GC-MS SCRUTINY OF PHYTOCONSTITUENTS, IN VITRO EVALUATION OF ANTIHYPERGLYCEMIC, ANTIADIPOGENIC ACTIVITIES, AND CYTOTOXIC EFFECT USING 3T3 L1 ADIPOCYTE CELL LINE AND MOLECULAR DOCKING STUDIES OF PREMNA CORYMBOSA

Authors

  • RADHIKA S Department of Biochemistry, Soka Ikeda College of Arts and Science for Women, Affiliated to University of Madras, Chennai, Tamil Nadu, India.
  • SENTHILKUMAR R Department of Biochemistry, Thanthai Periyar Government Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.

DOI:

https://doi.org/10.22159/ajpcr.2024v17i10.51986

Keywords:

Antihyperglycemic activity, Premna corymbosa, Phytochemicals, 3T3 L1 adipocyte Cell line, In silico molecular docking

Abstract

Objective: The present study was intended to list out the phytochemical multiples and to investigate the antihyperglycemic effect of Premna corymbosa using in vitro assays and in silico molecular docking methods.

Methods: The phytochemical multiples of methanol proportion of P. corymbosa leaves were appraised by Gas Chromatography-Mass Spectrometry (GC-MS) scrutiny to illustrate the attendance of phytochemical composites. Moreover, the in vitro antihyperglycemic, antiadipogenic activities, and cytotoxic effects of the extract were elucidated using a 3T3 L1 adipocyte cell line. Mode of action of phytochemical composites in methanol leaf extract of P. corymbosa was probed by Western blotting with IRS1, IRS2, mTOR, and glucose transporter type 4 (GLUT 4) receptors. At present, to probe the consequence of the aboriginal drugs, it is necessary to perform in silico docking on the diabetic receptor which could be useful for the progress of enhanced formulation for the psychoanalysis of diabetes.

Results: The GC-MS scrutiny depicted the being there of thirty-five phytochemical multipart. Amid the thirty-five multipart’s recognized, focal composites were Phytol, acetate (RT-16.78), n-Hexadecanoic acid (RT-18.16), Phytol (RT-19.51), 9,12,15-octadecatrienoic acid (Z,Z,Z) (RT-19.85), octadecanoic acid (RT-20.04), and Bis (2-ethylhexyl) phthalate (RT-23.09). The results of the glucose conception assay, adipocyte differentiation assay, and MTT assay showed potent in vitro antihyperglycemic activity with methanol leaf extract of P. corymbosa in 3T3l1Cell line. The results attained from western blotting revealed good antihyperglycemic activity of P. corymbosa. The in silico molecular docking results illustrated that the selected herbal lead compound is an effective target against the receptors. The compound showed favorable interactions with the amino acid residues thereby substantiating their proven efficacy as an antihyperglycemic compound.

Conclusion: The outcome of the current study substantiates the antihyperglycemic prospective of the methanol leaf extract of P. corymbosa on the hyperglycemic causal agents and its activity against diabetes by a molecular approach.

Downloads

Download data is not yet available.

References

Danaei G, Finucane MM, Lu Y, Singh GM, Cowan MJ, Paciorek CJ, et al. National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: Systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2.7 million participants. Lancet. 2011;378 (9785):31-40. doi: 10.1016/S0140-6736(11)60679-X, PMID: 21705069.

World Health Organization. WHO Report, Technical Report WHO/ EDM/TRM/2002. Vol. 21. Geneva, Switzerland: World Health Organization; 2002. p. 19-21.

Krishnamoorthy K, Krishnaswamy T, Subramaniam P, Sellamuthu M. Quantification of phytochemicals and in vitro antioxidant potential of various solvent extract of certain species of Acanthaceae. Int J Green Pharm. 2014;8:58-64.

Kaushik P, Lal S, Rana AC, Kaushik D. GC-MS analysis of bioactive constituents of Pinus roxburghii Sarg. (Pinaceae) from Northern India. Res J Phytochem. 2014;8(2):42-6. doi: 10.3923/rjphyto.2014.42.46

Alese MO, Adewole OS, Ljomone OM, Ajayi SA, Alese OO. Hypoglycemic and hypolipidemic activities of methanolic extract of Sphenocentrum jollyanum on streptozotocin-induced diabetic Wister rats. Eur J Med Plants. 2014;4(3):353-64. doi: 10.9734/ EJMP/2014/7618

Peteros NP. Antioxidant and cytotoxic activities and phytochemical screening of four Phillppine medicinal plants. J Med Plants Res. 2010;4(5):407-14.

Warjeet Singh L. Traditional medicinal plants of Manipur as anti-diabetics. J Med Plants Res. 2011;5:677-68.

Savithramma N, Linga Rao M, Prabha B. Phytochemical studies of Dysophylla myosuroides (Roth.) Benth. In: Wall. and Talinum cuneifolium (Vahl.) Willd. Res J Phytochem. 2011;5(3):163-9.

Karthishwaran K, Muthukkumarasamy S, Sankaran M. GCMS analysis of methanolic extract of aerial parts of Pergularia daemia. J Life Sci. 2012;1(1):50-5.

Murugan DD, Balan D, Wong PF. Adipogenesis and therapeutic potentials of antiobesogenic phytochemicals: Insights from preclinical studies. Phytother Res. 2021;35(11):5936-60. doi: 10.1002/ptr.7205, PMID 34219306

Sagbo IJ, van de Venter M, Koekemoer T, Bradley G. In vitro antidiabetic activity and mechanism of action of Brachylaena elliptica (Thunb.) DC. Evid Based Complement Alternat Med. 2018;2018:4170372. doi: 10.1155/2018/4170372

Alessi DR, Downes CP. The role of PI 3-kinase in insulin action. Biochimic Biophys Acta. 1998;1436(1-2):151-64. doi: 10.1016/s0005- 2760(98)00133-7, PMID 9838087

Ida Y, Watanabe M, Ohguro H, Hikage F. Simultaneous use of ROCK inhibitors and EP2 agonists induces unexpected effects on adipogenesis and the physical properties of 3T3-L1 preadipocytes. Int J Mol Sci. 2021;22(9):4648. doi: 10.3390/ijms22094648, PMID 33925005

Leney SE, Tavareé JM. The molecular basis of insulin-stimulated glucose uptake: Signalling, trafficking and potential drug targets. J Endocrinol. 2009;203(1):1-18. doi: 10.1677/JOE-09-0037, PMID 19389739

Lizák B, Szarka A, Kim Y, Choi KS, Németh CE, Marcolongo P, et al. Glucose transport and transporters in the endo membranes. Int J Mol Sci. 2019;20(23):5898. doi: 10.3390/ijms20235898, PMID 31771288

Pereira MJ, Palming J, Rizell M, Aureliano M, Carvalho E, Svensson MK, et al. mTOR inhibition with rapamycin causes impaired insulin signalling and glucose uptake in human subcutaneous and omental adipocytes. Mol Cell Endocrinol. 2012;355(1):96-105. doi: 10.1016/j.mce.2012.01.024, PMID 22333157

Sasaki-Suzuki N, Arai K, Ogata T, Kasahara K, Sakoda H., Chida K et al. Growth hormone inhibition of glucose uptake in adipocytes occurs without affecting GLUT4 translocation through an insulin receptor substrate-2-phosphatidylinositol 3-kinase-dependent pathway. J Biol Chem. 2009;284(10):6061-70. doi: 10.1074/jbc.M808282200, PMID 19122000

Chadt A, Al-Hasani H. Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease. Pflugers Arch. 2020;472(9):1273-1298. doi: 10.1007/s00424-020-02417-x, PMID 32591906

Hajalsiddig TT, Osman AB, Saeed AE. 2D-QSAR modeling and molecular docking studies on 1H-Pyrazole-1-carbothioamide derivatives as EGFR kinase inhibitors. ACS Omega. 2020;5(30):18662- 74. doi: 10.1021/acsomega.0c01323, PMID 32775868

Anthony J, Rangamaran VR, Shiva Sankarasubbiah KT, Gopal D, Ramalingam K. Applications of molecular docking: Its impact and importance outside the purview of drug discovery. In: Dastmalchi S, Hamzeh-Mivehroud M, Sokouti B, editors. Applied Case Studies and Solutions in Molecular Docking-based Drug Design. Pennsylvania: IGI Global; 2016. p. 278-306.

Xiang M, Cao Y, Fan W, Chen L, Mo Y. Computer-aided drug design: Lead discovery and optimization. Comb Chem High Throughput Screen. 2012;15(4):328-37. doi: 10.2174/138620712799361825, PMID 22221065

Dar AM, Mir S. Molecular docking: Approaches, types, applications and basic challenges. J Anal Bioanal Tech. 2017;8(2):356. doi: 10.4172/2155-9872.1000356

Dnyandev KM, Babasaheb GV, Chandrashekhar KV, Chandrakant MA, Vasant OK. A review on molecular docking. Int Res J Pure Appl Chem. 2021;22(3):60-8.

Gouthami K, Veeraraghavan V, Saratale GD, Rahdar A, Bilal M, Shah A, et al. Molecular Docking Used as an Advanced Tool to Determine Novel Compounds on Emerging Infectious Diseases: A Systematic Review. Research Square [Preprint]; 2021. doi: 10.21203/rs.3.rs-319858/v1

Kashikar VS, Tejaswita K. Indigenous remedies for diabetes mellitus. Int J Pharm Pharm Sci. 2011;3(3):22-9.

Kapoor LD. CRC Handbook of Ayurvedic Medicinal Plants, Herbal Reference Library. Vol. 70. Boca Raton London, New York Washington; CRC Press; 2001. p. 70.

Kathirvel A, Sujatha V. Phytochemical analysis and antioxidant activity of Barringtonia acutangula (L.) Gaetn. Leaves. Int J Pharm Pharm Sci. 2013;5(4):291-5.

Kanthal LK, Dey A, Satyavathi K, Bhojaraju P. GC-MS analysis of bio-active compounds in methanolic extract of Lactuca runcinata DC. Pharmacognosy Res. 2014;6(1):58-61. doi: 10.4103/0974- 8490.122919, PMID 24497744

Shemer J, Raizadag MK, Masters BA, Ota A, LeRoith D. Insulin-like growth factor I receptors in neuronal and glial cells - characterization and biological effects in primary culture. J Biol Chem. 1987;262(16):7693- 9. doi: 10.1016/S0021-9258(18)47623-5, PMID 2953724

Madsen L, Pedersen LM, Liaset B, Ma T, Petersen RK, van den Berg S, et al. CAMP-dependent signaling regulates the adipogenic effect of n-6 polyunsaturated fatty acids. J Biol Chem. 2008;283(11):7196-7205. doi: 10.1074/jbc.M707775200, PMID 18070879

Jung DW, Ha HH, Zheng X, Chang YT, Williams DR. Novel use of fluorescent glucose analogues to identify a new class of triazine-based insulin mimetics possessing useful secondary effects. Mol Biosyst. 2011;7(2):346-58. doi: 10.1039/c0mb00089b, PMID 20927436

Slater TF, Sawyer FB, Strauli U. Studies on succinate tetrazolium reductase systems. III. Points of coupling of four different tetrazolium salts. Biochim Biophys Acta. 1963;77:383-93. doi: 10.1016/0006- 3002(63)90513-4, PMID 14089413

Wright HM, Clish CB, Mikami T, Hauser S, Yanagi K, Hiramatsu R, et al. A synthetic antagonist for the peroxisome proliferator-activated receptor gamma inhibits adipocyte differentiation. J Biol Chem. 2000;275(3):1873-7. doi: 10.1074/jbc.275.3.1873, PMID 10636887

