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:

5-IP7 is a GPCR messenger mediating neural control of synaptotagmin-dependent insulin exocytosis and glucose homeostasis

Abstract

5-diphosphoinositol pentakisphosphate (5-IP7) is a signalling metabolite linked to various cellular processes. How extracellular stimuli elicit 5-IP7 signalling remains unclear. Here we show that 5-IP7 in β cells mediates parasympathetic stimulation of synaptotagmin-7 (Syt7)-dependent insulin release. Mechanistically, vagal stimulation and activation of muscarinic acetylcholine receptors triggers Gαq–PLC–PKC−PKD-dependent signalling and activates IP6K1, the 5-IP7 synthase. Whereas both 5-IP7 and its precursor IP6 compete with PIP2 for binding to Syt7, Ca2+ selectively binds 5-IP7 with high affinity, freeing Syt7 to enable fusion of insulin-containing vesicles with the cell membrane. β-cell-specific IP6K1 deletion diminishes insulin secretion and glucose clearance elicited by muscarinic stimulation, whereas mice carrying a phosphorylation-mimicking, hyperactive IP6K1 mutant display augmented insulin release, congenital hyperinsulinaemia and obesity. These phenotypes are absent in mice lacking Syt7. Our study proposes a new conceptual framework for inositol pyrophosphate physiology in which 5-IP7 acts as a GPCR second messenger at the interface between peripheral nervous system and metabolic organs, transmitting Gq-coupled GPCR stimulation to unclamp Syt7-dependent, and perhaps other, exocytotic events.

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: The M3R−Gαq/11–PLC–PKC–PKD–IP6K1 signalling axis activates IP6K1 by S118/S121 phosphorylation.
Fig. 2: The phospho-mimic IP6K1DD mutant mice display insulin hypersecretion, whereas β-cell-specific IP6K1 deletion decreases insulin secretion.
Fig. 3: Neural regulation of insulin secretion requires the pancreatic M3R–PKC–IP6K1 phosphorylation axis.
Fig. 4: 5-IP7 does not affect insulin biogenesis or Ca2+ influx.
Fig. 5: 5-IP7 promotes Syt7-triggered insulin secretion.
Fig. 6: 5-IP7 clamps Syt7 in a Ca2+-releasable manner to regulate Syt7–PIP2 interaction.

Similar content being viewed by others

Data availability

The atomic coordinates of the C2B domain of Syt-7 with IP6 were deposited to the Protein Data Bank under the accession codes PDB: 6LCY. All other data generated or analysed during this study are included in this published article (and its supplementary information files). Raw data and reagent requests should be addressed to F. Rao (raof@sustech.edu.cn). Source data are provided with this paper.

