Skip to main content
Log in

Upregulation of Intestinal NLRP6 Inflammasomes After Roux-en-Y Gastric Bypass Promotes Gut Immune Homeostasis

  • Original Contributions
  • Published:
Obesity Surgery Aims and scope Submit manuscript

Abstract

Objective

Roux-en-Y gastric bypass (RYGB) could affect immunological activity after surgery. We examined the role of RYGB on the NOD-like receptor pyrin domain containing-6 (NLRP6) in the intestine after surgery in rat models.

Methods

Expression of intestinal NLRP6 in the lean, obesity, RYGB, and sham-pair fed (PF) groups was analyzed by quantitative RT-PCR, Western blotting, and immunohistochemistry. Gut microbiota abundance was determined by 16S rRNA sequencing. Cohousing experiments were conducted to analyze the effects of gut microbiota. Inflammatory cell infiltration and gut permeability were further validated.

Results

Obese rats had decreased intestinal NLRP6 levels, which could be restored by RYGB but not by calorie restriction. This regulation was dependent on the gut microbiota-related metabolites, taurine, and histamine. After RYGB, there were increased levels of taurine, which could positively affect NLRP6 expression. The pair-fed groups showed increased histamine, which had the opposite effects on NLRP6. Obese rats had greater intestinal permeability along with increased CD8+ T cell infiltration. However, RYGB but not calorie restriction could restore these changes in a manner, dependent on gut NLRP6 expression.

Conclusions

In rat models, RYGB could efficiently restore abnormal gut permeability and reduce inflammation in the intestine, depending on reactivation of NLRP6.

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
Fig. 4

Similar content being viewed by others

References

  1. Koch L. Surgery: T2DM remission after RYGB-a ‘gutsy’ move. Nat Rev Endocrinol. 2013;9(10):564.

    Article  Google Scholar 

  2. Korybalska K, Luczak J, Swora-Cwynar E, et al. Weight loss-dependent and -independent effects of moderate calorie restriction on endothelial cell markers in obesity. J Physiol Pharmacol. 2017;68(4):597–608.

    CAS  PubMed  Google Scholar 

  3. Xia Z, Wang G, Li H, et al. Influence of bariatric surgery on the expression of nesfatin-1 in rats with type 2 diabetes mellitus. Curr Pharm Des. 2015;21(11):1464–71.

    Article  CAS  Google Scholar 

  4. Guo Y, Liu CQ, Shan CX, et al. Gut microbiota after Roux-en-Y gastric bypass and sleeve gastrectomy in a diabetic rat model: Increased diversity and associations of discriminant genera with metabolic changes. Diabetes Metab Res Rev. 2017;33(3)

    Article  Google Scholar 

  5. Cox AJ, West NP, Cripps AW. Obesity, inflammation, and the gut microbiota. Lancet Diabetes Endocrinol. 2015;3(3):207–15.

    Article  CAS  Google Scholar 

  6. Mraz M, Haluzik M. The role of adipose tissue immune cells in obesity and low-grade inflammation. J Endocrinol. 2014;222(3):R113–27.

    Article  CAS  Google Scholar 

  7. Boulangé CL, Neves AL, Chilloux J, et al. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016;8(1):42.

    Article  Google Scholar 

  8. Dalmas E, Venteclef N, Caer C, et al. T cell-derived IL-22 amplifies IL-1β-driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes. 2014;63(6):1966–77.

    Article  CAS  Google Scholar 

  9. Chen GY, Liu M, Wang F, et al. A functional role for Nlrp6 in intestinal inflammation and tumorigenesis. J Immunol. 2011;186(12):7187–94.

    Article  CAS  Google Scholar 

  10. Normand S, Delanoye-Crespin A, Bressenot A, et al. Nod-like receptor pyrin domain-containing protein 6 (NLRP6) controls epithelial self-renewal and colorectal carcinogenesis upon injury. Proc Natl Acad Sci U S A. 2011;108(23):9601–6.

    Article  CAS  Google Scholar 

  11. Bueter M, Abegg K, Seyfried F, et al. Roux-en-Y gastric bypass operation in rats. J Vis Exp. 2012;64:e3940. https://doi.org/10.3791/3940.

    Article  Google Scholar 

  12. Wang Q, Wang G, Hu C, et al. Optimization of a technique to standardize the rodent Roux-En-Y gastric bypass model and troubleshooting of postoperative failures. Obes Surg. 2019;29(5):1681–9.

    Article  Google Scholar 

  13. Hotz B, Arndt M, Dullat S, et al. Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin Cancer Res. 2007;13(16):4769–76.

    Article  CAS  Google Scholar 

  14. Rhee NA, Wahlgren CD, Pedersen J, et al. Effect of Roux-en-Y gastric bypass on the distribution and hormone expression of small-intestinal enteroendocrine cells in obese patients with type 2 diabetes. Diabetologia. 2015;58(10):2254–8.

    Article  CAS  Google Scholar 

  15. Saeidi N, Meoli L, Nestoridi E, et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science. 2013;341(6144):406–10.

    Article  CAS  Google Scholar 

  16. Mumphrey MB, Hao Z, Townsend RL, et al. Sleeve gastrectomy does not cause hypertrophy and reprogramming of intestinal glucose metabolism in rats. Obes Surg. 2015;25(8):1468–73.

