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:

A natural mutation in Pisum sativum L. (pea) alters starch assembly and improves glucose homeostasis in humans

Abstract

Elevated postprandial glucose (PPG) is a significant risk factor for non-communicable diseases globally. Currently, there is a limited understanding of how starch structures within a carbohydrate-rich food matrix interact with the gut luminal environment to control PPG. Here, we use pea seeds (Pisum sativum) and pea flour, derived from two near-identical pea genotypes (BC1/19RR and BC1/19rr) differing primarily in the type of starch accumulated, to explore the contribution of starch structure, food matrix and intestinal environment to PPG. Using stable isotope 13C-labelled pea seeds, coupled with synchronous gastric, duodenal and plasma sampling in vivo, we demonstrate that maintenance of cell structure and changes in starch morphology are closely related to lower glucose availability in the small intestine, resulting in acutely lower PPG and promotion of changes in the gut bacterial composition associated with long-term metabolic health improvements.

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: Starch biosynthetic pathway in pea seeds.
Fig. 2: Effects of acute consumption of 50 g dry weight RR and rr pea seeds and flour.
Fig. 3: Impact of genotype structure and processing on starch digestibility.
Fig. 4: The effect of structure and genotype of pea seeds and flour on small intestinal environment.
Fig. 5: Using stable isotope 13C-enriched RR and rr pea seeds and flour to understand the digestion and fermentation process further.
Fig. 6: The effect of consuming products derived from the two pea genotypes for 28 days on glucose homeostasis and gut microbiota.

Similar content being viewed by others

Data availability

All presented data are tabulated and detailed in the main text and the Supplementary Information. The experimental procedures are detailed in the Methods. Quantified data are freely available from the Mendeley Data Database at https://doi.org/10.17632/gtthhhp9wz.1.

References

  1. O’Keefe, J. H. & Bell, D. S. Postprandial hyperglycemia/hyperlipidemia (postprandial dysmetabolism) is a cardiovascular risk factor. Am. J. Cardiol. 100, 899–904 (2007).

    PubMed  Google Scholar 

  2. Wolever, T. M. & Bolognesi, C. Source and amount of carbohydrate affect postprandial glucose and insulin in normal subjects. J. Nutr. 126, 2798–2806 (1996).

    CAS  PubMed  Google Scholar 

  3. Jenkins, D. et al. Glycemic index of foods: a physiological basis for carbohydrate exchange. Am. J. Clin. Nutr. 34, 362–366 (1981).

    CAS  PubMed  Google Scholar 

  4. Jenkins, D. J. et al. Glycemic index: overview of implications in health and disease. Am. J. Clin. Nutr. 76, 266S–273S (2002).

    CAS  PubMed  Google Scholar 

  5. Greenwood, D. C. et al. Glycemic index, glycemic load, carbohydrates, and type 2 diabetes: systematic review and dose–response meta-analysis of prospective studies. Diabetes Care 36, 4166–4171 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Jenkins, D. J. et al. Effect of a low–glycemic index or a high–cereal fiber diet on type 2 diabetes: a randomized trial. J. Am. Med. Assoc. 300, 2742–2753 (2008).

    CAS  Google Scholar 

  7. Wang, S. & Copeland, L. Molecular disassembly of starch granules during gelatinization and its effect on starch digestibility: a review. Food Funct. 4, 1564–1580 (2013).

    CAS  PubMed  Google Scholar 

  8. Sievenpiper, J. et al. Effect of non-oil-seed pulses on glycaemic control: a systematic review and meta-analysis of randomised controlled experimental trials in people with and without diabetes. Diabetologia 52, 1479 (2009).

    CAS  PubMed  Google Scholar 

  9. Rayner, T. et al. Genetic variation controlling wrinkled seed phenotypes in Pisum: how lucky was Mendel? Int. J. Mol. Sci. 18, 1205 (2017).