References

  1. Rosenbaum, D. M., Rasmussen, S. G. & Kobilka, B. K. The structure and function of G-protein-coupled receptors. Nature 459, 356–363 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hatch, A. J. & York, J. D. SnapShot: inositol phosphates. Cell 143, 1030–1030 e1031 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Irvine, R. F. & Schell, M. J. Back in the water: the return of the inositol phosphates. Nat. Rev. Mol. Cell Biol. 2, 327–338 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Shears, S. B. Inositol pyrophosphates: why so many phosphates? Adv. Biol. Regul. 57, 203–216 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Wilson, M. S., Livermore, T. M. & Saiardi, A. Inositol pyrophosphates: between signalling and metabolism. Biochem. J. 452, 369–379 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Saiardi, A., Bhandari, R., Resnick, A. C., Snowman, A. M. & Snyder, S. H. Phosphorylation of proteins by inositol pyrophosphates. Science 306, 2101–2105 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Ganguli, S. et al. A high energy phosphate jump — from pyrophospho-inositol to pyrophospho-serine. Adv. Biol. Regul. 75, 100662 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Wu, M., Chong, L. S., Perlman, D. H., Resnick, A. C. & Fiedler, D. Inositol polyphosphates intersect with signaling and metabolic networks via two distinct mechanisms. Proc. Natl Acad. Sci. USA 113, E6757–E6765 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shears, S. B. Diphosphoinositol polyphosphates: metabolic messengers? Mol. Pharmacol. 76, 236–252 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, X. et al. Control of XPR1-dependent cellular phosphate efflux by InsP8 is an exemplar for functionally-exclusive inositol pyrophosphate signaling. Proc. Natl Acad. Sci. USA 117, 3568–3574 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sahu, S. et al. InsP7 is a small-molecule regulator of NUDT3-mediated mRNA decapping and processing-body dynamics. Proc. Natl Acad. Sci. USA 117, 19245–19253 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bittner, T. et al. Photolysis of caged inositol pyrophosphate InsP8 directly modulates intracellular Ca2+ oscillations and controls C2AB domain localization. J. Am. Chem. Soc. 142, 10606–10611 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Szijgyarto, Z., Garedew, A., Azevedo, C. & Saiardi, A. Influence of inositol pyrophosphates on cellular energy dynamics. Science 334, 802–805 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Fu, C. et al. Neuronal migration is mediated by inositol hexakisphosphate kinase 1 via alpha-actinin and focal adhesion kinase. Proc. Natl Acad. Sci. USA 114, 2036–2041 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rao, F. et al. Inositol pyrophosphates promote tumor growth and metastasis by antagonizing liver kinase B1. Proc. Natl Acad. Sci. USA 112, 1773–1778 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chakraborty, A. et al. Inositol pyrophosphates inhibit Akt signaling, thereby regulating insulin sensitivity and weight gain. Cell 143, 897–910 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Illies, C. et al. Requirement of inositol pyrophosphates for full exocytotic capacity in pancreatic beta cells. Science 318, 1299–1302 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Chakraborty, A., Kim, S. & Snyder, S. H. Inositol pyrophosphates as mammalian cell signals. Sci. Signal 4, re1 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Chakraborty, A. et al. Casein kinase-2 mediates cell survival through phosphorylation and degradation of inositol hexakisphosphate kinase-2. Proc. Natl Acad. Sci. USA 108, 2205–2209 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ghoshal, S., Tyagi, R., Zhu, Q. & Chakraborty, A. Inositol hexakisphosphate kinase-1 interacts with perilipin1 to modulate lipolysis. Int. J. Biochem. Cell Biol. 78, 149–155 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nishikawa, K., Toker, A., Johannes, F. J., Songyang, Z. & Cantley, L. C. Determination of the specific substrate sequence motifs of protein kinase C isozymes. J. Biol. Chem. 272, 952–960 (1997).

    Article  CAS  PubMed  Google Scholar 

  22. Losito, O., Szijgyarto, Z., Resnick, A. C. & Saiardi, A. Inositol pyrophosphates and their unique metabolic complexity: analysis by gel electrophoresis. PLoS ONE 4, e5580 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Wilson, M. S., Bulley, S. J., Pisani, F., Irvine, R. F. & Saiardi, A. A novel method for the purification of inositol phosphates from biological samples reveals that no phytate is present in human plasma or urine. Open Biol. 5, 150014 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Bah, A. & Forman-Kay, J. D. Modulation of intrinsically disordered protein function by post-translational modifications. J. Biol. Chem. 291, 6696–6705 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gogl, G., Kornev, A. P., Remenyi, A. & Taylor, S. S. Disordered protein kinase regions in regulation of kinase domain cores. Trends Biochem. Sci. 44, 300–311 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hubbard, K. B. & Hepler, J. R. Cell signalling diversity of the Gqα family of heterotrimeric G proteins. Cell Signal 18, 135–150 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Atwood, B. K., Lopez, J., Wager-Miller, J., Mackie, K. & Straiker, A. Expression of G protein-coupled receptors and related proteins in HEK293, AtT20, BV2, and N18 cell lines as revealed by microarray analysis. BMC Genomics 12, 14 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Arias-Del-Val, J. et al. Regulation of inositol 1,4,5-trisphosphate-induced Ca2+ release from the endoplasmic reticulum by AMP-activated kinase modulators. Cell Calcium 77, 68–76 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Padmanabhan, U., Dollins, D. E., Fridy, P. C., York, J. D. & Downes, C. P. Characterization of a selective inhibitor of inositol hexakisphosphate kinases: use in defining biological roles and metabolic relationships of inositol pyrophosphates. J. Biol. Chem. 284, 10571–10582 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Noguchi, G. M. & Huising, M. O. Integrating the inputs that shape pancreatic islet hormone release. Nat. Metab. 1, 1189–1201 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wicksteed, B. et al. Conditional gene targeting in mouse pancreatic ss-cells: analysis of ectopic Cre transgene expression in the brain. Diabetes 59, 3090–3098 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tamarina, N. A., Roe, M. W. & Philipson, L. Characterization of mice expressing Ins1 gene promoter driven CreERT recombinase for conditional gene deletion in pancreatic beta-cells. Islets 6, e27685 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Bhandari, R., Juluri, K. R., Resnick, A. C. & Snyder, S. H. Gene deletion of inositol hexakisphosphate kinase 1 reveals inositol pyrophosphate regulation of insulin secretion, growth, and spermiogenesis. Proc. Natl Acad. Sci. USA 105, 2349–2353 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rajasekaran, S. S. et al. Inositol hexakisphosphate kinase 1 is a metabolic sensor in pancreatic beta-cells. Cell Signal 46, 120–128 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ahren, B. Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat. Rev. Drug Disco. 8, 369–385 (2009).

