Andrographis paniculata Ethanol Extract Alleviates High Glucose-induced Senescence of Human Umbilical Vein Endothelial Cells via the Regulation of mTOR and SIRT1 Pathways
Abstract
BACKGROUND: Chronic exposure of high glucose (HG) in endothelial cell induces senescence which may contribute to the development and progression of age-related diseases including insulin resistance. Andrographis paniculata improves insulin resistance in recent in vitro and in vivo studies. Anti-inflammatory and antioxidant properties of A. paniculata may be the new therapeutic approach to inhibiting premature senescence. However, the senolytic effect of A. paniculata on endothelial cells has not been investigated comprehensively. This study was conducted to evaluate the effect of A. paniculata extract on HG-induced endothelial cell senescence and the underlying mechanisms.
METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with 33 mM HG and 7.5 μg/mL A. paniculata extract for 48 hours. The expressions of p16, p21, interleukin (IL)-6, IL-8, insulin receptor substrate (IRS)-1, mammalian target of rapamycin, and sirtuin 1 (SIRT1) were measured by performing real-time quantitative polymerase chain reaction (RT-qPCR). The senescence-associated-β-galactosidase (SA-β-gal) staining was performed to observe the positive-stained senescent cells, while the cell surface expression of IL-1α was examined with flow cytometry method.
RESULTS: A. paniculata extract reversed senescence in HUVECs under HG conditions by reducing mRNA expressions of p16 and p21, the number of SA-β-gal-positive-stained cells, and the expression of IL-1α on cell surface, which decreased the activation of IL-6 and IL-8. In addition, A. paniculata extract decreased the mRNA expression of mTOR and increased the mRNA expressions of IRS-1 as well as SIRT1.
CONCLUSION: A. paniculata extract ameliorated senescence and improved insulin sensitivity by regulating the mTOR, SIRT1, and IRS-1 mRNA expressions on HG-treated HUVECs.
KEYWORDS: Andrographis paniculata, endothelial cell, senescence, high glucose, nutrient-sensing pathways
Full Text:
PDFReferences
González-Gualda E, Baker AG, Fruk L, Muñoz-Espín D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J. 2021; 288(1): 56-80, CrossRef.
Barinda AJ, Arozal W, Yuasa S. A review of pathobiological mechanisms and potential application of medicinal plants for vascular aging: focus on endothelial cell senescence. Med J Indones. 2022; 31(2): 132-40, CrossRef.
Orjalo AV, Bhaumik D, Gengler BK, Scott GK, Campisi J. Cell surface-bound IL-1α is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc Natl Acad Sci USA. 2009; 106(40): 17031-6, CrossRef.
Shimizu I, Minamino T. Cellular senescence in cardiac diseases. J Cardiol. 2019; 74(4): 313-9, CrossRef.
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013; 153(6): 1194, CrossRef.
Rabiee A, Krüger M, Ardenkjær-Larsen J, Kahn CR, Emanuelli B. Distinct signalling properties of insulin receptor substrate (IRS)-1 and IRS-2 in mediating insulin/IGF-1 action. Cell Signal. 2018; 47: 1-15, CrossRef.
Barinda AJ, Ikeda K, Nugroho DB, Wardhana DA, Sasaki N, Honda S, et al. Endothelial progeria induces adipose tissue senescence and impairs insulin sensitivity through senescence associated secretory phenotype. Nat Commun. 2020; 11(1): 481, CrossRef.
Power H, Valtchev P, Dehghani F, Schindeler A. Strategies for senolytic drug discovery. Aging Cell. 2023; 22(10): e13948, CrossRef.
Islam MT, Tuday E, Allen S, Kim J, Trott DW, Holland WL, et al. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell. 2023; 22(2): e13767, CrossRef.
Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine. 2019; 47: 446-56, CrossRef.
Wong LP, Alias H, Tan KM, Wong PF, Murugan DD, Hu Z, et al. Exploring the perspectives of pharmaceutical experts and healthcare practitioners on senolytic drugs for vascular aging-related disorder: A qualitative study. Front Pharmacol. 2023; 14: 1254470, CrossRef.