    PubMed Central  Google Scholar 

  10. Bhattacharyya, M. K. et al. The wrinkled-seed character of pea described by Mendel is caused by a transposon-like insertion in a gene encoding starch-branching enzyme. Cell 60, 115–122 (1990).

    CAS  PubMed  Google Scholar 

  11. Satoh, H. et al. Starch-branching enzyme I-deficient mutation specifically affects the structure and properties of starch in rice endosperm. Plant Physiol. 133, 1111–1121 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Sun, Y. et al. Generation of high-amylose rice through CRISPR/Cas9-mediated targeted mutagenesis of starch branching enzymes. Front. Plant Sci. 8, 298 (2017).

    PubMed  PubMed Central  Google Scholar 

  13. Hazard, B. et al. Induced mutations in the starch branching enzyme II (SBEII) genes increase amylose and resistant starch content in durum wheat. Crop Sci. 52, 1754–1766 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Schönhofen, A., Zhang, X. & Dubcovsky, J. Combined mutations in five wheat STARCH BRANCHING ENZYME II genes improve resistant starch but affect grain yield and bread-making quality. J. Cereal Sci. 75, 165–174 (2017).

    Google Scholar 

  15. Tuncel, A. et al. Cas9‐mediated mutagenesis of potato starch‐branching enzymes generates a range of tuber starch phenotypes. Plant Biotechnol. J. 17, 2259–2271 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Ghoos, Y. F. et al. Measurement of gastric emptying rate of solids by means of a carbon-labeled octanoic acid breath test. Gastroenterology 104, 1640–1647 (1993).

    CAS  PubMed  Google Scholar 

  17. McCleary, B. V. & Monaghan, D. A. Measurement of resistant starch. J. AOAC Int. 85, 665–675 (2002).

    CAS  PubMed  Google Scholar 

  18. Gidley, M. et al. Molecular order and structure in enzyme-resistant retrograded starch. Carbohydr. Polym. 28, 23–31 (1995).

    CAS  Google Scholar 

  19. Lopez-Rubio, A. et al. Molecular rearrangement of starch during in vitro digestion: toward a better understanding of enzyme resistant starch formation in processed starches. Biomacromolecules 9, 1951–1958 (2008).

    CAS  PubMed  Google Scholar 

  20. Shrestha, A. K. et al. Enzyme resistance and structural organization in extruded high amylose maize starch. Carbohydr. Polym. 80, 699–710 (2010).

    CAS  Google Scholar 

  21. Skrabanja, V. et al. Influence of genotype and processing on the in vitro rate of starch hydrolysis and resistant starch formation in peas (Pisum sativum L.). J. Agric. Food Chem. 47, 2033–2039 (1999).

    CAS  PubMed  Google Scholar 

  22. Zhou, Y., Hoover, R. & Liu, Q. Relationship between α-amylase degradation and the structure and physicochemical properties of legume starches. Carbohydr. Polym. 57, 299–317 (2004).

    CAS  Google Scholar 

  23. Lambeth, S. M. et al. Composition, diversity and abundance of gut microbiome in prediabetes and type 2 diabetes. J. Diabetes 2, 1–7 (2015).

    Google Scholar 

  24. Edwards, C. H. et al. Manipulation of starch bioaccessibility in wheat endosperm to regulate starch digestion, postprandial glycemia, insulinemia, and gut hormone responses: a randomized controlled trial in healthy ileostomy participants, 2. Am. J. Clin. Nutr. 102, 791–800 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Tahir, R. et al. Study of the structure and properties of native and hydrothermally processed wild-type, lam and r variant pea starches that affect amylolysis of these starches. Biomacromolecules 12, 123–133 (2010).

    PubMed  Google Scholar 

  26. Edwards, C. H. et al. A comparison of the kinetics of in vitro starch digestion in smooth and wrinkled peas by porcine pancreatic alpha-amylase. Food Chem. 244, 386–393 (2018).