    Article  CAS  Google Scholar 

  36. Persaud, S. J. Islet G-protein coupled receptors: therapeutic potential for diabetes. Curr. Opin. Pharmacol. 37, 24–28 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Rodriguez-Diaz, R. & Caicedo, A. Neural control of the endocrine pancreas. Best. Pr. Res Clin. Endocrinol. Metab. 28, 745–756 (2014).

    Article  CAS  Google Scholar 

  38. Nakajima, K. et al. Minireview: novel aspects of M3 muscarinic receptor signaling in pancreatic beta-cells. Mol. Endocrinol. 27, 1208–1216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gautam, D. et al. A critical role for beta cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo. Cell Metab. 3, 449–461 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Tache, Y., Maeda-Hagiwara, M., Goto, Y. & Garrick, T. Central nervous system action of TRH to stimulate gastric function and ulceration. Peptides 9, 9–13 (1988).

    Article  PubMed  Google Scholar 

  41. Mussa, B. M., Sartor, D. M., Rantzau, C. & Verberne, A. J. Effects of nitric oxide synthase blockade on dorsal vagal stimulation-induced pancreatic insulin secretion. Brain Res. 1394, 62–70 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. Rodriguez-Diaz, R. et al. Innervation patterns of autonomic axons in the human endocrine pancreas. Cell Metab. 14, 45–54 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rodriguez-Diaz, R. et al. Alpha cells secrete acetylcholine as a non-neuronal paracrine signal priming beta cell function in humans. Nat. Med. 17, 888–892 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Song, Q., Huang, M., Wang, B., Kang, X. & Wang, C. Bidirectional regulation of Ca2+ in exo–endocytosis coupling. Sci. China Life Sci. 61, 1583–1585 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Brunger, A. T., Choi, U. B., Lai, Y., Leitz, J. & Zhou, Q. Molecular mechanisms of fast neurotransmitter release. Annu. Rev. Biophys. 47, 469–497 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gustavsson, N. et al. Impaired insulin secretion and glucose intolerance in synaptotagmin-7 null mutant mice. Proc. Natl Acad. Sci. USA 105, 3992–3997 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wang, S., Li, Y. & Ma, C. Synaptotagmin-1 C2B domain interacts simultaneously with SNAREs and membranes to promote membrane fusion. eLife 5, e14211 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Fukuda, M., Aruga, J., Niinobe, M., Aimoto, S. & Mikoshiba, K. Inositol-1,3,4,5-tetrakisphosphate binding to C2B domain of IP4BP/synaptotagmin II. J. Biol. Chem. 269, 29206–29211 (1994).

    Article  CAS  PubMed  Google Scholar 

  49. Veiga, N. et al. Coordination, microprotonation equilibria and conformational changes of myo-inositol hexakisphosphate with pertinence to its biological function. Dalton Trans. 43, 16238–16251 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Hager, A. et al. Cellular cations control conformational switching of inositol pyrophosphate analogues. Chemistry 22, 12406–12414 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Voleti, R., Tomchick, D. R., Sudhof, T. C. & Rizo, J. Exceptionally tight membrane-binding may explain the key role of the synaptotagmin-7 C2A domain in asynchronous neurotransmitter release. Proc. Natl Acad. Sci. USA 114, E8518–E8527 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chakraborty, A. The inositol pyrophosphate pathway in health and diseases. Biol. Rev. Camb. Philos. Soc. 93, 1203–1227 (2018).

