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:

Slc12a8 is a nicotinamide mononucleotide transporter

Matters Arising to this article was published on 12 July 2019

An Author Correction to this article was published on 12 July 2019

This article has been updated

Abstract

Nicotinamide mononucleotide (NMN) is a biosynthetic precursor of nicotinamide adenine dinucleotide (NAD+) known to promote cellular NAD+ production and counteract age-associated pathologies associated with a decline in tissue NAD+ levels. How NMN is taken up into cells has not been entirely clear. Here we show that the Slc12a8 gene encodes a specific NMN transporter. We find that Slc12a8 is highly expressed and regulated by NAD+ in the mouse small intestine. Slc12a8 knockdown abrogates the uptake of NMN in vitro and in vivo. We further show that Slc12a8 specifically transports NMN, but not nicotinamide riboside, and that NMN transport depends on the presence of sodium ion. Slc12a8 deficiency significantly decreases NAD+ levels in the jejunum and ileum, which is associated with reduced NMN uptake as traced by doubly labelled isotopic NMN. Finally, we observe that Slc12a8 expression is upregulated in the aged mouse ileum, which contributes to the maintenance of ileal NAD+ levels. Our work identifies a specific NMN transporter and demonstrates that Slc12a8 has a critical role in regulating intestinal NAD+ metabolism.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Identification and characterization of the Slc12a8 gene.
Fig. 2: The kinetic features of the Slc12a8 NMN transporter and its specificity, sodium dependency and effects on NAD+ biosynthesis.
Fig. 3: In vivo knockdown of Slc12a8 in the small intestine.
Fig. 4: The age-associated upregulation of Slc12a8 in the ileum and its effect on NMN uptake and ileal NAD+ biosynthesis in aged mice.

Similar content being viewed by others

Data availability

The microarray data used in this study has been deposited into the National Center for Biotechnology Information Gene Expression Omnibus (GEO) database (GEO accession numbers GSE49784 and GSE118365). All data generated or analysed during this study are included in the article and its Supplementary Information.

Change history

  • 12 July 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Canto, C., Menzies, K. J. & Auwerx, J. NAD+ metabolism and the control of energy homeostasis: a balancing act between mitochondria and the nucleus. Cell Metab. 22, 31–53 (2015).

    Article  CAS  Google Scholar 

  2. Rajman, L., Chwalek, K. & Sinclair, D. A. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 27, 529–547 (2018).

    Article  CAS  Google Scholar 

  3. Verdin, E. NAD+ in aging, metabolism, and neurodegeneration. Science 350, 1208–1213 (2015).

    Article  CAS  Google Scholar 

  4. Yoshino, J., Baur, J. A. & Imai, S. I. NAD+ intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 27, 513–528 (2018).

    Article  CAS  Google Scholar 

  5. Yoshino, J., Mills, K. F., Yoon, M. J. & Imai, S. Nicotinamide mononucleotide, a key NAD+ intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 14, 528–536 (2011).

    Article  CAS  Google Scholar 

  6. Mouchiroud, L. et al. The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell 154, 430–441 (2013).

    Article  CAS  Google Scholar 

  7. Camacho-Pereira, J. et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 23, 1127–1139 (2016).

    Article  CAS  Google Scholar 

  8. Garten, A. et al. Physiological and pathophysiological roles of NAMPT and NAD metabolism. Nat. Rev. Endocrinol. 11, 535–546 (2015).

    Article  CAS  Google Scholar 

  9. Imai, S. Nicotinamide phosphoribosyltransferase (Nampt): a link between NAD biology, metabolism, and diseases. Curr. Pharm. Des. 15, 20–28 (2009).

    Article  CAS  Google Scholar 

  10. Belenky, P. et al. Nicotinamide riboside promotes Sir2 silencing and extends lifespan via Nrk and Urh1/Pnp1/Meu1 pathways to NAD+. Cell 129, 473–484 (2007).

    Article  CAS  Google Scholar 

  11. Caton, P. W., Kieswich, J., Yaqoob, M. M., Holness, M. J. & Sugden, M. C. Nicotinamide mononucleotide protects against pro-inflammatory cytokine-mediated impairment of mouse islet function. Diabetologia 54, 3083–3092 (2011).

    Article  CAS  Google Scholar 

  12. de Picciotto, N. E. et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell 15, 522–530 (2016).

    Article  Google Scholar 

  13. Gomes, A. P. et al. Declining NAD+ induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 155, 1624–1638 (2013).

    Article  CAS  Google Scholar 

  14. Long, A. N. et al. Effect of nicotinamide mononucleotide on brain mitochondrial respiratory deficits in an Alzheimer’s disease-relevant murine model. BMC Neurology 15, 19 (2015).

