Chemiluminescence Detection. Application note no. 6, Fujifilm Life Sciences, Tokyo, Japan, (1998) 〈http://www.fujifilm.de/media/science/6_fundamentals_in_chemiluminescence_detection.pdf〉.

Download references

Acknowledgements

This work was supported by NIH Roadmap Initiative Grant U54HG003916 (E.A.S.) and NIH Grant RC1 HL101899 (J.L.M.).

Author information

Authors and Affiliations

Authors

Contributions

J.L.M. identified and validated TNAP as a potential therapeutic target. E.A.S. designed and optimized the assay and performed the screening. E.A.S. wrote the paper with guidance and discussion from J.L.M.

Corresponding author

Correspondence to Eduard A Sergienko.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sergienko, E., Millán, J. High-throughput screening of tissue-nonspecific alkaline phosphatase for identification of effectors with diverse modes of action. Nat Protoc 5, 1431–1439 (2010). https://doi.org/10.1038/nprot.2010.86

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2010.86

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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