    Article  PubMed  Google Scholar 

  53. Lee, S., Kim, M. G., Ahn, H. & Kim, S. Inositol Pyrophosphates: Signaling Molecules with Pleiotropic Actions in Mammals. Molecules 25, 2208 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  54. Trusov, Y. & Botella, J. R. Plant G-proteins come of age: breaking the bond with animal models. Front Chem. 4, 24 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Laha, D. et al. Arabidopsis ITPK1 and ITPK2 have an evolutionarily conserved phytic acid kinase activity. ACS Chem. Biol. 14, 2127–2133 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Laha, D. et al. VIH2 regulates the synthesis of inositol pyrophosphate InsP8 and jasmonate-dependent defenses in arabidopsis. Plant Cell 27, 1082–1097 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wild, R. et al. Control of eukaryotic phosphate homeostasis by inositol polyphosphate sensor domains. Science 352, 986–990 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. Scherer, P. C. et al. Inositol hexakisphosphate (IP6) generated by IP5K mediates cullin–COP9 signalosome interactions and CRL function. Proc. Natl Acad. Sci. USA 113, 3503–3508 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Carafoli, E. & Krebs, J. Why calcium? How calcium became the best communicator. J. Biol. Chem. 291, 20849–20857 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Brunger, A. T., Leitz, J., Zhou, Q., Choi, U. B. & Lai, Y. Ca2+-triggered synaptic vesicle fusion initiated by release of inhibition. Trends Cell Biol. 28, 631–645 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Courtney, N. A., Bao, H., Briguglio, J. S. & Chapman, E. R. Synaptotagmin 1 clamps synaptic vesicle fusion in mammalian neurons independent of complexin. Nat. Commun. 10, 4076 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Shears, S. B. Assessing the omnipotence of inositol hexakisphosphate. Cell Signal 13, 151–158 (2001).

    Article  CAS  PubMed  Google Scholar 

  63. Sasakawa, N. et al. Dissociation of inositol polyphosphates from the C2B domain of synaptotagmin facilitates spontaneous release of catecholamines in adrenal chromaffin cells. A suggestive evidence of a fusion clamp by synaptotagmin. Neuropharmacology 60, 1364–1370 (2011).

    Article  CAS  PubMed  Google Scholar 

  64. Gustavsson, N. et al. Synaptotagmin-7 is a principal Ca2+ sensor for Ca2+-induced glucagon exocytosis in pancreas. J. Physiol. 587, 1169–1178 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Gustavsson, N. et al. Synaptotagmin-7 as a positive regulator of glucose-induced glucagon-like peptide-1 secretion in mice. Diabetologia 54, 1824–1830 (2011).

    Article  CAS  PubMed  Google Scholar 

  66. Chu, B. B. et al. Cholesterol transport through lysosome-peroxisome membrane contacts. Cell 161, 291–306 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Lee, T. S. et al. Inositol pyrophosphates inhibit synaptotagmin-dependent exocytosis. Proc. Natl Acad. Sci. USA 113, 8314–8319 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Park, S. J. et al. Inositol pyrophosphate metabolism regulates presynaptic vesicle cycling at central synapses. iScience 23, 101000 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Rao, F. et al. Inositol pyrophosphates mediate the DNA–PK/ATM–p53 cell death pathway by regulating CK2 phosphorylation of Tti1/Tel2. Mol. Cell 54, 119–132 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wilson, M. S. & Saiardi, A. Inositol phosphates purification using titanium dioxide beads. Bio. Protoc. 8, e2959 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lin, H. et al. Basis for metabolite-dependent Cullin–RING ligase deneddylation by the COP9 signalosome. Proc. Natl Acad. Sci. USA 117, 4117–4124 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Rao, F. et al. Inositol hexakisphosphate kinase-1 mediates assembly/disassembly of the CRL4-signalosome complex to regulate DNA repair and cell death. Proc. Natl Acad. Sci. USA 111, 16005–16010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lin, H. et al. IP6-assisted CSN–COP1 competition regulates a CRL4–ETV5 proteolytic checkpoint to safeguard glucose-induced insulin secretion against hyperinsulinemia. Nat. Commun. 12, 2461 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhu, K. et al. Kindlin-2 modulates MafA and beta-catenin expression to regulate beta-cell function and mass in mice. Nat. Commun. 11, 484 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Virtue, S. & Vidal-Puig, A. GTTs and ITTs in mice: simple tests, complex answers. Nat. Metab. 3, 883–886 (2021).

    Article  PubMed  Google Scholar 

  76. Suriben, R. et al. β-cell insulin secretion requires the ubiquitin ligase COP1. Cell 163, 1457–1467 (2015).

    Article  CAS  PubMed  Google Scholar 

  77. Lin, H. et al. IP6-assisted CSN–COP1 competition regulates a CRL4–ETV5 proteolytic checkpoint to safeguard glucose-induced insulin secretion. Nat. Commun. 12, 2461 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Stephens, P. J., Devlin, F. J., Chabalowski, C. F. & Frisch, M. J. Ab-Initio calculation of vibrational absorption and circular-dichroism spectra using density-functional force-fields. J. Phys. Chem. 98, 11623–11627 (1994).

    Article  CAS  Google Scholar 

  79. Grimme, S., Antony, J., Ehrlich, S. & Krieg, H. A consistent and accurate ab initio parametrization of density functional dispersion correction (DFT-D) for the 94 elements H–Pu. J. Chem. Phys. 132, 154104 (2010).

    Article  PubMed  CAS  Google Scholar 

  80. Goerigk, L. & Grimme, S. A thorough benchmark of density functional methods for general main group thermochemistry, kinetics, and noncovalent interactions. Phys. Chem. Chem. Phys. 13, 6670–6688 (2011).

    Article  CAS  PubMed  Google Scholar 

  81. Hay, P. J. & Wadt, W. R. Abinitio effective core potentials for molecular calculations — potentials for K to Au including the outermost core orbitals. J. Chem. Phys. 82, 299–310 (1985).

    Article  CAS  Google Scholar 

  82. Bochevarov, A. D. et al. Jaguar: a high-performance quantum chemistry software program with strengths in life and materials sciences. Int. J. Quantum Chem. 113, 2110–2142 (2013).

    Article  CAS  Google Scholar 

  83. Tannor, D. J. et al. Accurate first principles calculation of molecular charge-distributions and solvation energies from ab-initio quantum-mechanics and continuum dielectric theory. J. Am. Chem. Soc. 116, 11875–11882 (1994).

    Article  CAS  Google Scholar 

  84. Marten, B. et al. New model for calculation of solvation free energies: correction of self-consistent reaction field continuum dielectric theory for short-range hydrogen-bonding effects. J. Phys. Chem. 100, 11775–11788 (1996).

    Article  CAS  Google Scholar 

  85. Ferrer-Orta, C. et al. Structural characterization of the rabphilin–3A–SNAP25 interaction. Proc. Natl Acad. Sci. USA 114, E5343–E5351 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. H. Snyder and F. Yu for generously sharing reagents and tools for this research, the Southern University of Science and Technology (SUSTech) Core Research Facilities and Peking University Laboratory Animal Center of Shenzhen Graduate School for technical assistance, and the Shanghai Synchrotron Radiation Facility beamline BL17U1 and BL19U1 for X-ray beam time. This work was supported by grants from the National Natural Science Foundation of China (31872798 and 91853129 to F.R.; 31670734 and 91953110, to C. W.), the Shenzhen Science and Technology Program (KQTD20200820113040070 to F.R.) and the Shenzhen Municipal Government (KQJSCX20180322152418316, JCYJ20170412153517422 and JCYJ20170817104311912 to F. R.), the Department of Science and Technology of Guangdong Province (2018A030313207, to F. R.), the Ministry of Science and Technology of the People’s Republic of China (2019YFA0508402, to C. W.), USTC Research Funds of Double First-Class Initiative (YD9100002006), the Fundamental Research Funds for the Central Universities (WK9100000029 and WK9100000013) and the Chinese Academy of Sciences Pioneer Hundred Talents Program (C. W.).

Author information

Authors and Affiliations

Authors

Contributions

F. R., C. W., X. Z., J. Z., C. M., and C. H. W. designed research; X. Z., J. Z., Y. Z., Y. Y., Y. L., B. Z., Z.X., N. L., Xiuyan Yang, Xiaoli Yang, D. C., A. W., B. W., N. M., S. W., Z. Z., C. Y., D. Y., K.Z., B. L. and Z. K. performed research; F. R., C. W., C. H. W., C. M., Y. R. and W. Z. supervised research; N. J., Z. L., M. L., Q. W., Z. H., X. Q., G. X. and W. H. contributed new reagents and analytic tools; H. L., Y. D., Q. F., T.-N. Z. and F. R. analysed data; and F. R. wrote the draft of the paper with input from X. Z., N. L., J. Z. and C. W.

Corresponding authors

Correspondence to Chao Wang or Feng Rao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Guy Rutter and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary handling editor: Christoph Schmitt.

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

Extended data

Extended Data Fig. 1 PKC and PKD phosphorylates IP6K1 at S121 and S118, respectively.

(a) Phos-tag gel electrophoresis of immunoprecipitated IP6K1 reveals a slower migrating species that is absent under normal PAGE. (b-c) Mass-spectrum of immunoprecipitated IP6K1 reveals phosphorylation at S118/S121 (b) and S127 (c). (d) Sequence alignments of the three human IP6K isoforms. The poorly-conserved unstructured region is marked by red rectangles. (e) The disorder tendency of IP6K1 determined by IUPred (http://iupred.enzim.hu/). IDR: intrinsic disordered region. (f) Phosphorylation status of IP6K1 and its mutants examined with antibodies specifically recognizing phosphorylated PKC and PKD substrates. (g-h) In vitro phosphorylation of GST-IP6K1 purified from HEK293 cells by purified PKCβ (g) and PKD (h). GST-IP6K1 overexpressed in a 10-cm plate was pull-down, washed extensively and then incubated with commercial PKCβ and PKD.

Source data

Extended Data Fig. 2 Activation of IP6K1 by PKC/D-mediated phosphorylation and consequent conformational changes.

(a) The catalytic activity of IP6K1 with or without phosphorylation by PKC or PKD, assayed by the PAGE gel method, suggest that single kinase phosphorylation only modestly improves IP6K1 enzymatic activity. (b) The catalytic activities of the S118D/S121D (DD) phospho-mimic mutant is higher than wildtype IP6K1. (c) The catalytic activity of IP6K1 wildtype, S118D and S121D single mutants are comparable. (d) Limited tryptic proteolysis of unmodified or phosphorylated IP6K1 reveal slowed digest upon phosphorylation. (e) Circular dichroism analysis of WT (upper panel) or DD mutant (lower panel) IP6K1 with or without urea at the indicated concentrations. 3-4 M urea totally denatures WT but not DD mutant IP6K1. (f) Scheme depicting the proposed mechanism of IP6K1 activation by PKC phosphorylation.

Source data

Extended Data Fig. 3 A M3R-Gαq/11-PLC-nPKC-IP6K1 signaling axis revealed by a phospho-IP6K1 antibody specifically detecting p-S118/S121.

(a) The specificity of a rabbit monoclonal antibody (p-IP6K1) that specifically recognizes IP6K1 phosphorylated at S118/S121 is validated by using IP6K1 knockdown and knockout cells as well as male IP6K1 knockout mice. (b) Effect of in-lysate treatment with calf intestinal phosphatase (CIP, 1:100) on IP6K1-S118/S121 phosphorylation. An asterisk (*) indicates a nonspecific band. (c) The signal of the p-IP6K1 antibody is not affected by the neighboring S127A mutation. (d) Effect of in cell treatment with the PKC activator PMA (5 μM, 5 min) and the PKC inhibitor Go6983 (5 μM, 20 min) on IP6K1-S118/S121 phosphorylation. (e) Time-dependent analysis of IP6K1 S118/S121 phosphorylation upon carbachol (CCH) stimulation. (f) Effect of M3R antagonists Atropine (10 μM, 20 min) and J104129 (100 nM, 20 min) on carbachol stimulated IP6K1 phosphorylation. (g) Effect of overexpressing Gαq, Gα11, and their constitutively active Q209L mutants on carbachol stimulated IP6K1 phosphorylation. (h) Effect of Gαq and Gα11 knockdown on carbachol stimulated IP6K1 phosphorylation. An asterisk (*) indicates a nonspecific band. (i) Effect of histamine (50 μM) stimulation on IP6K1 S118/121 phosphorylation in HeLa cells. (j) Effect of Gαq/11 inhibition by YM-254890 (100 nM, 30 min), PLC inhibition by U73122 (10 μM, 30 min), PKC inhibition by Go6983 (5 μM, 30 min), and PKD inhibition CRT0066101 (2 μM, 30 min) on Histamine-stimulated (50 μM, 3 min) IP6K1 phosphorylation.

Source data

Extended Data Fig. 4 The phosphor-mimic IP6K1DD mutant mice display insulin hypersecretion, whereas β cell-specific IP6K1 deletion decrease insulin secretion.