Liu LZ, Zhao HL, Zuo J, Ho SK, Chan JC, Meng Y, et al. Protein kinase Czeta mediates insulin-induced glucose transport through actin remodeling in L6 muscle cells. Mol Biol Cell. 2006;17(5):2322-30. doi: 10.1091/mbc.e05-10-0969, PMID 16525020

Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976 May 7;72:248-54. doi: 10.1006/ abio.1976.9999, PMID 942051

Pass GJ, Becker W, Kluge R, Linnartz K, Plum L, Giesen K, et al. Effect of hyperinsulinemia and type II diabetes-like hyperglycemia on expression of hepatic cytochrome p450 and glutathione s-transferase isoforms in a New Zealand obese-derived mouse backcross population. J Pharmacol Exp Ther. 2002;302(2):442-50.

Lalitharani S, Mohan VR, Regini GS, Kalidass C. GC-MS analysis of ethanolic extracts of Pothos scandens L. J Herb Med Toxicol. 2009;3:159-60.

Kala SM, Tresina PS, Mohan VR. Hepatoprotective effect of Eugenia singampattiana Bedd leaf extract on carbon tetrachloride induced jaundice. Int J Pharm Sci Rev Res. 2013;21(1):41-5.

Rao CV, Newmark HL, Reddy BS. Chemopreventive effect of squalene on colon cancer. Carcinogenesis. 1998;19(2):287-90. doi: 10.1093/ carcin/19.2.287, PMID 9498278

Alagammal M, Tresina PS, Mohan VR. GC-MS Determination of bioactive components of Polygala javana DC. Int J Curr Pharm Res. 2012;4(2):42-4.

Sathyaprabha G, Kumaravel S, Ruffina D, Praveen Kumar P. A comparative study on antioxidant, proximate analysis, antimicrobial activity and phytochemical analysis of Aloe vera and Cissus quadrangularis by GC-MS. J Pharm Res. 2010;3(12):2970-3.

Jones PJ. Clinical nutrition: 7. Functional foods - more than just nutrition. CMAJ. 2002;166(12):1555-63. PMID 12074125

Jegadeeswari P, Nishanthini A, Muthukumarasamy S, Mohan VR. GC-MS analysis of bioactive components of Aristolochia krysagathra (Aristolochiaceae). J Curr Chem Pharm Sci. 2012;2(4):226-32.

Muller DP. Vitamin E and neurological function- review. Mol Nutr Food Res. 2010;54(5):710-8. doi: 10.1002/mnfr.200900460, PMID 20183831

Whitney EN, Rolfes SR, editors. Understanding Nutrition. 12th ed. Wadsworth, California: Cengage Learning; 2011.

Balamurugan R, Stalin A, Ignacimuthu S. Molecular docking of gamma-sitosterol with some target related to diabetes. Eur J Med Chem. 2012;47(1):3938-43. doi: 10.1016/j.ejmech.2011.10.007, PMID 22078765

Asokan A, Thangavel M. In-vitro cytotoxic studies of crude methanolic extract of Saraca indica bark extract. IOSR J Pharm Biol Sci. 2014;9(4):26-30. doi: 10.9790/3008-09412630

Prathapan A, Krishna MS, Lekshmi PC, Raghu KG, Menon NA. Modulation of adipogenesis and glucose uptake by Curcuma longa extract in 3T3L1 and L6 cell lines-an in vitro study. Asian Pac J Trop Dis. 2012;2(Suppl 1):S163-5. doi: 10.1016/S2222-1808(12)60144-3

Arumughan M, Vijayan P, Raghu C, Ashok G, Dhanraj SA, Kumarappan CT. Antiadipogenic activity of Capsicum annum (Solanaceae) in 3T3L1. J Complement Integr Med. 2008;5(1):1-9.

Schordinger Suite Induced Fit Docking Protocol, Glide Version 5.6; Prime version 2.2. New York: Schrodinger LLC; 2010.

Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, et al. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidyl inositol 3-kinase activity in muscle. J Biol Chem. 2002;277(52):50230-6. doi: 10.1074/jbc. M200958200, PMID 12006582

Storz P, Doöppler H, Wernig A, Pfizenmaier K, Muüller G. Cross-talk mechanisms in the development of insulin resistance of skeletal muscle cells palmitate rather than tumour necrosis factor inhibits insulin-dependent protein kinase B (PKB)/Akt stimulation and glucose uptake. Eur J Biochem. 1999;266(1):17-25. doi: 10.1046/j.1432- 1327.1999.00809.x, PMID 10542046

Sakaue HW, Ogawa MW, Matsumoto SM, Kuroda MS, Takata TM, Sugimoto BM, et al. Posttranscriptional control of adipocyte differentiation through activation of phosphoinositide 3-kinase. J Biol Chem. 1998;273(44):28945-52. doi: 10.1074/jbc.273.44.28945, PMID 9786898

Barak Y, Nelson MC, Ong ES, Jones YZ, Ruize-Lozano P, Chien KR, et al. PPAR gamma is required for placental, cardiac, and adipose tissue development. Mol Cell. 1999;4(4):585-95. doi: 10.1016/s1097- 2765(00)80209-9, PMID 10549290

Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, et al. PPAR gamma mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance. Mol Cell. 1999;4(4):597-609. doi: 10.1016/s1097-2765(00)80210-5, PMID 10549291

Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, et al. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell. 1999;4(4):611-7. doi: 10.1016/s1097- 2765(00)80211-7, PMID 10549292

Tanaka T, Yoshida N, Kishimoto T, Akira S. Defective adipocyte differentiation in mice lacking the C/EBPb and C/EBPd gene. EMBO J. 1997;16(24):7432-43. doi: 10.1093/emboj/16.24.7432, PMID 9405372

Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17(4):448-53. doi: 10.1038/nm.2307, PMID 21423183

Wu Z, Rosen ED, Brun R, Hauser S, Adelmant G, Troy AE, et al. Cross regulation of C/EBP alpha and PPAR gamma controls the transcriptional pathway of adipogenesis and insulin sensitivity. Mol Cell. 1999;3(2):151-8. doi: 10.1016/s1097-2765(00)80306-8, PMID 10078198

Fasshauer M, Klein J, Ueki K, Kriauciunas KM, Benito M, White MF, et al. Essential role of insulin receptor substrate-2 in insulin stimulation of Glut4 translocation and glucose uptake in brown adipocytes. J Biol Chem. 2000;275(33):25494-501. doi: 10.1074/jbc.M004046200, PMID 10829031

Cao Z, Umek RM, McKnight SL. Regulated expression of three C/EBP isoforms during adipocyte conversion of 3T3-L1 cells. Genes Dev. 1991;5(9):1538-52. doi: 10.1101/gad.5.9.1538, PMID 1840554

Wu Z, Bucher NL, Farmer SR. Induction of peroxisome proliferator-activated receptor g during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPb, C/EBPd and glucocorticoid. Mol Cell Biol. 1996;16(8):4128-36. doi: 10.1128/MCB.16.8.4128, PMID 8754811

Kubinyi H. Combinatorial and computational approaches in structure-based drug design. Curr Opin Drg Discov Dev 1998;1:16-27.

Girija CR, Karunakar P, Poojari CS, Begum NS, Syed AA. Molecular docking studies of curcumin derivatives with multiple protein targets for pro carcinogen activating enzyme inhibition. J Proteomics Bioinform. 2010;3(6):200-3.

Published

07-10-2024

How to Cite

RADHIKA S, and SENTHILKUMAR R. “GC-MS SCRUTINY OF PHYTOCONSTITUENTS, IN VITRO EVALUATION OF ANTIHYPERGLYCEMIC, ANTIADIPOGENIC ACTIVITIES, AND CYTOTOXIC EFFECT USING 3T3 L1 ADIPOCYTE CELL LINE AND MOLECULAR DOCKING STUDIES OF PREMNA CORYMBOSA”. Asian Journal of Pharmaceutical and Clinical Research, vol. 17, no. 10, Oct. 2024, pp. 106-1, doi:10.22159/ajpcr.2024v17i10.51986.

Issue

Section

Original Article(s)