Atractylodes chinensis volatile oil up-regulated IGF-1 to improve diabetic gastroparesis in rats

Document Type : Original Article

Authors

1 Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China Science and Technology, Qingdao, China

2 Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China

3 Shandong Xinhua Pharmaceutical Company Limited, Zibo, China

4 Qingdao Women and Children’s Hospital, Qingdao, China

5 Department of Pharmacy College of Chemical Engineering Qingdao University of Science and Technology Qingdao, 266042 China

Abstract

Objective(s): Diabetic gastroparesis (DGP) is one of the main complications of diabetes, and more than half of diabetes cases are accompanied by gastroparesis. This study aims to explore the effect of Atractylodes chinensis volatile oil (ACVO) on DGP rats.
Materials and Methods: The rats were injected with STZ combined with a high-sugar and high-fat diet in an irregular manner to establish the DGP model. ACVO at different doses (9.11 mg/kg, 18.23 mg/kg, and 36.45 mg/kg)  were given by intragastric administration. A mixture of cisapride and metformin was used as the positive control. At the end of the experiment, gastric emptying and intestinal propulsion were determined. Then the tissue samples and blood were taken from each group for serum analysis, western blot and immunopathological examination. 
Results: After treatment with ACVO, body weight increased and blood glucose decreased when compared with rats in the DGP group. Gastric emptying and intestinal propulsion were accelerated, and gastric acid secretion increased. The serum insulin-like growth factor-1 (IGF-1) level was increased. Protein expressions and positive cells of IGF-1 receptor (IGF-1R), acetylcholine transferase (CHAT), and stem cell factors (SCF) in the stomach were significantly increased determined by western blot and immunofluorescence staining. The morphology and the number of interstitial cells of Cajal (ICCs) in the stomach were restored, determined by hematoxylin and eosin staining and immunohistochemical staining, respectively. 
Conclusion: ACVO effectively alleviated DGP in rats, and its mechanism may be related to the up-regulation of IGF-1/IGF-1R signaling.