    Article  Google Scholar 

  17. Netto BD, Bettini SC, Clemente AP, et al. Roux-en-Y gastric bypass decreases pro-inflammatory and thrombotic biomarkers in individuals with extreme obesity. Obes Surg. 2015;25(6):1010–8.

    Article  Google Scholar 

  18. Mocanu AO, Mulya A, Huang H, et al. Effect of Roux-en-Y gastric bypass on the NLRP3 Inflammasome in adipose tissue from obese rats. PLoS One. 2015;10(10):e0139764.

    Article  Google Scholar 

  19. Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17(2):179–88.

    Article  CAS  Google Scholar 

  20. Pan F, Zhang L, Li M, et al. Predominant gut Lactobacillus murinus strain mediates anti-inflammaging effects in calorie- restricted mice. Microbiome. 2018;6(1):54.

    Article  Google Scholar 

  21. Guo Y, Huang ZP, Liu CQ, et al. Modulation of the gut microbiome: a systematic review of the effect of bariatric surgery. Eur J Endocrinol. 2018;178(1):43–56.

    Article  CAS  Google Scholar 

  22. Elinav E, Strowig T, Kau AL, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745–57.

    Article  CAS  Google Scholar 

  23. Blanchard C, Moreau F, Chevalier J, et al. Sleeve gastrectomy alters intestinal permeability in diet-induced obese mice. Obes Surg. 2017;27(10):2590–8.

    Article  Google Scholar 

  24. Levy M, Thaiss CA, Zeevi D, et al. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 Inflammasome signaling. Cell. 2015;163(6):1428–43.

    Article  CAS  Google Scholar 

  25. de La Serre CB, Ellis CL, Lee J, et al. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am J Physiol Gastrointest Liver Physiol. 2010;299(2):G440–8.

    Article  Google Scholar 

  26. Anand PK, Malireddi RK, Lukens JR, et al. NLRP6 negatively regulates innate immunity and host defence against bacterial pathogens. Nature. 2012;488(7411):389–93.

    Article  CAS  Google Scholar 

  27. Mamantopoulos M, Ronchi F, Van Hauwermeiren F, et al. Nlrp6- and ASC- dependent inflammasomes do not shape the commensal gut microbiota composition. Immunity. 2017;47(2):339–348.e4.

    Article  CAS  Google Scholar 

  28. Wlodarska M, Thaiss CA, Nowarski R, et al. NLRP6 inflammasome orchestrates the colonic host-microbial interface by regulating goblet cell mucus secretion. Cell. 2014;156(5):1045–59.

    Article  CAS  Google Scholar 

  29. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542(7640):177–85.

    Article  CAS  Google Scholar 

  30. Lin L, Zhang J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017;18(1):2.

    Article  Google Scholar 

  31. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15(8):914–20.

    Article  CAS  Google Scholar 

  32. Sasaki T, Liu K, Agari T, et al. Anti-high mobility group box 1 antibody exerts neuroprotection in a rat model of Parkinson's disease. Exp Neurol. 2016;275(Pt 1):220–31.

    Article  CAS  Google Scholar 

  33. Malek N, Kostrzewa M, Makuch W, et al. The multiplicity of spinal AA-5-HT anti-nociceptive action in a rat model of neuropathic pain. Pharmacol Res. 2016;111:251–63.

    Article  CAS  Google Scholar 

Download references

Funding

This study was supported by National Key Basic Research Program of China (No.2015CB5540007); National Natural Science Foundation of China (No.81472740, 81200276 and 81700488); Natural Science Foundation of Hubei Province of China (No.2014CFA060 and 2015CFB710); Research Fund of Public Welfare in Health Industry, Health and Family Plan Committee of China (No.201402015); Natural Science Foundation of Huazhong University of Science and Technology (No.5001530030), and Health and Family Planning Youth Project Foundation of Hubei Province, China (No.WJ2015Q001).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Zefeng Xia.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

A Statement of Animal Rights/Ethical Approval

All procedures in this study were approved by the Ethics Committee for Animal Research of Tongji Medicine College.

Additional information

Publisher’s Note

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

Geng Wang and Qingbo Wang are co-first authors.

Electronic Supplementary Material

Supplementary Figure1

(SFig 1). A. Changes of body weight after surgery. B-C. Expression of NLRP6 by explants cultured with increasing doses of histamine or taurine. D. Infiltration of macrophages and CD4+T cells in the intestine in ND, HFD, RYGB and Sham-PF groups. E. Intestinal Muc gene expressions in different groups. ND: normal diet group. HFD: high fat diet group. RYGB: HFD rats after RYGB surgery. Sham-PF: HFD rats after sham surgery and received pair-fed control (PNG 1663 kb)

High resolution image (TIF 20855 kb)

Supplementary Table 1

The primer sequences of the target. (DOCX 13 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, G., Wang, Q., Bai, J. et al. Upregulation of Intestinal NLRP6 Inflammasomes After Roux-en-Y Gastric Bypass Promotes Gut Immune Homeostasis. OBES SURG 30, 327–335 (2020). https://doi.org/10.1007/s11695-019-04152-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11695-019-04152-4

Keywords

Navigation