    CAS  PubMed  Google Scholar 

  27. Pilichiewicz, A. N. et al. Load-dependent effects of duodenal glucose on glycemia, gastrointestinal hormones, antropyloroduodenal motility, and energy intake in healthy men. Am. J. Physiol. Endocrinol. Metab. 293, E743–E753 (2007).

    CAS  PubMed  Google Scholar 

  28. Zhang, G., Ao, Z. & Hamaker, B. R. Slow digestion property of native cereal starches. Biomacromolecules 7, 3252–3258 (2006).

    CAS  PubMed  Google Scholar 

  29. Donohoe, D. R. et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 13, 517–526 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Panwar, H. et al. Probiotics as potential biotherapeutics in the management of type 2 diabetes–prospects and perspectives. Diabetes Metab. Res. Rev. 29, 103–112 (2013).

    CAS  PubMed  Google Scholar 

  31. Julious, S. A. Sample size of 12 per group rule of thumb for a pilot study. Pharm. Stat. 4, 287–291 (2005).

    Google Scholar 

  32. Te Morenga, L. et al. Effect of a relatively high-protein, high-fiber diet on body composition and metabolic risk factors in overweight women. Eur. J. Clin. Nutr. 64, 1323–1331 (2010).

    PubMed  Google Scholar 

  33. Preston, T. & McMillan, D. Rapid sample throughput for biomedical stable isotope tracer studies. Biomed. Environ. Mass Spectrom. 16, 229–235 (1988).

    CAS  PubMed  Google Scholar 

  34. Edwards, C. et al. Production of 13C labelled pea flour for use in human digestion and fermentation studies. Isot. Environ. Health Stud. 38, 139–147 (2002).

    MathSciNet  CAS  Google Scholar 

  35. Kreymann, B., Ghatei, M. A., Williams, G. & Bloom, S. R. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet 330, 1300–1304 (1987).

    Google Scholar 

  36. Garcia-Perez, I. et al. Identifying unknown metabolites using NMR-based metabolic profiling techniques. Nat. Protoc. 15, 2538–2567 (2020).

    CAS  PubMed  Google Scholar 

  37. Morrison, D. J., O'Hara, J. P., King, R. F. G. J. & Preston, T. Quantitation of plasma 13C-galactose and 13C-glucose during exercise by liquid chromatography/isotope ratio mass spectrometry. Rapid Commun. Mass Spectrom. 25, 2484–2488 (2011).

    ADS  CAS  PubMed  Google Scholar 

  38. Morrison, D. J. et al. A streamlined approach to the analysis of volatile fatty acids and its application to the measurement of whole‐body flux. Rapid Commun. Mass Spectrom. 18, 2593–2600 (2004).

    ADS  CAS  PubMed  Google Scholar 

  39. Minekus, M. et al. A standardised static in vitro digestion method suitable for food–an international consensus. Food Funct. 5, 1113–1124 (2014).

    CAS  PubMed  Google Scholar 

  40. Perez-Moral, N. et al. Ultra-high performance liquid chromatography-size exclusion chromatography (UPLC-SEC) as an efficient tool for the rapid and highly informative characterisation of biopolymers. Carbohydr. Polym. 196, 422–426 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Flanagan, B. M., Gidley, M. J. & Warren, F. J. Rapid quantification of starch molecular order through multivariate modelling of 13C CP/MAS NMR spectra. J. Chem. Commun. 51, 14856–14858 (2015).