    Article  Google Scholar 

  15. Mills, K. F. et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 24, 795–806 (2016).

    Article  CAS  Google Scholar 

  16. Stein, L. R. & Imai, S. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J. 33, 1321–1340 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang, X., Hu, X., Yang, Y., Takata, T. & Sakurai, T. Nicotinamide mononucleotide protects against β-amyloid oligomer-induced cognitive impairment and neuronal death. Brain Res. 1643, 1–9 (2016).

    Article  CAS  Google Scholar 

  18. Yamamoto, T. et al. Nicotinamide mononucleotide, an intermediate of NAD+ synthesis, protects the heart from ischemia and reperfusion. PLoS ONE 9, e98972 (2014).

    Article  Google Scholar 

  19. Ramsey, K. M., Mills, K. F., Satoh, A. & Imai, S. Age-associated loss of Sirt1-mediated enhancement of glucose-stimulated insulin secretion in β cell-specific Sirt1-overexpressing (BESTO) mice. Aging Cell 7, 78–88 (2008).

    Article  CAS  Google Scholar 

  20. Revollo, J. R. et al. Nampt/PBEF/visfatin regulates insulin secretion in β cells as a systemic NAD biosynthetic enzyme. Cell Metab. 6, 363–375 (2007).

    Article  CAS  Google Scholar 

  21. Ratajczak, J. et al. NRK1 controls nicotinamide mononucleotide and nicotinamide riboside metabolism in mammalian cells. Nat. Commun. 7, 13103 (2016).

    Article  CAS  Google Scholar 

  22. Airhart, S. E. et al. An open-label, non-randomized study of the pharmacokinetics of the nutritional supplement nicotinamide riboside (NR) and its effects on blood NAD+ levels in healthy volunteers. PLoS ONE 12, e0186459 (2017).

    Article  Google Scholar 

  23. Liu, L. et al. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab. 27, 1067–1080 (2018).

    Article  CAS  Google Scholar 

  24. Hebert, S. C., Mount, D. B. & Gamba, G. Molecular physiology of cation-coupled Cl cotransport: the SLC12 family. Pflug. Arch. 447, 580–593 (2004).

    Article  CAS  Google Scholar 

  25. Ramsey, K. M. et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science 324, 651–654 (2009).

    Article  CAS  Google Scholar 

  26. Yamada, K., Hara, N., Shibata, T., Osago, H. & Tsuchiya, M. The simultaneous measurement of nicotinamide adenine dinucleotide and related compounds by liquid chromatography/electrospray ionization tandem mass spectrometry. Anal. Biochem. 352, 282–285 (2006).

    Article  CAS  Google Scholar 

  27. Shi, W. et al. Effects of a wide range of dietary nicotinamide riboside (NR) concentrations on metabolic flexibility and white adipose tissue (WAT) of mice fed a mildly obesogenic diet. Mol. Nutr. Food Res. 61, 1600878 (2017).

    Article  Google Scholar 

  28. Sociali, G. et al. Antitumor effect of combined NAMPT and CD73 inhibition in an ovarian cancer model. Oncotarget 7, 2968–2984 (2016).

    Article  Google Scholar 

  29. Alessi, D. R. et al. The WNK-SPAK/OSR1 pathway: master regulator of cation-chloride cotransporters. Sci. Signal. 7, re3 (2014).

    Article  Google Scholar 

  30. Belenky, P., Bogan, K. L. & Brenner, C. NAD+ metabolism in health and disease. Trends Biochem. Sci. 32, 12–19 (2007).

    Article  CAS  Google Scholar 

  31. Bieganowski, P. & Brenner, C. Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss-Handler independent route to NAD+ in fungi and humans. Cell 117, 495–502 (2004).

    Article  CAS  Google Scholar 

  32. Ummarino, S. et al. Simultaneous quantitation of nicotinamide riboside, nicotinamide mononucleotide and nicotinamide adenine dinucleotide in milk by a novel enzyme-coupled assay. Food Chem. 221, 161–168 (2017).

    Article  CAS  Google Scholar 

  33. Hewett, D. et al. Identification of a psoriasis susceptibility candidate gene by linkage disequilibrium mapping with a localized single nucleotide polymorphism map. Genomics 79, 305–314 (2002).

    Article  CAS  Google Scholar 

  34. Feigin, M. E. et al. Recurrent noncoding regulatory mutations in pancreatic ductal adenocarcinoma. Nat. Genet. 49, 825–833 (2017).

    Article  CAS  Google Scholar 

  35. Kim, J. E. et al. Associations between genetic polymorphisms of membrane transporter genes and prognosis after chemotherapy: meta-analysis and finding from Seoul Breast Cancer Study (SEBCS). Pharmacogenomics J. 18, 633–645 (2018).

    Article  CAS  Google Scholar 

  36. Wang, H. H., Patel, S. B., Carey, M. C. & Wang, D. Q. Quantifying anomalous intestinal sterol uptake, lymphatic transport, and biliary secretion in Abcg8 −/− mice. Hepatology 45, 998–1006 (2007).

    Article  CAS  Google Scholar 

  37. Yoshino, J. & Imai, S. Accurate measurement of nicotinamide adenine dinucleotide (NAD+) with high-performance liquid chromatography. Methods Mol. Biol. 1077, 203–215 (2013).

    Article  CAS  Google Scholar 

  38. Grimm, A. A., Brace, C. S., Wang, T., Stormo, G. D. & Imai, S. A nutrient-sensitive interaction between Sirt1 and HNF-1α regulates Crp expression. Aging Cell 10, 305–317 (2011).

    Article  CAS  Google Scholar 

  39. Revollo, J. R., Grimm, A. A. & Imai, S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J. Biol. Chem. 279, 50754–50763 (2004).

    Article  CAS  Google Scholar 

  40. Bruzzone, S. et al. The plant hormone abscisic acid increases in human plasma after hyperglycemia and stimulates glucose consumption by adipocytes and myoblasts. FASEB J. 26, 1251–1260 (2012).

    Article  CAS  Google Scholar 

  41. Bruzzone, S., Guida, L., Zocchi, E., Franco, L. & De Flora, A. Connexin 43 hemi channels mediate Ca2+-regulated transmembrane NAD+ fluxes in intact cells. FASEB J. 15, 10–12 (2001).

    Article  CAS  Google Scholar 

  42. Franco, L. et al. The transmembrane glycoprotein CD38 is a catalytically active transporter responsible for generation and influx of the second messenger cyclic ADP-ribose across membranes. FASEB J. 12, 1507–1520 (1998).

    Article  CAS  Google Scholar 

  43. Satoh, A. et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 18, 416–430 (2013).

    Article  CAS  Google Scholar 

  44. Sato, T. & Clevers, H. Primary mouse small intestinal epithelial cell cultures. Methods Mol. Biol. 945, 319–328 (2013).

    Article  Google Scholar 

  45. Kolodziejska-Huben, M., Kaminski, Z. & Paneth, P. Preparation of 18O-labelled nicotinamide. J. Label. Compd. Radiopharm. 45, 1005–1010 (2002).

    Article  CAS  Google Scholar 

  46. Chatterjee, A., Hazra, A. B., Abdelwahed, S., Hilmey, D. G. & Begley, T. P. A “radical dance” in thiamin biosynthesis: mechanistic analysis of the bacterial hydroxymethylpyrimidine phosphate synthase. Angew. Chem. Int. Ed. Engl. 49, 8653–8656 (2010).

    Article  CAS  Google Scholar 

  47. Fouquerel, E. et al. ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ depletion. Cell Rep. 8, 1819–1831 (2014).

    Article  CAS  Google Scholar 

  48. Cheadle, C., Vawter, M. P., Freed, W. J. & Becker, K. G. Analysis of microarray data using Z score transformation. J. Mol. Diagn. 5, 73–81 (2003).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. Cantoni for flow cytometric analysis, L. Guida for support in proteoliposome preparation and R. Lewis for the production of Slc12a8-KO mice. We also thank E. Schulak for his generous support to A.G., members of the S.I. lab for critical comments and suggestions on this study and staff members in the core facilities provided by the Diabetes Research Center (P30 DK020579), the Nutrition Obesity Research Center (P30 DK56341) and the Hope Center for Neurological Disorders at Washington University. This work was also performed in a facility supported by NCRR grant C06 RR015502. A.G. was supported as the Tanaka Scholar by T. Tanaka and M. Tanaka. M.E.M was supported by UK Research Councils and Biotechnology and Biological Science Research Council (BBSRC; BB/N001842/1). This work was mainly supported by grants from the National Institute on Aging (AG024150, AG037457, AG047902) to S.I.

Author information

Authors and Affiliations

Authors

Contributions

S.I. conceived the project, and A.G. and S.I. mainly designed research, analysed data and wrote the manuscript. A.G. performed most experiments. K.F.M. performed the analyses of gut-specific Slc12a8-KD mice and whole-body Slc12a8-KO mice with A.G. J.Y. performed microarray analyses. S.B. and G.S. conducted proteoliposome experiments and analysed the results with A.G. and S.I. K.T. and A.G. performed immunostaining, and H.C.L. and A.G. conducted Slc12a8 overexpression in vivo. R.C. and M.E.M. synthesized O18-D-NR and O18-D-NMN for this study, and Y.S. and A.G. conducted mass spectrometry analyses.

Corresponding author

Correspondence to Shin-ichiro Imai.

Ethics declarations

Competing interests

A.G. and S.I. are inventors on a patent (PCT/US18/46233) about the Slc12a8 NMN transporter, whose applicant is Washington University and which has been licensed by Teijin Limited (Japan). Other authors declare no competing interests.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grozio, A., Mills, K.F., Yoshino, J. et al. Slc12a8 is a nicotinamide mononucleotide transporter. Nat Metab 1, 47–57 (2019). https://doi.org/10.1038/s42255-018-0009-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-018-0009-4

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