(a) levels of 5-IP7 and IP6 levels in the various organs of male WT and IP6K1DD (DD) mouse littermates. (b) Levels of IP6K1 protein in the pancreas of male WT and DD mutant mice. (c) Lelves of 5-IP7 and IP6 in male WT and DD mouse embryonic fibroblasts (MEF) (*p = 0.02). (d) Weight of the various adipose tissues from male WT and DD mice at 4-months old (*p = 0.01, **p < 0.01). Right panel: Representative photographs of the white adipose tissues (WAT). EWAT: epididymal WAT; RWAT: retroperitoneal WAT; IWAT: inguinal WAT. (e-h) O2 consumption (e), CO2 production (f), respiratory exchange ratio (g), energy expenditure (h) in male WT and DD mutant mice, measured using CLAMS cages for 48 h (n = 6 mice per group). (i-j) Levels of serum glycerol (i) and non-esterified fatty acids (j) in male WT and DD mice before and after fasting (*p < 0.05, **p = 0.006). (k) Glucose-induced insulin secretion with glucose applied by oral gavage. (2 g/Kg body weight) tolerance test (GTT) (*p < 0.05, **p = 0.007). (l-n) Body weight (l), levels of feeding and fasting glucose (m), and levels of feeding and fasting insulin (n) in female WT vs DD mice (*p < 0.05, **p = 0.009, ***p = 0.0006). (o) Intraperitoneal glucose-induced insulin secretion in female WT and DD mice (*p < 0.05). (p) Bodyweight and serum insulin levels of male WT and DD mice prior to and six weeks after streptozocin treatment (STZ, 50 mg/kg, 4 days) (**p = 0.004). (q) Bodyweight and serum insulin levels of male WT and DD mice prior to and six weeks after streptozocin treatment (STZ, 50 mg/kg, 4 days) (*p = 0.02, **p = 0.007). (r) Insulin secretion from pancreatic islets isolated from male WT and mice (*p = 0.009). (s) Western-blot demonstrating successful depletion of IP6K1 protein in male IP6K1fl/fl:MIP-CreERT mice upon tamoxifen induction (*p = 9.5 x 10-11). (t-u) Levels of feeding and fasting glucose (t) and feeding and fasting insulin (u) in male IP6K1fl/fl and IP6K1fl/fl:MIP-CreERT mice after tamoxifen-induced IP6K1 deletion. n.s.: not significant. For Extended Data Fig. 4b-d and i-u, data represent means ± SEM; n represents number of mice (each data point = 1 mouse). Two-tailed t-tests were used for statistical analysis.

Source data

Extended Data Fig. 5 Glucose does not stimulate IP6K1 phosphorylation.

(a) Effect of glucose on IP6K1 phosphorylation in 293 and INS1 cells. An asterisk (*) indicates a nonspecific band. (b) Effect of glucose, applied via oral gavage or i.p. injection, on IP6K1 phosphorylation in pancreas. (c) Effect of IP6K1 knockdown on insulin secretion from INS1 cells at 2.8 mM and 16.8 mM glucose concentrations. For Extended Data Fig. 5c, data represent means ± SEM; n represents number of mice (each data point = 1 mouse). Two-tailed t-tests were used for statistical analysis.

Source data

Extended Data Fig. 6 Cholinergic, but not adrenergic, regulation of insulin secretion requires the pancreatic M3R-PKC-IP6K1 phosphorylation axis.

(a) Time-dependent stimulation of IP6K1 S118/S121 phosphorylation by the muscarinic agonist Oxotremorine M (Oxo-M, 2.5 μM) in isolated mouse islets. (b) Carbachol-induced IP6K1 phosphorylation is reversed by pretreatment with the Gαq inhibitor YM254890 (100 nM, 15 min). (c) Isoprenaline (i.p., 750 μg/kg, 10 min)-induced insulin secretion is similar between tamoxifen-treated male IP6K1fl/fl and IP6K1fl/fl:MIP-CreERT mice. (d) Intracisternal injection of thyrotropin releasing hormone (TRH) (0.5 μg in 100 nL) application elicits IP6K1 phosphorylation in male mouse pancreas. For Extended Data Fig. 6c, data represent means ± SEM; n = 3 independent biological experiments. Two-tailed t-tests were used for statistical analysis.

Source data

Extended Data Fig. 7 Muscarinic stimulation of human islets elicits IP6K1 phosphorylation and insulin secretion.

