リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Effects of imeglimin on mitochondrial function, AMPK activity, and gene expression in hepatocytes」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Effects of imeglimin on mitochondrial function, AMPK activity, and gene expression in hepatocytes

Hozumi, Kaori Sugawara, Kenji Ishihara, Takaya Ishihara, Naotada Ogawa, Wataru 神戸大学

2023.01.13

概要

Imeglimin is a recently launched antidiabetic drug structurally related to metformin. To provide insight into the pharmacological properties of imeglimin, we investigated its effects on hepatocytes and compared them with those of metformin. The effects of imeglimin on mitochondrial function in HepG2 cells or mouse primary hepatocytes were examined with an extracellular flux analyzer and on gene expression in HepG2 cells by comprehensive RNA-sequencing analysis. The effects of the drug on AMPK activity in HepG2 cells, mouse primary hepatocytes, and mouse liver were also examined. Treatment of HepG2 cells or mouse primary hepatocytes with imeglimin reduced the oxygen consumption rate coupled to ATP production. Imeglimin activated AMPK in these cells whereas the potency was smaller than metformin. Bolus administration of imeglimin in mice also activated AMPK in the liver. Whereas the effects of imeglimin and metformin on gene expression in HepG2 cells were similar overall, the expression of genes encoding proteins of mitochondrial respiratory complex III and complex I was upregulated by imeglimin but not by metformin. Our results suggest that imeglimin and metformin exert similar pharmacological effects on mitochondrial respiration, AMPK activity, and gene expression in cultured hepatocytes, whereas the two drugs differ in their effects on the expression of certain genes related to mitochondrial function.

この論文で使われている画像

参考文献

1. Yendapally, R. et al. A review of phenformin, metformin, and imeglimin. Drug. Dev. Res. 81, 390–401 (2020).

2. Pirags, V. et al. Imeglimin, a novel glimin oral antidiabetic, exhibits a good efficacy and safety profile in type 2 diabetic patients.

Diabetes Obes. Metab. 14, 852–858 (2012).

3. Pacini, G. et al. Imeglimin increases glucose-dependent insulin secretion and improves β-cell function in patients with type 2

diabetes. Diabetes Obes. Metab. 17, 541–545 (2015).

4. Fouqueray, P. et al. Imeglimin—a new oral anti-diabetic that targets the three key defects of type 2 diabetes. J. Diabetes Metab. 2,

4 (2011).

5. Perry, R. J. et al. Imeglimin lowers glucose primarily by amplifying glucose-stimulated insulin secretion in high-fat-fed rodents.

Am. J. Physiol. Endocrinol. Metab. 311, E461–E470 (2016).

6. Vial, G. et al. Imeglimin normalizes glucose tolerance and insulin sensitivity and improves mitochondrial function in liver of a

high-fat, high-sucrose diet mice model. Diabetes 64, 2254–2264 (2015).

7. Hallakou-Bozec, S., Kergoat, M., Moller, D. E. & Bolze, S. Imeglimin preserves islet β-cell mass in Type 2 diabetic ZDF rats. Endocrinol. Diabetes Metab. 4, e00193 (2020).

8. Hallakou-Bozec, S., Kergoat, M., Fouqueray, P., Bolze, S. & Moller, D. E. Imeglimin amplifies glucose-stimulated insulin release

from diabetic islets via a distinct mechanism of action. PLoS ONE 16, e0241651 (2021).

9. Foretz, M., Guigas, B. & Viollet, B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat.

Rev. Endocrinol. 15, 569–589 (2019).

10. Zhou, G. et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108, 1167–1174 (2001).

11. Musi, N., Hirshman, M. F., Nygren, J., Svanfeldt, M. & Rooyackers, O. Metformin increases AMP-activated protein kinase activity

in skeletal muscle of subjects with type 2 diabetes. Diabetes 51, 2074–2081 (2002).

12. Cool, B. et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes

and the metabolic syndrome. Cell Metab. 3, 403–416 (2006).

13. Wang, Y. et al. Metformin improves mitochondrial respiratory activity through activation of AMPK. Cell Rep. 29, 1511-1523.e5

(2019).

14. Foretz, M. et al. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease

in hepatic energy state. J. Clin. Invest. 120, 2355–2369 (2010).

15. Cokorinos, E. C. et al. Activation of skeletal muscle AMPK promotes glucose disposal and glucose lowering in non-human primates

and mice. Cell Metab. 25, 1147-1159.e10 (2017).

16. Boudaba, N. et al. AMPK re-activation suppresses hepatic steatosis but its downregulation does not promote fatty liver development. EBioMedicine 28, 194–209 (2018).

17. Miller, R. A. et al. Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP. Nature 494, 256–260

(2013).

18. Hunter, R. W. et al. Metformin reduces liver glucose production by inhibition of fructose-1-6-bisphosphatase. Nat. Med. 24,

1395–1406 (2018).

19. Esquejo, R. M. et al. Activation of liver AMPK with PF-06409577 corrects NAFLD and lowers cholesterol in rodent and primate

preclinical models. EBioMedicine 31, 122–132 (2018).

20. Vial, G. et al. The mechanism by which imeglimin inhibits gluconeogenesis in rat liver cells. Endocrinol. Diabetes Metab. 4, e00211

(2021).

21. Turrens, J. F. Mitochondrial formation of reactive oxygen species. J. Physiol. 552, 335–344 (2003).

22. Zhao, R. Z., Jiang, S., Zhang, L. & Yu, Z. B. Mitochondrial electron transport chain, ROS generation and uncoupling. Int. J. Mol.

Med. 44, 3–15 (2019).