    CAS  Google Scholar 

  42. Wang, T. L., Bogracheva, T. Y. & Hedley, C. L. Starch: as simple as A, B, C? J. Exp. Bot. 49, 481–502 (1998).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank E. Panteliou and L. Mendoza for their help during nasogastric and nasoduodenal tube insertion. We thank A. Sukkar and A. Cherta Murillo for their assistance in trial 4 and G. Franco Becker and C. Byrne for their help with the radioimmunoassays in trial 4. We thank E. McKay for technical assistance with the plasma [13C]glucose and urine 13C-labelled SCFA assay and S. Small for technical assistance with the breath 13C and urine 13C-labelled SCFA assay and growth of isotope-labelled pea seeds. We thank M. Parker for valuable discussions and interpretation of the microscopy images. We thank B. Hazard, QIB, for very helpful discussions of starch mutations in cereals. All clinical trials were conducted at the NIHR Imperial Clinical Research Facility; we thank all of the staff and volunteers who took part in the study. The Division of Integrative Systems Medicine and Digestive Disease at Imperial College London receives financial support from the NIHR Imperial Biomedical Research Centre based at Imperial College Healthcare NHS Trust and Imperial College London, in line with the Gut Health research theme. I.G.-P. is supported by a NIHR fellowship (NIHR-CDF-2017-10-032). C.D. gratefully acknowledges support from the Department for Environment, Food & Rural Affairs (CH0103 and CH0111, Pulse Crop Genetic Improvement Network; and LK09126) and from the Biotechnology and Biological Sciences Research Council (BBSRC; BB/L025531/1 and BBS/E/J/000PR9799). We also gratefully acknowledge the support of the BBSRC through the BBSRC Institute Strategic Programme Food Innovation and Health BB/R012512/1 and its constituent project(s) BBS/E/F/000PR10343 (Theme 1, Food Innovation) and BBS/E/F/000PR10345 (Theme 2, Digestion in the Upper GI Tract). Infrastructure support was provided by the NIHR Imperial Biochemical Research Centre and the NIHR Imperial Clinical Research Facility. G.S.F. is an NIHR Senior Investigator. This research was funded by the BBSRC (grant nos. BB/L025582/1, BB/L025418/1, BB/L025531/1 and BB/L025566/1). The views expressed in this publication are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health and Social Care.

Author information

Authors and Affiliations

Authors

Contributions

K.P. and L.J.S. should be considered as joint first authors. G.S.F. oversaw the design and implementation of the in vivo experiments. K.P. managed, assisted in the design and performed all the experimental studies in vivo, sample collection, processing and data analysis. E.S.C. designed and applied for ethics approval for the human studies. R.A. and M.K. assisted in experimental human studies 2 and 3, and M.K. also assisted in study 4. N.P. was responsible for the nasogastric and nasoduodenal tube insertion. Metabolomics analysis was performed by K.P. and I.G.-P. Metabolite identification was performed by I.G.-P. and J.I.S.-C. D.J.M. and T.P. oversaw the stable isotope analysis and data analysis, and T.P. also oversaw the labelled crop production. L.J.S. performed simulated digestions, starch analysis of pea seeds, particle size analysis of pea fragments, sample preparation for light microscopy and scanning electron microscopy, and light microscopy imaging. Preparation, sectioning and imaging of pea tissue sections were performed by R.S. and K.L.C.; K.L.C. performed scanning electron microscopy. N.P. carried out simulated digestions of flours and subsequent starch analysis; diffusion experiments using fluorescence microscopy were also performed by N.P. R.A. performed the compression experiments and M.N.C. oversaw the implementation of these experiments. T.K. and Y.Z.K. carried out solid-state NMR experiments. P.J.W., F.J.W. and C.H.E. oversaw the design and implementation of the digestions in vitro and microscopy studies. C.D. oversaw field trials of the variant pea lines and multiplication of their seeds, with quality testing for all experiments. K.P., J.A.K.M., R.C.S. and J.M.B. performed 16S rRNA gene sequencing and data analysis. G.S.F. and K.P. led the initial drafts of the manuscript. All authors contributed to the final draft of the manuscript.

Corresponding author

Correspondence to Gary S. Frost.

Ethics declarations

Competing interests

The 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

Supplementary Information

Supplementary Figs. 1–9, Tables 1–14, methods and note.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Petropoulou, K., Salt, L.J., Edwards, C.H. et al. A natural mutation in Pisum sativum L. (pea) alters starch assembly and improves glucose homeostasis in humans. Nat Food 1, 693–704 (2020). https://doi.org/10.1038/s43016-020-00159-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43016-020-00159-8

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