open access

Vol 74, No 3 (2023)
Original paper
Submitted: 2023-01-03
Accepted: 2023-03-16
Published online: 2023-05-15
Get Citation

Irisin attenuates pyroptosis in high glucose-induced pancreatic beta cells via the miR-133a-3p/FOXO1 axis

Anjun Tan1, Tianrong Li1, Jingjing Yang1, Jinwen Yu1, Hewen Chen1
·
Pubmed: 37335063
·
Endokrynol Pol 2023;74(3):277-284.
Affiliations
  1. Department of Geriatric Medicine, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China

open access

Vol 74, No 3 (2023)
Original Paper
Submitted: 2023-01-03
Accepted: 2023-03-16
Published online: 2023-05-15

Abstract

Introduction: Irisin is closely related to type 2 diabetes mellitus (T2DM) and other metabolic diseases. It can improve the homeostasis of T2DM. MiR-133a-3p is decreased in the peripheral blood of patients with T2DM. Forkhead box protein O1 (FOXO1) is widely expressed in beta-cells and affects the occurrence of diabetes through transcriptional regulation and signalling pathway regulation.

Material and methods: The miR-133a-3p inhibitor was constructed to verify the effect of irisin on pyroptosis through miR-133a-3p. Next, we predicted the presence of targeted binding sequences between FOXO1 and miR-133a-3p by bioinformatics software, which was then confirmed with a double fluorescence assay. Finally, the FOXO1 overexpression vector was used to further verify the effect of irisin through the miR-133a-3p/FOXO1 axis.

Results: We first observed that irisin inhibited the protein levels of N-terminal gasdermin D (GSDMD-N) and cleaved caspase-1 and the secretion of interleukins (IL): IL-1beta and IL-18 in Min6 cells treated with high glucoes (HG). Irisin inhibited pyroptosis of Min6 cells treated with HG by reinforcing miR-133a-3p. Then, FOXO1 was validated to be the target gene of miR-133a. Both miR-133a-3p inhibitor and overexpression of FOXO1 restrained the force of irisin on pyroptosis in HG-induced Min6 cells.

Conclusion: We explored the protective effect of irisin on HG-induced pyroptosis of islet b-cells in vitro and explained its mechanism of inhibiting pyroptosis through the miR-133a-3p/FOXO1 axis, to provide a theoretical basis for finding new molecular targets to delay beta-cell failure and the treatment of T2DM.

Abstract

Introduction: Irisin is closely related to type 2 diabetes mellitus (T2DM) and other metabolic diseases. It can improve the homeostasis of T2DM. MiR-133a-3p is decreased in the peripheral blood of patients with T2DM. Forkhead box protein O1 (FOXO1) is widely expressed in beta-cells and affects the occurrence of diabetes through transcriptional regulation and signalling pathway regulation.

Material and methods: The miR-133a-3p inhibitor was constructed to verify the effect of irisin on pyroptosis through miR-133a-3p. Next, we predicted the presence of targeted binding sequences between FOXO1 and miR-133a-3p by bioinformatics software, which was then confirmed with a double fluorescence assay. Finally, the FOXO1 overexpression vector was used to further verify the effect of irisin through the miR-133a-3p/FOXO1 axis.

Results: We first observed that irisin inhibited the protein levels of N-terminal gasdermin D (GSDMD-N) and cleaved caspase-1 and the secretion of interleukins (IL): IL-1beta and IL-18 in Min6 cells treated with high glucoes (HG). Irisin inhibited pyroptosis of Min6 cells treated with HG by reinforcing miR-133a-3p. Then, FOXO1 was validated to be the target gene of miR-133a. Both miR-133a-3p inhibitor and overexpression of FOXO1 restrained the force of irisin on pyroptosis in HG-induced Min6 cells.

Conclusion: We explored the protective effect of irisin on HG-induced pyroptosis of islet b-cells in vitro and explained its mechanism of inhibiting pyroptosis through the miR-133a-3p/FOXO1 axis, to provide a theoretical basis for finding new molecular targets to delay beta-cell failure and the treatment of T2DM.

Get Citation

Keywords

irisin; T2DM; FOXO1; miR-133a-3p; pyroptosis

About this article
Title

Irisin attenuates pyroptosis in high glucose-induced pancreatic beta cells via the miR-133a-3p/FOXO1 axis

Journal

Endokrynologia Polska

Issue

Vol 74, No 3 (2023)

Article type

Original paper

Pages

277-284

Published online

2023-05-15

Page views

1538

Article views/downloads

495

DOI

10.5603/EP.a2023.0035

Pubmed

37335063

Bibliographic record

Endokrynol Pol 2023;74(3):277-284.

