Skip to main content

Advertisement

Log in

Acute Enterocyte Adaptation to Luminal Glucose: A Posttranslational Mechanism for Rapid Apical Recruitment of the Transporter GLUT2

  • 2011 SSAT Plenary Presentation
  • Published:
Journal of Gastrointestinal Surgery Aims and scope

Abstract

Background

Glucose absorption postprandially increases markedly to levels far greater than possible by the classic glucose transporter sodium–glucose cotransporter 1 (SGLT1).

Hypothesis

Luminal concentrations of glucose >50 mM lead to rapid, phenotypic, non-genomic adaptations by the enterocyte to recruit another transporter, glucose transporter 2 (GLUT2), to the apical membrane to increase glucose absorption.

Methods

Isolated segments of jejunum were perfused in vivo with glucose-containing solutions in anesthetized rats. Carrier-mediated glucose uptake was measured in 10 and 100 mM glucose solutions (n = 6 rats each) with and without selective inhibitors of SGLT1 and GLUT2.

Results

The mean rate of carrier-mediated glucose uptake increased in rats perfused with 100 mM versus 10 mM glucose to 13.9 ± 2.9 μmol from 2.1 ± 0.1 μmol, respectively (p < 0.0001). Using selective inhibitors, the relative contribution of GLUT2 to glucose absorption was 56% in the 100 mM concentration of glucose compared to the 10 mM concentration (27%; p < 0.01). Passive absorption accounted for 6% of total glucose absorption at 100 mM glucose.

Conclusion

A small amount of GLUT2 is active at the lesser luminal concentrations of glucose, but when exposed to concentrations of 100 mM, the enterocyte presumably changes its phenotype by recruiting GLUT2 apically to markedly augment glucose absorption.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Alvarado F, Lherminier M, Phan H. Hamster intestinal disaccharide absorption: extracellular hydrolysis precedes transport of the monosaccharide products. J Physiol 1984;355:493–507

    PubMed  CAS  Google Scholar 

  2. Hediger MA, Coady MJ, Ikeda TS, Wright EM. Expression cloning and cDNA sequencing of the Na+/glucose cotransporter. Nature 1987;330:379–381

    PubMed  CAS  Google Scholar 

  3. Kellett GL, Brot-Laroche E, Mace OJ, Leturque A. Sugar absorption in the intestine: the role of GLUT2. Ann Rev Nutr 2008;28:35–54

    CAS  Google Scholar 

  4. Scow JS, Iqbal CW, Jones TW 3 rd, Qandeel HG, Zheng Y, Duenes JA, Nagao M, Madhavan S, Sarr MG. Absence of evidence of translocation of GLUT2 to the apical membrane of enterocytes in everted intestinal sleeves. J Surg Res 2011;167:56–61

    PubMed  CAS  Google Scholar 

  5. Crane RK. The gradient hypothesis and other models of carrier-mediated active transport. Rev Physiol Biochem Pharmacol 1977;78:99–159

    PubMed  CAS  Google Scholar 

  6. Schultz SG, Curran PF. Coupled transport of sodium and organic solutes. Physiol Rev 1970;50:637–718

    PubMed  CAS  Google Scholar 

  7. Semenza G, Kessler M, Hosang M, Weber J, Schmidt U. Biochemistry of the Na+, D-glucose cotransporter of the small intestinal membrane: state of the art in 1984. Biochim Biophys Acta 1984;779:343–379

    PubMed  CAS  Google Scholar 

  8. Stevens BR, Kaunitz JD, Wright EM. Intestinal transport of amino acids and sugars: advances using membrane vesicles. Ann Rev Physiol 1984;46:417–433

    CAS  Google Scholar 

  9. Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell J. Molecular Cell Biology, 4th edition. New York: W.H. Freeman and Co, 1999, pp 602–604

    Google Scholar 

  10. Kellett GL, Helliwell PA. The diffusive component of intestinal glucose absorption is mediated by the glucose-induced recruitment of GLUT2 to the brush-border membrane. Biochem J 2000;350:155–162

    PubMed  CAS  Google Scholar 

  11. Fullerton PM, Parsons DS. The absorption of sugars and water from rat intestine in vivo. Q J Exp Physiol 1956;41:387–397

    CAS  Google Scholar 

  12. Debnam ES, Levin RJ. An experimental method of identifying and quantifying the active transfer electrogenic component from the diffusive component during sugar absorption measured in vivo. J Physiol 1975;246:181–196

    PubMed  CAS  Google Scholar 

  13. Lostao MP, Berjon A, Barber A, Ponz F. On the multiplicity of glucose analogues transport systems in rat intestine. Rev Espan Fisiol 1991;47:209–216

    PubMed  CAS  Google Scholar 

  14. Pappenheimer JR, Reiss KZ. Contribution of solvent drag through intercellular junctions to absorption of nutrients by the small intestine of the rat. J Membr Biol 1987;100:123–136

    PubMed  CAS  Google Scholar 

  15. Helliwell POA, Richardson M, Affleck J, Kellett GL. Stimulation of fructose transport across the intestinal brush-border membrane by PMA is mediated by GLUT2 and dynamically regulated by protein kinase C. Biochem J 2000;350 pt.1:149–154

    Google Scholar 

  16. Kellett GL. The facilitated component of intestinal glucose absorption. J Physiol. 2001;531:585–595

    PubMed  CAS  Google Scholar 

  17. Affleck JA, Helliwell PA, Kellett GL. Immunocytochemical detection of GLUT2 at the rat intestinal brush-border membrane. J Histochem Cytochem 2003;51:1567–1574

    PubMed  CAS  Google Scholar 

  18. Zheng Y, Scow JS, Duenes JA, Sarr MG. Mechanisms of glucose uptake in intestinal cell lines: role of GLUT2. Surgery 2011; in press

  19. Houghton SG, Zarroug AE, Duenes JA, Fernandez-Zapico ME, Sarr MG. The diurnal periodicity of hexose transporter mRNA and protein levels in the rat jejunum: role of vagal innervation. Surgery 2006;139;542–549

    PubMed  Google Scholar 

  20. Helliwell PA, Kellett GL. The active and passive components of glucose absorption in rat jejunum under low and high perfusion stress. J Physiol 2002;544:579–589

    PubMed  CAS  Google Scholar 

  21. Lane JS, Whang EE, Rigberg DA, Hines OJ, Kwan D, Zinner MJ, McFadden DW, Diamond J, Ashley SW. Paracellular glucose transport plays a minor role in the unanesthetized dog. Am J Physiol 1999;276:789–794

    Google Scholar 

  22. Ferraris RS, Yarshapour S, Lloyd KC, Mirzayan R, Diamond JM. Luminal glucose concentrations in the gut under normal conditions. Am J Physiol 1990;259:822–837

    Google Scholar 

  23. Pappenheimer JR. On the coupling of membrane digestion with intestinal absorption of sugars and amino acids. Am J Physiol 1993;265:409–417

    Google Scholar 

  24. Rand EB, DePaoli AM, Davidson NO, Bell GI, Burant CF. Sequence, tissue distribution, and functional characterization of the rat fructose transporter GLUT5. Am J Physiol 1993;27:G1169-G1176

    Google Scholar 

  25. Stumpel F, Burcelin R, Jungermann K, Thorens B. Normal kinetics of intestinal glucose absorption in the absence of GLUT2: evidence for a transport pathway requiring glucose phosphorylation and transfer into the endoplasmic reticulum. Proc National Acad Sci 2001;98:11330–11335.

    CAS  Google Scholar 

  26. Scow JS, Tavakkolizadeh A, Zheng Y, Sarr MG. Acute adaptation by the small intestinal enterocyte: A posttranscriptional mechanism involving apical translocation of nutrient transporters. Surgery 2011;149:601–605

    PubMed  Google Scholar 

  27. Morgan EL, Mace OJ, Affleck J, Kellet GL. Apical GLUT2 and Cav 1.3: regulation of rat intestinal glucose and calcium absorption. J Physiol 2007;580.2:593–604

    Google Scholar 

  28. Helliwell PA, Rumsby MG, Kellett GL. Intestinal sugar absorption is regulated by phosphorylation and turnover of protein kinase C beta II mediated by phosphatidylinositol 3-kinase-and mammalian target of rapamycin-dependent pathways. J Biol Chem 2003;278:28644–28650

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This work was supported in part by a grant from the National Institutes of Health-DK39337 (Dr. Sarr).

The authors wish to thank Deborah Frank for her expertise in preparing the manuscript for submission.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael G. Sarr.

Additional information

Discussant

Dr. Stanley W. Ashley (Boston, MA): This was a terrific presentation of a well-designed and nicely conducted study examining what remains a relatively controversial hypothesis, i.e., that the GLUT2 transporter, traditionally believed to be primarily a basolateral membrane transporter, plays a role in apical glucose transport as well. Twenty years ago, this debate focused on the question of whether the apical sodium–glucose contransporter SGLT1 could explain all such transports or whether there was also some component of transport through the intercellular junctions with water, at least at high glucose concentrations. The hypothesis that GLUT2 also contributes to this is a relatively new one, and the authors have added significantly to the evidence supporting this and strengthened the argument that paracellular transport plays a minimal role. I have a couple of questions: (1) Phlorizin and phloretin seem equally effective in inhibiting transport at high glucose concentrations—does this mean that the inhibitors are less specific than we would hope or is there some interaction between the transporters? (2) Some investigators in this area have suggested that differences in species may explain some of the discrepancies between studies—do you think this is a rodent-only phenomenon or more generalizable? (3) Why have such a complex mechanism of acute adaptation? Would not it just be more simple to have the transporters there at all times rather than to insert them into the membrane only when glucose concentrations are high? Do the authors think this adaptation has any clinical significance?

Closing Discussant

Dr. Rizwan Chaudry: Thank you for the comments and excellent questions. In regard to your first question, phlorizin and phloretin at the concentrations used have been shown to be quite specific in their actions on transporters SGLT1 and GLUT2, respectively, as has been validated in several previous studies (Sarr et al. and Kellet et al.). Furthermore, in our current study, phlorizin and phloretin display significantly different effects on absorption at the 10 mM concentration of glucose, with phlorizin causing a much greater relative decrease in absorption as expected. The reason we see a similar effect on absorption (i.e., a significant decrease) with both inhibitors at the greater glucose concentration suggests that SGLT1 is necessary in GLUT2 translocation and may act as the rate-limiting step in the process; once translocated, however, GLUT2 accounted for the majority of the absorption (56%) at the greater concentration.

Although recent investigators have been studying intestinal glucose transport in vivo using a rat model, this phenomenon is not limited to this particular species. For example, a similar in vivo study was conducted in unanesthetized mongrel dogs in 1999 by Ashley et al. that showed paracellular transport to be minimal (2–5%) in its contribution to glucose absorption at supraphysiologic concentrations of glucose. Our experimental results for passive absorption at greater concentrations of glucose, although conducted in rats, paralleled what occurred in live dogs. Neither we nor to the best of our knowledge others have looked for this phenomenon in mice or other species.

Your third question, sir, is a bit more difficult to answer since we ourselves are trying to figure out the intricacies of this process. It may be that GLUT2 resides in the cytoplasm of the enterocyte and inserts to the apical membrane, or even the basolateral membrane, through an on-demand basis. As SGLT1, a high-affinity, low-capacity transporter becomes saturated, GLUT2, a low-affinity, high-capacity transporter, is rapidly available through activation of intracellular pathways involving a protein kinase C isoenzyme. The reason why GLUT2 just does not simply reside only at the apical membrane, and thus be readily available to transport glucose when luminal concentrations exceed 30 mM, could be because it may be needed in different amounts at the basolateral membrane as well, based on the amount of glucose needed to be absorbed systemically compared to the amount of glucose needed by the enterocyte itself for energy metabolism. Metabolic derangements such as hypo- or hyperglycemia may demand different amounts of GLUT2 to be readily available at certain locations of the cell Thus, a “pool” of GLUT2 deposits residing within the cytosol, being available on-demand for apical or basolateral glucose transport, makes sense.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chaudhry, R.M., Scow, J.S., Madhavan, S. et al. Acute Enterocyte Adaptation to Luminal Glucose: A Posttranslational Mechanism for Rapid Apical Recruitment of the Transporter GLUT2. J Gastrointest Surg 16, 312–319 (2012). https://doi.org/10.1007/s11605-011-1752-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11605-011-1752-y

Keywords

Navigation