(a) Oxo-M treatment (2.5 μM, 10 min) of isolated human islets stimulates IP6K1 S118/121 phosphorylation, which can be blocked by Atropine pretreatment (10 μM, 15 min). (b) Effect of IP6K1 inhibition on Oxo-M-induced insulin secretion under low and high glucose conditions (***p = 0.0004, ****p = 1.7 x 10-6). (c) Effect of IP6K1 knockdown on Oxo-M-induced insulin secretion under low and high glucose conditions (**p = 0.001, ***p = 0.0005, ****p = 2 x 10-5). For Extended Data Fig. 7b-c, data represent means ± SEM; n = 4 independent biological experiments. Two-tailed t-tests were used for statistical analysis.

Source data

Extended Data Fig. 8 IP6K1 does not affect insulin biogenesis.

(a-b) H&E staining-based morphometric analysis of pancreatic islets from tamoxifen-treated male IP6K1fl/fl and IP6K1fl/fl:MIP-CreERT mice at 3 months old. Number of islets per pancreas (a) and average islet area (b) were measured by examining 20 slides with every five slides in between, covering the whole pancreas (n = 4 mice). (c) Total insulin content of pancreatic islet from male IP6K1fl/fl or IP6K1fl/fl:MIP-CreERT mice measured using ELISA assay. (*p = 0.03, n = 5 randomly selected islets). (d) Average cytoplasmic Ca2+ response to depolarization induced by 5 μM Oxo-M, assayed with Fura2-AM (5 μM, 30 mins) on islets from tamoxifen-treated male IP6K1fl/fl and IP6K1fl/fl:MIP-CreERT mice. Images were acquired at 2 seconds intervals. n = 4 (IP6K1fl/fl) or 3 independent (IP6K1fl/fl:MIP-CreERT) biological experiments from mouse islets. For Extended Data Fig. 8a-d, data represent means ± SEM; n represents number of mice (each data point = 1 mouse). Two-tailed t-tests were used for statistical analysis. n.s.: not significant.

Source data

Extended Data Fig. 9 5-IP7 does not affect calcium influx, but cooperates with Ca2+ to promote Syt7-dependent insulin secretion.

(a) Effect of IP6K1 knockdown on insulin secretion from INS1 cells at 5mM and 30 mM KCl concentrations (***p = 0.0006, ****p = 9 x 10-5). (b) Extracellular calcium (Ca2+o) removal greatly abolishes insulin secretion from male wildtype (WT) and IP6K1DD (DD) islets (**p = 0.001, *p = 0.048). (c) Deleting the juxtamembrane segment of Syt7 (aa 38-104) abolishes Flag-IP6K1 co-immunoprecipitation with HA-Syt7.

Source data

Extended Data Fig. 10 5-IP7 clamps Syt7 in a Ca2+-releasable manner to regulate Syt7-PIP2 interaction.

(a) FRET assay measuring the binding of NBD-labelled Syt7 to Rhodamine-PE labelled liposome, in the presence of 0.5 mM MgCl2 and increasing concentrations of IP6 (middle panel) or 5-IP7 (right panel). In this experiment, 1 µM IANBD-Syt7, 100 µM (total lipids) liposome, 25 µM CaCl2, 0.5 mM MgCl2, and indicated concentrations of IP6/5-IP7 were mixed and measured in 1-cm cuvette at 25 °C. Data points are presented as means ± s.d. (n = 3, technical replicates). (b) Concentration-dependent effect of 5-IP7 and IP6 on the interaction between Ca2+ (2 μM) and Fura2 dye (2 μM).

Supplementary information

Supplementary Information

Supplementary Figure 1, Supplementary Tables 1 and 2 and Supplementary Protocol for 5-IP7 synthesis.

Reporting Summary

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 1

Unprocessed western blots and gels.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 2

Unprocessed western blots and gels.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 3

Unprocessed western blots and gels.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 5

Unprocessed western blots and gels.

Source Data Extended Data Fig. 1

Unprocessed western blots and gels.

Source Data Extended Data Fig. 2

Unprocessed western blots and gels.

Source Data Extended Data Fig. 3

Unprocessed western blots and gels.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 4

Unprocessed western blots and gels.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 5

Unprocessed western blots and gels.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 6

Unprocessed western blots and gels.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 7

Unprocessed western blots and gels.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 9

Statistical Source Data.

Source Data Extended Data Fig. 9

Unprocessed western blots and gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, X., Li, N., Zhang, J. et al. 5-IP7 is a GPCR messenger mediating neural control of synaptotagmin-dependent insulin exocytosis and glucose homeostasis. Nat Metab 3, 1400–1414 (2021). https://doi.org/10.1038/s42255-021-00468-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00468-7

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