Vol 60, No 1 (2022)
Original paper
Published online: 2022-02-14

open access

Page views 5988
Article views/downloads 748
Get Citation

Connect on Social Media

Connect on Social Media

Insulin-like growth factor-1 inhibits apoptosis of rat gastric smooth muscle cells under high glucose condition via adenosine monophosphate-activated protein kinase (AMPK) pathway

Xiang-zi Zhang12, Yan Sun1, Mo-han Zhang1, Zheng Jin1
Pubmed: 35156189
Folia Histochem Cytobiol 2022;60(1):74-88.

Abstract

Introduction. Diabetic gastroparesis (DGP) is a common chronic complication of diabetes characterized by decreased gastric motility, and an effective number of gastric smooth muscle cells (GSMCs) ensures gastric motility. A previous study documented that apoptosis was present in gastric smooth muscles in rats with DGP and adenosine monophosphate-activated protein kinase (AMPK) was an important factor of apoptosis of rat GSMCs cultured under high glucose conditions. This study aimed to explore the effect of insulin-like growth factor-1 (IGF-1) on apoptosis of high glucose cultured rat GSMCs after silencing of AMPK and elucidate the underlying mechanism.

Material and methods. A total of 120 rats were divided into normal control (NC, n = 20), diabetic gastroparesis (DGP, n = 50) and DGP + IGF-1 (n = 50) groups. After establishing the rat model of DGP, rats in the DGP+IGF-1 group received an intraperitoneal injection of IGF-1 at a dose of 1.5 μg/kg/d for 10 weeks. The level of AMPK activity, liver kinase B1 (LKB1) activity, and calcium/calmodulin-dependent protein kinase b (CaMKKb) expression in rat gastric smooth muscle tissues was detected by Western blot analysis. Apoptosis in rat gastric smooth muscle tissues was detected by TUNEL assay. We also cultured rat GSMCs in vitro under high glucose (HG) condition (35 mM), incubated cells with IGF-1, and silenced AMPK with siRNA. The cells were divided into HG, HG + IGF-1, HG + siRNA, and HG + siRNA + IGF-1 groups. The apoptosis rates of rat GSMCs after silencing AMPK were detected by TUNEL assay and flow cytometry, and apoptosis-related protein expression in rat GSMCs was detected by Western blot.

Results. IGF-1 decreased LKB1 activity, CaMKKb expression, AMPK activity, and inhibited apoptosis in rat gastric smooth muscle tissues. Compared with rat GSMCs cultured in vitro under HG conditions, apoptosis rates were reduced after treatment with IGF-1 and AMPK silencing (both p < 0.01). Apoptosis rates were higher in the HG + siRNA group compared with the HG + IGF-1 group (p < 0.05). IGF-1 down-regulated the expression of calcium/calmodulin-dependent kinase II (CaMKII) and p53, up-regulated the expression of p21, PLC-b3, PI3K p110 Ser1070, and the activities of Akt, p70S6K, mTORC1, and mTORC2. IGF-1 also up-regulated Bcl-2 expression and down-regulated the expression of BAX and Caspase-3.

Conclusions. IGF-1 can inhibit the apoptosis of rat GSMCs under high glucose conditions, its mechanism may be related to the regulation of expression and activity of p53, PI3K, TSC-2, Akt, mTOR, 4E-BP1, p70S6K, p21, CaMKII, and PLC-b3 in rat GSMCs acting through AMPK pathway.