23. Owen, M. R., Doran, E. & Halestrap, A. P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex

1 of the mitochondrial respiratory chain. Biochem. J. 348, 607–614 (2000).

24. El-Mir, M. Y. et al. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I.

J. Biol. Chem. 275, 223–228 (2000).

25. Stephenne, X. et al. Metformin activates AMP-activated protein kinase in primary human hepatocytes by decreasing cellular energy

status. Diabetologia 54, 3101–3110 (2011).

26. Hardie, D. G. AMPK—sensing energy while talking to other signaling pathways. Cell Metab. 20, 939–952 (2014).

27. Foretz, M., Guigas, B., Bertrand, L., Pollak, M. & Viollet, B. Metformin: From mechanisms of action to therapies. Cell Metab. 20,

953–966 (2014).

28. Heckman-Stoddard, B. M., DeCensi, A., Sahasrabuddhe, V. V. & Ford, L. G. Repurposing metformin for the prevention of cancer

and cancer recurrence. Diabetologia 60, 1639–1647 (2017).

29. Detaille, D. et al. Metformin prevents high-glucose-induced endothelial cell death through a mitochondrial permeability transitiondependent process. Diabetes 54, 2179–2187 (2005).

30. Kim, J. et al. Metformin suppresses lipopolysaccharide (LPS)-induced inflammatory response in murine macrophages via activating transcription factor-3 (ATF-3) induction. J. Biol. Chem. 289, 23246–23255 (2014).

31. Vasamsetti, S. B. et al. Metformin inhibits monocyte-to-macrophage differentiation via AMPK-mediated inhibition of STAT3

activation: Potential role in atherosclerosis. Diabetes 64, 2028–2041 (2015).

32. Day, E. A. et al. Metformin-induced increases in GDF15 are important for suppressing appetite and promoting weight loss. Nat.

Metab. 1, 1202–1208 (2019).

33. Coll, A. P. et al. GDF15 mediates the effects of metformin on body weight and energy balance. Nature 578, 444–448 (2020).

34. Liu, Z. et al. Metallothionein 1 family profiling identifies MT1X as a tumor suppressor involved in the progression and metastastatic

capacity of hepatocellular carcinoma. Mol. Carcinog. 57, 1435–1444 (2018).

35. Lei, Y. et al. Comprehensive analysis of partial epithelial mesenchymal transition-related genes in hepatocellular carcinoma. J. Cell

Mol. Med. 25, 448–462 (2021).

36. Park, K. C. et al. Identification of differential phosphorylation and sub-cellular localization of the metastasis suppressor, NDRG1.

Biochim. Biophys. Acta Mol. Basis Dis. 1864, 2644–2663 (2018).

37. Ben-Sahra, I. et al. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer

Res. 71, 4366–4372 (2011).

38. Wilcock, C. & Bailey, C. J. Accumulation of metformin by tissues of the normal and diabetic mouse. Xenobiotica 24, 49–57 (1994).

39. He, L. & Wondisford, F. E. Metformin action: concentrations matter. Cell Metab. 21, 159–162 (2015).

40. Fontaine, E. Metformin-induced mitochondrial complex I inhibition: Facts, uncertainties, and consequences. Front. Endocrinol.

(Lausanne) 19, 753 (2018).

41. Berry, M. N. & Friend, D. S. High-yield preparation of isolated rat liver parenchymal cells: a biochemical and fine structural study.

J. Cell Biol. 43, 506–520 (1969).

42. Matsumoto, M. et al. Role of the insulin receptor substrate 1 and phosphatidylinositol 3-kinase signaling pathway in insulin-induced

expression of sterol regulatory element binding protein 1c and glucokinase genes in rat hepatocytes. Diabetes 51, 1672–1680 (2002).

43. Yoshino, K. et al. Canagliflozin ameliorates hepatic fat deposition in obese diabetic mice: Role of prostaglandin E2. Biochem.

Biophys. Res. Commun. 557, 62–68 (2021).

Scientific Reports |

(2023) 13:746 |

https://doi.org/10.1038/s41598-023-27689-y

11

Vol.:(0123456789)

www.nature.com/scientificreports/

Acknowledgements

We thank C. Aoki (Kobe University) for assistance with mouse experiments.

Author contributions

K.H., K.S., and W.O. designed experiments and analyzed the data. K.H. performed experiments. K.S. and T.I.

contributed to experiments. K.H. and W.O. wrote the manuscript. K.S., T.I., and N.I. contributed to discussion.

W.O. is the guarantor of this work.

Funding

This work was supported by Sumitomo Pharma Co. Ltd (to W.O.) and AMED-CREST (Grant Number

JP22gm1110006 to N.I.) as well as Japan Society for the Promotion of Science KAKENHI grants (20K08884),

Takeda Science Foundation, MSD Life Science Foundation, Cell Science Research Foundation, Foundation for

Biomedical Research and Innovation at Kobe, and Suzuken Memorial Foundation (all to K.S).

Competing interests WO has received lecture fees from Sumitomo Pharma. All other authors declare no conflict of interest.

Additional information

Supplementary Information The online version contains supplementary material available at https://​doi.​org/​

10.​1038/​s41598-​023-​27689-y.

Correspondence and requests for materials should be addressed to W.O.

Reprints and permissions information is available at www.nature.com/reprints.

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

institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International

License, which permits use, sharing, adaptation, distribution and reproduction in any medium or

format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the

Creative Commons licence, and indicate if changes were made. The images or other third party material in this

article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the

material. If material is not included in the article’s Creative Commons licence and your intended use is not

permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/.

© The Author(s) 2023

Scientific Reports |

Vol:.(1234567890)

(2023) 13:746 |

https://doi.org/10.1038/s41598-023-27689-y

12

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る