Benzie IFF, Wachtel-Galor S. Herbal Medicine: Biomolecular and Clinical Aspects. 2nd ed. Boca Raton: Taylor & Francis; 2011, CrossRef.
Eziefule OM, Arozal W, Wanandi SI, Dewi S, Nafrialdi, Saraswati M, et al. Andrographis paniculata: A potential supplementary therapy for cardiovascular diseases - A systematic review of its effects and molecular actions. J Pharm Pharmacogn Res. 2024; 12(3): 487-513, CrossRef.
Mussard E, Jousselin S, Cesaro A, Legrain B, Lespessailles E, Esteve E, et al. Andrographis paniculata and its bioactive diterpenoids against inflammation and oxidative stress in keratinocytes. Antioxidants. 2020; 9(6): 530, CrossRef.
Mussard E, Jousselin S, Cesaro A, Legrain B, Lespessailles E, Esteve E, et al. Andrographis paniculata and its bioactive diterpenoids protect dermal fibroblasts against inflammation and oxidative stress. Antioxidants. 2020; 9(5): 432, CrossRef.
Lu CY, Yang YC, Li CC, Liu KL, Lii CK, Chen HW. Andrographolide inhibits TNFot-induced ICAM-1 expression via suppression of NADPH oxidase activation and induction of HO-1 and GCLM expression through the PI3K/Akt/Nrf2 and PI3K/Akt/AP-1 pathways in human endothelial cells. Biochem Pharmacol. 2014; 91(1): 40-50, CrossRef.
Lindsay CB, Zolezzi JM, Rivera DS, Cisternas P, Bozinovic F, Inestrosa NC. Andrographolide reduces neuroinflammation and oxidative stress in aged octodon degus. Mol Neurobiol. 2020; 57(2): 1131-45, CrossRef.
Serrano FG, Tapia-Rojas C, Carvajal FJ, Hancke J, Cerpa W, Inestrosa NC. Andrographolide reduces cognitive impairment in young and mature AβPPswe/PS-1 mice. Mol Neurodegener. 2014; 9: 61, CrossRef.
Tarigan TJE, Purwaningsih EH, Yusra, Abdullah M, Nafrialdi, Prihartono J, et al. Effects of sambiloto (Andrographis paniculata) on GLP-1 and DPP-4 concentrations between normal and prediabetic subjects: A crossover study. Evid Based Complement Alternat Med. 2022; 2022: 1535703, CrossRef.
Arozal W, Wanandi S, Louisa M, Wuyung PE, Noviana D, Eziefule OM, et al. Evaluating the acute toxicity and in vivo protective effect of standardized Andrographis paniculata extract against doxorubicin-induced cardiotoxicity in sprague-dawley Rats. Trop J Nat Prod Res. 2024; 8(5): 7134-41, CrossRef.
Wong PF, Tong KL, Jamal J, Khor ES, Lai SL, Mustafa MR. Senescent HUVECs-secreted exosomes trigger endothelial barrier dysfunction in young endothelial cells. EXCLI J. 2019; 18: 764-76, PMID.
Yi T, Zhao H, Zhang J, Shan H, Meng X, Zhang J. Effect of aspirin on high glucose-induced senescence of endothelial cells . Chin Med J. 2009; 122(24): 3055-61, PMID.
Ren H, Han W, Wang S, Zhao B, Miao J, Lin Z. A novel sulfur dioxide probe inhibits high glucose-induced endothelial cell senescence. Front Physiol. 2022; 13: 979986, CrossRef.
Song Z, Wang M, Liu Y, Hao B, Niu H, Liu D, et al. Hydrogen sulfide ameliorates high glucose-induced endothelial cell senes-cence by suppressing oxidative stress. Chin J Pathophysiol. 2014; 12: 1345-50, CrossRef.
Xue WL, Chen RQ, Zhang QQ, Li XH, Cao L, Li MY, et al. Hydrogen sulfide rescues high glucose-induced migration dysfunction in HUVECs by upregulating miR-126-3p. Am J Physiol Cell Physiol. 2020; 318(5): C857-69, CrossRef.
Xu J, Liu M, Yu M, Shen J, Zhou J, Hu J, et al. RasGRP1 is a target for VEGF to induce angiogenesis and involved in the endothelial‐protective effects of metformin under high glucose in HUVECs. IUBMB Life. 2019; 71(9): 1391-400, CrossRef.