Article available in PDF format

View PDF Download PDF file

References

  1. Zhao J, Frøkjaer JB, Drewes AM, et al. Upper gastrointestinal sensory-motor dysfunction in diabetes mellitus. World J Gastroenterol. 2006; 12(18): 2846–2857.
  2. Ramzan Z, Duffy F, Gomez J, et al. Continuous glucose monitoring in gastroparesis. Dig Dis Sci. 2011; 56(9): 2646–2655.
  3. Hrdinka M, Yabal M. Inhibitor of apoptosis proteins in human health and disease. Genes Immun. 2019; 20(8): 641–650.
  4. Zhang MH, Fang XS, Guo JY, et al. Effects of AMPK on Apoptosis and Energy Metabolism of Gastric Smooth Muscle Cells in Rats with Diabetic Gastroparesis. Cell Biochem Biophys. 2019; 77(2): 165–177.
  5. Pang L, Yang K, Zhang Z. High-glucose environment accelerates annulus fibrosus cell apoptosis by regulating endoplasmic reticulum stress. Biosci Rep. 2020; 40(7).
  6. Yang M, Lin Y, Wang Y, et al. High-glucose induces cardiac myocytes apoptosis through Foxo1 /GRK2 signaling pathway. Biochem Biophys Res Commun. 2019; 513(1): 154–158.
  7. Meng S, Cao J, He Q, et al. Metformin activates AMP-activated protein kinase by promoting formation of the αβγ heterotrimeric complex. J Biol Chem. 2015; 290(6): 3793–3802.
  8. Nambam B, Schatz D. Growth hormone and insulin-like growth factor-I axis in type 1 diabetes. Growth Horm IGF Res. 2018; 38: 49–52.
  9. Li H, Kong R, Wan B, et al. Initiation of PI3K/AKT pathway by IGF-1 decreases spinal cord injury-induced endothelial apoptosis and microvascular damage. Life Sci. 2020; 263: 118572.
  10. Luo Li, Lu AM, Wang Y, et al. Chronic resistance training activates autophagy and reduces apoptosis of muscle cells by modulating IGF-1 and its receptors, Akt/mTOR and Akt/FOXO3a signaling in aged rats. Exp Gerontol. 2013; 48(4): 427–436.
  11. Vanamala J, Reddivari L, Radhakrishnan S, et al. Resveratrol suppresses IGF-1 induced human colon cancer cell proliferation and elevates apoptosis via suppression of IGF-1R/Wnt and activation of p53 signaling pathways. BMC Cancer. 2010; 10: 238.
  12. Zhang MH, Jiang JZ, Cai YL, et al. Significance of dynamic changes in gastric smooth muscle cell apoptosis, PI3K-AKT-mTOR and AMPK-mTOR signaling in a rat model of diabetic gastroparesis. Mol Med Rep. 2017; 16(2): 1530–1536.
  13. Fang XS, Zhang MH, Zhang XZ, et al. Insulin-like growth factor-1 inhibits the apoptosis of rat gastric smooth muscle cells cultured under high glucose condition through PI3K-Akt-PKC-Ca2+ pathway. Biotechnology & Biotechnological Equipment. 2019; 33(1): 456–464.
  14. Miricescu D, Totan A, Stanescu-Spinu II, et al. PI3K/AKT/mTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects. Int J Mol Sci. 2020; 22(1).
  15. Martini M, De Santis MC, Braccini L, et al. PI3K/AKT signaling pathway and cancer: an updated review. Ann Med. 2014; 46(6): 372–383.
  16. Yan J, Wang C, Jin Y, et al. Catalpol ameliorates hepatic insulin resistance in type 2 diabetes through acting on AMPK/NOX4/PI3K/AKT pathway. Pharmacol Res. 2018; 130: 466–480.
  17. Liu Y, Deng J, Fan D. Ginsenoside Rk3 ameliorates high-fat-diet/streptozocin induced type 2 diabetes mellitus in mice via the AMPK/Akt signaling pathway. Food Funct. 2019; 10(5): 2538–2551.
  18. Ersahin T, Tuncbag N, Cetin-Atalay R. The PI3K/AKT/mTOR interactive pathway. Mol Biosyst. 2015; 11(7): 1946–1954.
  19. Shariati M, Meric-Bernstam F. Targeting AKT for cancer therapy. Expert Opin Investig Drugs. 2019; 28(11): 977–988.
  20. Yin H, Zhao L, Li S, et al. Impaired Cellular Energy Metabolism Contributes to Duck-Enteritis-Virus-Induced Autophagy via the AMPK-TSC2-MTOR Signaling Pathway. Front Cell Infect Microbiol. 2017; 7: 423.
  21. McCampbell AS, Mittelstadt ML, Dere R, et al. Loss of p27 Associated with Risk for Endometrial Carcinoma Arising in the Setting of Obesity. Curr Mol Med. 2016; 16(3): 252–265.
  22. Bonucci M, Kuperwasser N, Barbe S, et al. mTOR and S6K1 drive polycystic kidney by the control of Afadin-dependent oriented cell division. Nat Commun. 2020; 11(1): 3200.
  23. Rozas NS, Redell JB, Hill JL, et al. Genetic activation of mTORC1 signaling worsens neurocognitive outcome after traumatic brain injury. J Neurotrauma. 2015; 32(2): 149–158.
  24. Al-Attar R, Childers CL, Nguyen VuC, et al. Differential protein phosphorylation is responsible for hypoxia-induced regulation of the Akt/mTOR pathway in naked mole rats. Comp Biochem Physiol A Mol Integr Physiol. 2020; 242: 110653.
  25. Al Dera H, Eleawa SM, Al-Hashem FH, et al. Enhanced hepatic insulin signaling in the livers of high altitude native rats under basal conditions and in the livers of low altitude native rats under insulin stimulation: a mechanistic study. Arch Physiol Biochem. 2017; 123(3): 145–158.
  26. Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell. 2017; 168(6): 960–976.
  27. Sangaunchom P, Dharmasaroja P. Caffeine Potentiates Ethanol-Induced Neurotoxicity Through mTOR/p70S6K/4E-BP1 Inhibition in SH-SY5Y Cells. Int J Toxicol. 2020; 39(2): 131–140.
  28. Mukaida S, Evans BA, Bengtsson T, et al. Adrenoceptors promote glucose uptake into adipocytes and muscle by an insulin-independent signaling pathway involving mechanistic target of rapamycin complex 2. Pharmacol Res. 2017; 116: 87–92.
  29. Sutton LP, Caron MG. Essential role of D1R in the regulation of mTOR complex1 signaling induced by cocaine. Neuropharmacology. 2015; 99: 610–619.
  30. Zhang XH, Chen SY, Tang L, et al. Myricetin induces apoptosis in HepG2 cells through Akt/p70S6K/bad signaling and mitochondrial apoptotic pathway. Anticancer Agents Med Chem. 2013; 13(10): 1575–1581.
  31. Ragan TJ, Ross DB, Keshwani MM, et al. Expression, purification, and characterization of a structurally disordered and functional C-terminal autoinhibitory domain (AID) of the 70 kDa 40S ribosomal protein S6 kinase-1 (S6K1). Protein Expr Purif. 2008; 57(2): 271–279.
  32. Keshwani MM, von Daake S, Newton AC, et al. Hydrophobic motif phosphorylation is not required for activation loop phosphorylation of p70 ribosomal protein S6 kinase 1 (S6K1). J Biol Chem. 2011; 286(26): 23552–23558.
  33. Magnuson B, Ekim B, Fingar DC. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 2012; 441(1): 1–21.
  34. Cheng CY, Kao ST, Lee YC. Ferulic acid ameliorates cerebral infarction by activating Akt/mTOR/4E‑BP1/Bcl‑2 anti‑apoptotic signaling in the penumbral cortex following permanent cerebral ischemia in rats. Mol Med Rep. 2019; 19(2): 792–804.
  35. Cheng CY, Kao ST, Lee YC. Ferulic acid ameliorates cerebral infarction by activating Akt/mTOR/4E‑BP1/Bcl‑2 anti‑apoptotic signaling in the penumbral cortex following permanent cerebral ischemia in rats. Mol Med Rep. 2019; 19(2): 792–804.
  36. Qin X, Jiang B, Zhang Y. 4E-BP1, a multifactor regulated multifunctional protein. Cell Cycle. 2016; 15(6): 781–786.
  37. So L, Lee J, Palafox M, et al. The 4E-BP-eIF4E axis promotes rapamycin-sensitive growth and proliferation in lymphocytes. Sci Signal. 2016; 9(430): ra57.
  38. Lieschke E, Wang Z, Kelly GL, et al. Discussion of some 'knowns' and some 'unknowns' about the tumour suppressor p53. J Mol Cell Biol. 2019; 11(3): 212–223.
  39. Liu Y, Tavana O, Gu W. p53 modifications: exquisite decorations of the powerful guardian. J Mol Cell Biol. 2019; 11(7): 564–577.
  40. Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst). 2016; 42: 63–71.
  41. Yu D, Liu Q, Qiao Bo, et al. Exposure to acrylamide inhibits uterine decidualization via suppression of cyclin D3/p21 and apoptosis in mice. J Hazard Mater. 2020; 388: 121785.
  42. Kaluzki I, Hailemariam-Jahn T, Doll M, et al. Dimethylfumarate Inhibits Colorectal Carcinoma Cell Proliferation: Evidence for Cell Cycle Arrest, Apoptosis and Autophagy. Cells. 2019; 8(11).
  43. He P, Li Z, Xu F, et al. AMPK Activity Contributes to G2 Arrest and DNA Damage Decrease via p53/p21 Pathways in Oxidatively Damaged Mouse Zygotes. Front Cell Dev Biol. 2020; 8: 539485.
  44. Yang D, Zhang Qi, Ma Y, et al. Augmenting the therapeutic efficacy of adenosine against pancreatic cancer by switching the Akt/p21-dependent senescence to apoptosis. EBioMedicine. 2019; 47: 114–127.
  45. McConkey D, Orrenius S. The Role of Calcium in the Regulation of Apoptosis. Biochem Biophys Res Commun. 1997; 239(2): 357–366.
  46. Finkelstein M, Etkovitz N, Breitbart H. Ca signaling in mammalian spermatozoa. Mol Cell Endocrinol. 2020; 516: 110953.
  47. Makhlouf G, Murthy K. Signal Transduction in Gastrointestinal Smooth Muscle. Cell Signal. 1997; 9(3-4): 269–276.
  48. Nalli AD, Kumar DP, Al-Shboul O, et al. Regulation of Gβγi-dependent PLC-β3 activity in smooth muscle: inhibitory phosphorylation of PLC-β3 by PKA and PKG and stimulatory phosphorylation of Gαi-GTPase-activating protein RGS2 by PKG. Cell Biochem Biophys. 2014; 70(2): 867–880.
  49. Simon B, Huart AS, Temmerman K, et al. Molecular mechanisms of protein kinase regulation by calcium/calmodulin. Bioorg Med Chem. 2015; 23(12): 2749–2760.
  50. Banerjee C, Khatri P, Raman R, et al. Role of calmodulin-calmodulin kinase II, cAMP/protein kinase A and ERK 1/2 on Aeromonas hydrophila-induced apoptosis of head kidney macrophages. PLoS Pathog. 2014; 10(4): e1004018.
  51. Jayanthi LD, Wilson JJ, Montalvo J, et al. Differential regulation of mammalian brain-specific proline transporter by calcium and calcium-dependent protein kinases. Br J Pharmacol. 2000; 129(3): 465–470.



Folia Histochemica et Cytobiologica