Keywords


1. Bharucha AE, Kudva YC, Prichard DO. Diabetic gastroparesis. Endocr Rev 2019; 40:1318-1352.
2. Kumar M, Chapman A, Javed S, Alam U, Malik RA, Azmi S. The investigation and treatment of diabetic gastroparesis. Clin Ther 2018; 40:850-861.
3. Avalos DJ, Sarosiek I, Loganathan P, McCallum RW. Diabetic gastroparesis: Current challenges and future prospects. Clin Exp Gastroenterol 2018; 11:347-363.
4. Sullivan A, Temperley L, Ruban A. Pathophysiology, aetiology and treatment of gastroparesis. Dig Dis Sci 2020; 65:1615-1631.
5. Zhang H, Han T, Sun LN, Huang BK, Chen YF, Zheng HC, et al. Regulative effects of essential oil from Atractylodes lancea on delayed gastric emptying in stress-induced rats. Phytomedicine 2008; 15:602-611.
6. Luo XQ, Gu YH, Wu ZY. [Comparison of the effect of eight kinds of volatile oil of Chinese material medica on percutaneous absorption of ibuprofen in vitro]. Zhong Yao Cai 2007; 30:571-573.
7. Ishii T, Okuyama T, Noguchi N, Nishidono Y, Okumura T, Kaibori M, et al. Antiinflammatory constituents of Atractylodes chinensis rhizome improve glomerular lesions in immunoglobulin A nephropathy model mice. J Nat Med 2020; 74:51-64.
8. Hossen MJ, Chou JY, Li SM, Fu XQ, Yin C, Guo H, et al. An ethanol extract of the rhizome of Atractylodes chinensis exerts anti-gastritis activities and inhibits Akt/NF-kappaB signaling. J Ethnopharmacol 2019; 228:18-25.
9. Shimato Y, Ota M, Asai K, Atsumi T, Tabuchi Y, Makino T. Comparison of byakujutsu (Atractylodes rhizome) and sojutsu (Atractylodes lancea rhizome) on anti-inflammatory and immunostimulative effects in vitro. J Nat Med 2018; 72:192-201.
10. Chu SS, Jiang GH, Liu ZL. Insecticidal compounds from the essential oil of Chinese medicinal herb Atractylodes chinensis. Pest Manag Sci 2011; 67:1253-1257.
11. Kim JH, Doh EJ, Lee G. Chemical differentiation of genetically identified atractylodes japonica, A. macrocephala, and A. chinensis rhizomes using high-performance liquid chromatography with chemometric analysis. Evid Based Complement Alternat Med 2018; 2018:4860371.
12. Feng W, Ao H, Yue S, Peng C. Systems pharmacology reveals the unique mechanism features of Shenzhu Capsule for treatment of ulcerative colitis in comparison with synthetic drugs. Sci Rep 2018; 8:16160.
13. Yang S, Wu B, Sun H, Sun T, Han K, Li D, et al. Impaired insulin/IGF-1 is responsible for diabetic gastroparesis by damaging myenteric cholinergic neurones and interstitial cells of Cajal. Biosci Rep 2017; 37.
14. Ren K, Yong C, Yuan H, Cao B, Zhao K, Wang J. TNF-alpha inhibits SCF, ghrelin, and substance P expressions through the NF-kappaB pathway activation in interstitial cells of Cajal. Braz J Med Biol Res 2018; 51:e7065.
15. Gligorijevic N, Robajac D, Nedic O. Enhanced platelet sensitivity to IGF-1 in patients with type 2 diabetes mellitus. Biochemistry (Mosc) 2019; 84:1213-1219.
16. Brussee V, Cunningham FA, Zochodne DW. Direct insulin signaling of neurons reverses diabetic neuropathy. Diabetes 2004; 53:1824-1830.
17. Zhuang HX, Snyder CK, Pu SF, Ishii DN. Insulin-like growth factors reverse or arrest diabetic neuropathy: effects on hyperalgesia and impaired nerve regeneration in rats. Exp Neurol 1996; 140:198-205.
18. Byrne CD, Targher G. NAFLD: A multisystem disease. J Hepatol 2015; 62:S47-64.
19. Mule F, Amato A, Serio R. Gastric emptying, small intestinal transit and fecal output in dystrophic (mdx) mice. J Physiol Sci 2010; 60:75-79.
20. Improta G, Broccardo M. Effect of selective mu 1, mu 2 and delta 2 opioid receptor agonists on gastric functions in the rat. Neuropharmacology 1994; 33:977-981.
21. Brage R, Cortijo J, Esplugues J, Esplugues JV, Marti-Bonmati E, Rodriguez C. Effects of calcium channel blockers on gastric emptying and acid secretion of the rat in vivo. Br J Pharmacol 1986; 89:627-633.
22. Zhang GQ, Yang S, Li XS, Zhou DS. Expression and possible role of IGF-IR in the mouse gastric myenteric plexus and smooth muscles. Acta Histochem 2014; 116:788-794.
23. Krishnasamy S, Abell TL. Diabetic gastroparesis: Principles and current trends in management. Diabetes Ther 2018; 9:1-42.
24. Wang B, Zeng KW, Hong ZF, Ti GX, Wang LY, Lu P, et al. Banxia xiexin decoction treats diabetic gastroparesis through PLC-IP3-Ca(2+)/NO-cGMP-PKG signal pathway. Chin J Integr Med 2020; 26:833-838.
25. Zhang MH, Fang XS, Guo JY, Jin Z. Effects of AMPK on apoptosis and energy metabolism of gastric smooth muscle cells in rats with diabetic gastroparesis. Cell Biochem Biophys 2019; 77:165-177.
26. Sanders KM, Ward SM, Koh SD. Interstitial cells: Regulators of smooth muscle function. Physiol Rev 2014; 94:859-907.
27. Chen Y, Wang H, Li H, Liu S. Long-pulse gastric electrical stimulation repairs interstitial cells of cajal and smooth muscle cells in the gastric antrum of diabetic rats. Gastroenterol Res Pract 2018; 2018:6309157.
28. Wang Y, Xu XY, Tang YR, Yang WW, Yuan YF, Ning YJ, et al. Effect of endogenous insulin-like growth factor and stem cell factor on diabetic colonic dysmotility. World J Gastroenterol 2013; 19:3324-3331.
29. Torihashi S, Yoshida H, Nishikawa S, Kunisada T, Sanders KM. Enteric neurons express Steel factor-lacZ transgene in the murine gastrointestinal tract. Brain Res 1996; 738:323-328.
30. Rich A, Miller SM, Gibbons SJ, Malysz J, Szurszewski JH, Farrugia G. Local presentation of Steel factor increases expression of c-kit immunoreactive interstitial cells of Cajal in culture. Am J Physiol Gastrointest Liver Physiol 2003; 284:G313-320.
31. Horvath VJ, Vittal H, Lorincz A, Chen H, Almeida-Porada G, Redelman D, et al. Reduced stem cell factor links smooth myopathy and loss of interstitial cells of cajal in murine diabetic gastroparesis. Gastroenterology 2006; 130:759-770.
32. Anetsberger D, Kurten S, Jabari S, Brehmer A. Morphological and immunohistochemical characterization of human intrinsic gastric neurons. Cells Tissues Organs 2018; 206:183-195.
33. Bu J, Qiao X, He Y, Liu J. Colonic electrical stimulation improves colonic transit in rotenone-induced Parkinson’s disease model through affecting enteric neurons. Life Sci 2019; 231:116581.