Chen L, Yin Y, Liu G. Metformin alleviates bevacizumab-induced vascular endothelial injury by up-regulating GDF15 and activating the PI3K/AKT/FOXO/ PPARγ signaling pathway. Ann Transl Med. 2021; 9(20): 1547, CrossRef.
Goel N, Gajbhiye RL, Saha M, Nagendra C, Reddy AM, Ravichandiran V, et al. A comparative assessment of in vitro cytotoxic activity and phytochemical profiling of Andrographis nallamalayana J.L.Ellis and Andrographis paniculata (Burm. f.) Nees using UPLC-QTOF-MS/MS approach. RSC Adv. 2021; 11(57): 35918-36, CrossRef.
Kumar S, Singh B, Bajpai V. Andrographis paniculata (Burm.f.) Nees: Traditional uses, phytochemistry, pharmacological properties and quality control/quality assurance. J Ethnopharmacol. 2021; 275: 114054, CrossRef.
Laiso RAN, Ferreira JC, Rici REG, Cabral LG de S, Maria DA. Cytotoxic effect of Andrographis paniculata associated with 2-aminoethyl dihydrogen phosphate in triple-negative breast cells. Curr Issues Mol Biol. 2024; 46(1): 527-41, CrossRef.
You J, Roh KB, Li Z, Liu G, Tang J, Shin S, et al. The antiaging properties of andrographis paniculata by activation epidermal cell stemness. Molecules. 2015; 20(9): 17557-69, CrossRef.
Chen HW, Chen CC. Andrographis paniculata ameliorates insulin resistance in high fat diet-fed mice and 3T3-L1 adipocytes (P21-074-19). Curr Dev Nutr. 2019; 3(Suppl 1): nzz041.P21-074-19, CrossRef.
Ardika R, Budiono B, Widiastiti N, Maharani N, Susilaningsih N, Sandra F. Andrographis paniculata leaves extract inhibit TNF-α and caspase-3 expression of septic rats' intestinal tissues. Indones Biomed J. 2024; 16(1): 66-71, CrossRef.
Kevin M, Widyastiti N, Budijitno S, Prajoko Y, Susilaningsih N. The protective effect of Andrographis paniculata against lipopolysaccharide-induced sepsis in lung tissues of a rat model through the decrease of ICAM-1 and E-selectin expression. Indones Biomed J. 2023; 15(6): 411-9, CrossRef.
Duan MX, Zhou H, Wu QQ, Liu C, Xiao Y, Deng W, et al. Andrographolide protects against HG-induced inflammation, apoptosis, migration, and impairment of angiogenesis via PI3K/AKT-eNOS signalling in HUVECs. Mediators Inflamm. 2019; 2019: 6168340, CrossRef.
Dallaglio K, Bruno A, Cantelmo AR, Esposito AI, Ruggiero L, Orecchioni S, et al. Paradoxic effects of metformin on endothelial cells and angiogenesis. Carcinogenesis. 2014; 35(5): 1055-66, CrossRef.
Barinda AJ, Hardi H, Louisa M, Khatimah NG, Marliau RM, Felix I, et al. Repurposing effect of cardiovascular-metabolic drug to increase lifespan: A systematic review of animal studies and current clinical trial progress. Front Pharmacol. 2024; 15: 1373458, CrossRef.
Yi G, He Z, Zhou X, Xian L, Yuan T, Jia X, et al. Low concentration of metformin induces a p53-dependent senescence in hepatoma cells via activation of the AMPK pathway. Int J Oncol. 2013; 43(5): 1503-10, CrossRef.
Takahashi A, Kimura F, Yamanaka A, Takebayashi A, Kita N, Takahashi L, et al. Metformin impairs growth of endometrial cancer cells via cell cycle arrest and concomitant autophagy and apoptosis. Cancer Cell Int. 2014; 14(1): 53, CrossRef.
DOI: https://doi.org/10.18585/inabj.v16i4.3067
Copyright (c) 2024 The Prodia Education and Research Institute
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
Indexed by:
The Prodia Education and Research Institute