Keywords

irisin
T2DM
FOXO1
miR-133a-3p
pyroptosis

Authors

Anjun Tan
Tianrong Li
Jingjing Yang
Jinwen Yu
Hewen Chen

References (32)
  1. Sun H, Saeedi P, Karuranga S, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022; 183: 109119.
  2. Petersmann A, Müller-Wieland D, Müller UA, et al. Definition, Classification and Diagnosis of Diabetes Mellitus. Exp Clin Endocrinol Diabetes. 2019; 127(S 01): S1–S7.
  3. Holmes D. Diabetes: Why β cells fail in T2DM. Nat Rev Endocrinol. 2017; 13(8): 440.
  4. Group UPDSU. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998; 352(9131): 837–853.
  5. Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem Sci. 2017; 42(4): 245–254.
  6. de Vasconcelos NM, Lamkanfi M. Recent Insights on Inflammasomes, Gasdermin Pores, and Pyroptosis. Cold Spring Harb Perspect Biol. 2020; 12(5).
  7. Rohm TV, Meier DT, Olefsky JM, et al. Inflammation in obesity, diabetes, and related disorders. Immunity. 2022; 55(1): 31–55.
  8. Maedler K, Sergeev P, Ris F, et al. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest. 2002; 110(6): 851–860.
  9. Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol. 2021; 22(5): 550–559.
  10. McClelland AD, Kantharidis P. microRNA in the development of diabetic complications. Clin Sci (Lond). 2014; 126(2): 95–110.
  11. Kokkinopoulou I, Maratou E, Mitrou P, et al. Decreased expression of microRNAs targeting type-2 diabetes susceptibility genes in peripheral blood of patients and predisposed individuals. Endocrine. 2019; 66(2): 226–239.
  12. Kitamura T. The role of FOXO1 in β-cell failure and type 2 diabetes mellitus. Nat Rev Endocrinol. 2013; 9(10): 615–623.
  13. Fatima K, Mathew S, Faheem M, et al. The Dual Specificity Role of Transcription Factor FOXO in Type 2-diabetes and Cancer. Curr Pharm Des. 2018; 24(24): 2839–2848.
  14. Nathanael J, Suardana P, Vianney YM, et al. The role of FoxO1 and its modulation with small molecules in the development of diabetes mellitus: A review. Chem Biol Drug Des. 2022; 99(2): 344–361.
  15. Zeng R, Luo DX, Li HP, et al. MicroRNA-135b alleviates MPP-mediated Parkinson's disease in in vitro model through suppressing FoxO1-induced NLRP3 inflammasome and pyroptosis. J Clin Neurosci. 2019; 65: 125–133.
  16. Heo MiJ, Kim TH, You JS, et al. Alcohol dysregulates miR-148a in hepatocytes through FoxO1, facilitating pyroptosis via TXNIP overexpression. Gut. 2019; 68(4): 708–720.
  17. Li Y, Liu X, Lin X, et al. Chemical compound cinobufotalin potently induces FOXO1-stimulated cisplatin sensitivity by antagonizing its binding partner MYH9. Signal Transduct Target Ther. 2019; 4: 48.
  18. Waseem R, Shamsi A, Mohammad T, et al. FNDC5/Irisin: Physiology and Pathophysiology. Molecules. 2022; 27(3).
  19. Liu J, Qi B, Gan L, et al. A Bibliometric Analysis of the Literature on Irisin from 2012-2021. Int J Environ Res Public Health. 2022; 19(10).
  20. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001; 25(4): 402–408.
  21. Gora IM, Ciechanowska A, Ladyzynski P. NLRP3 Inflammasome at the Interface of Inflammation, Endothelial Dysfunction, and Type 2 Diabetes. Cells. 2021; 10(2).
  22. He Y, Hara H, Núñez G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci. 2016; 41(12): 1012–1021.
  23. Chen C, Rong P, Yang M, et al. The Role of Interleukin-1β in Destruction of Transplanted Islets. Cell Transplant. 2020; 29: 963689720934413.
  24. Choi YK, Kim MK, Bae KH, et al. Serum irisin levels in new-onset type 2 diabetes. Diabetes Res Clin Pract. 2013; 100(1): 96–101.
  25. Song H, Wu F, Zhang Y, et al. Irisin promotes human umbilical vein endothelial cell proliferation through the ERK signaling pathway and partly suppresses high glucose-induced apoptosis. PLoS One. 2014; 9(10): e110273.
  26. Zhang Y, Mu Q, Zhou Z, et al. Protective Effect of Irisin on Atherosclerosis via Suppressing Oxidized Low Density Lipoprotein Induced Vascular Inflammation and Endothelial Dysfunction. PLoS One. 2016; 11(6): e0158038.
  27. Sugiyama Y, Asai K, Yamada K, et al. Decreased levels of irisin, a skeletal muscle cell-derived myokine, are related to emphysema associated with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2017; 12: 765–772.
  28. Natalicchio A, Marrano N, Biondi G, et al. The Myokine Irisin Is Released in Response to Saturated Fatty Acids and Promotes Pancreatic β-Cell Survival and Insulin Secretion. Diabetes. 2017; 66(11): 2849–2856.
  29. Liu S, Du F, Li X, et al. Effects and underlying mechanisms of irisin on the proliferation and apoptosis of pancreatic β cells. PLoS One. 2017; 12(4): e0175498.
  30. Deng X, Huang W, Peng J, et al. Irisin Alleviates Advanced Glycation End Products-Induced Inflammation and Endothelial Dysfunction via Inhibiting ROS-NLRP3 Inflammasome Signaling. Inflammation. 2018; 41(1): 260–275.
  31. Peng J, Deng X, Huang W, et al. Irisin protects against neuronal injury induced by oxygen-glucose deprivation in part depends on the inhibition of ROS-NLRP3 inflammatory signaling pathway. Mol Immunol. 2017; 91: 185–194.
  32. Mazur-Bialy AI, Pocheć E, Zarawski M. Anti-Inflammatory Properties of Irisin, Mediator of Physical Activity, Are Connected with TLR4/MyD88 Signaling Pathway Activation. Int J Mol Sci. 2017; 18(4).

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl