131
Views
13
CrossRef citations to date
0
Altmetric
Original Research

Sterculia tragacantha Lindl Leaf Extract Ameliorates STZ-Induced Diabetes, Oxidative Stress, Inflammation and Neuronal Impairment

ORCID Icon, ORCID Icon, , , , , ORCID Icon, , , , & ORCID Icon show all
Pages 6749-6764 | Published online: 09 Dec 2021

References

  • Bilous R, Donnelly R, Idris I. Handbook of Diabetes. John Wiley & Sons; 2021.
  • Saeedi P, Petersohn I, Salpea P, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas. Diabetes Res Clin Pract. 2019;157:107843. doi:10.1016/j.diabres.2019.107843
  • Deng Y, Li N, Wu Y, et al. Global, regional, and national burden of diabetes-related chronic kidney disease from 1990 to 2019. Front Endocrinol (Lausanne). 2021;12:809. doi:10.3389/fendo.2021.672350
  • Darenskaya M, Kolesnikova L, Kolesnikov S. Oxidative stress: pathogenetic role in diabetes mellitus and its complications and therapeutic approaches to correction. Bull Exp Biol Med. 2021;171(2):179–189. doi:10.1007/s10517-021-05191-7
  • Park S, Park S-Y. Can antioxidants be effective therapeutics for type 2 diabetes? Yeungnam Univ J Med. 2021;38(2):83. doi:10.12701/yujm.2020.00563
  • Agada R, Usman WA, Shehu S, Thagariki D. In vitro and in vivo inhibitory effects of Carica papaya seed on α-amylase and α-glucosidase enzymes. Heliyon. 2020;6(3):e03618. doi:10.1016/j.heliyon.2020.e03618
  • Hamed SA. Brain injury with diabetes mellitus: evidence, mechanisms and treatment implications. Expert Rev Clin Pharmacol. 2017;10(4):409–428. doi:10.1080/17512433.2017.1293521
  • Jin HY, Moon -S-S, Calcutt NA. Lost in translation? Measuring diabetic neuropathy in humans and animals. Diabetes Metab J. 2021;45(1):27–42. doi:10.4093/dmj.2020.0216
  • Azmi S, Alam U, Burgess J, Malik RA. State-of-the-art pharmacotherapy for diabetic neuropathy. Expert Opin Pharmacother. 2021;22(1):55–68. doi:10.1080/14656566.2020.1812578
  • Pohl F, Kong Thoo Lin P. The potential use of plant natural products and plant extracts with antioxidant properties for the prevention/treatment of neurodegenerative diseases: in vitro, in vivo and clinical trials. Molecules. 2018;23(12):3283. doi:10.3390/molecules23123283
  • Orhan I, Aslan S, Kartal M, Şener B, Başer KHC. Inhibitory effect of Turkish Rosmarinus officinalis L. on acetylcholinesterase and butyrylcholinesterase enzymes. Food Chem. 2008;108(2):663–668. doi:10.1016/j.foodchem.2007.11.023
  • Shim Y-J, Doo H-K, Ahn S-Y, et al. Inhibitory effect of aqueous extract from the gall of Rhus chinensis on alpha-glucosidase activity and postprandial blood glucose. J Ethnopharmacol. 2003;85(2–3):283–287. doi:10.1016/S0378-8741(02)00370-7
  • Bashir L, Shittu O, Sani S, Busari M, Adeniyi K. African natural products with potential antitrypanosomal properties: a review. Int J Biochem Res Rev. 2015;7(2):45–79. doi:10.9734/IJBCRR/2015/16039
  • Lawal B, Shittu OK, Kabiru AY, et al. Potential antimalarials from African natural products: a review. J Intercult Ethnopharmacol. 2015;4(4):318. doi:10.5455/jice.20150928102856
  • Lawal B, Shittu OK, Oibiokpa FI, Berinyuy EB, Mohammed H. African natural products with potential antioxidants and hepatoprotectives properties: a review. Clin Phytosci. 2017;2(1):1–66.
  • Ndako M, Jigam AA, Kabiru AY, Umar SI, Lawal B. Polar extracts from Gymnosporia senegalensis (syn. Maytenus senegalensis) root bark, its effects on nociception, edema, and malarial infection. Phytomed Plus. 2021;1(4):100113. doi:10.1016/j.phyplu.2021.100113
  • Zdrowia ŚO; Organization WH. World Report on Knowledge for Better Health: Strengthening Health Systems. World Health Organization; 2004.
  • Sadeer NB, Llorent-Martínez EJ, Bene K, et al. Chemical profiling, antioxidant, enzyme inhibitory and molecular modelling studies on the leaves and stem bark extracts of three African medicinal plants. J Pharm Biomed Anal. 2019;174:19–33. doi:10.1016/j.jpba.2019.05.041
  • Udegbunam R, Asuzu U, Kene R, Udegbunam S, Nwaehujor C. Anti-nociceptive, anti-inflammatory and anti-oxidant effects of the methanol leaf extract of Sterculia tragacantha Lindl. J Pharmacol Toxicol. 2011;6(5):516–524. doi:10.3923/jpt.2011.516.524
  • Mogbojuri OM, Adedapo AA, Abatan MO. Phytochemical screening, safety evaluation, anti-inflammatory and analgesic studies of the leaf extracts of Sterculia tragacantha. J Complement Integr Med. 2016;13(3):221–228. doi:10.1515/jcim-2015-0114
  • Ajiboye BO, Oyinloye BE, Awurum JC, Onikanni SA, Adefolalu A, Oluba OM. Protective role of Sterculia tragacantha aqueous extract on pancreatic gene expression and oxidative stress parameters in streptozotocin-induced diabetic rats. J Complement Integr Med. 2021. doi:10.1515/jcim-2021-0020
  • Udegbunam RI, Asuzu UI, Kene RO, Oyiga CT, Udegbunam SO, Nwaehujor CC. Anti-inflammatory and anti-oxidant effects of Sterculia tragacantha fractions in mice. Afr J Biotechnol. 2013;12:6.
  • Ajiboye BO, Oyinloye BE, Essien PE, Onikanni SA, Ojo OA, Kappo AP. Ameliorative potential of Sterculia tragacantha aqueous extract on renal gene expression and biochemical parameters in streptozotocin-induced diabetic rats. J Pharm Invest. 2021;51(1):103–113. doi:10.1007/s40005-020-00506-8
  • Jerome SZ, Benson BB, Foungoye OA, M-bj A, Yves-Alain B. Analysis by GC (Ir), GC/MS and mosquito repellent effect of essential oils against Anopheles gambiae: case of stem bark of Sterculia tragacantha Lindl (Sterculiaceae) from Côte d'Ivoire. J Med Plants Res. 2021;15(7):309–320. doi:10.5897/JMPR2021.7132
  • Palve A, Shetty P, Pimpliskar M, Jadhav R. Study on antibacterial and antifungal activities of Sterculia lychnophora extracts [J]. Int J Curr Microbiol Appl Sci. 2015;4(11):336–341.
  • Prista LN, Alves AC, Da Costa M. A pharmacological study of Sterculia tragacantha. Garcia de Orta. 1960;8:67–80.
  • Sofowora A. Research on medicinal plants and traditional medicine in Africa. J Altern Complement Med. 1996;2(3):365–372. doi:10.1089/acm.1996.2.365
  • Hagerman AE, Riedl KM, Jones Ga, et al. High molecular weight plant polyphenolics (Tannins) as biological antioxidants. J Agric Food Chem. 1998;46(5):1887–1892. doi:10.1021/jf970975b
  • Ghorai N, Chakraborty S, Gucchait S, Saha SK, Biswas S. Estimation of Total Terpenoids Concentration in Plant Tissues Using a Monoterpene. Linalool as standard reagent; 2012.
  • Tsado N, Lawal B, Ossa P, et al. Antioxidants and antimicrobial activities of methanol extract of Newbouldia laevis and Crateva adansonii. J Pharm Allied Health Sci. 2016;6:14–19. doi:10.3923/jpahs.2016.14.19
  • Hajimahmoodi M. Antioxidant activity, reducing power and total phenolic content of Iranian olive cultivar. 第十四届世界食品科技大会. 2008;8:676.
  • Kasangana PB, Haddad PS, Stevanovic T. Study of polyphenol content and antioxidant capacity of Myrianthus arboreus (Cecropiaceae) root bark extracts. Antioxidants. 2015;4(2):410–426. doi:10.3390/antiox4020410
  • Worthington K. Alpha Amylase Worthington Enzyme Manual. Lakewood, NJ: Worthington Biochemical Corporation[Google Scholar]; 1993:36–41.
  • Apostolidis E, Y-i K, Shetty K. Inhibitory potential of herb, fruit, and fungal-enriched cheese against key enzymes linked to type 2 diabetes and hypertension. Innovat Food Sci Emerg Technol. 2007;8(1):46–54. doi:10.1016/j.ifset.2006.06.001
  • Abatan M, Mogbojuri O. Evaluation of the anti-inflammatory and analgesic activities of leaf-extracts of Sterculia tragacantha in rats. FASEB J. 2015;29(S1):616.615. doi:10.1096/fasebj.29.1_supplement.616.5
  • Kakkar P, Das B, Viswanathan P. A modified spectrophotometric assay of superoxide dismutase. Indian J Biochem Biophys. 1984;21(2):130–132.
  • Sinha AK. Colorimetric assay of catalase. Anal Biochem. 1972;47(2):389–394. doi:10.1016/0003-2697(72)90132-7
  • Rotruck JT, Pope AL, Ganther HE, Swanson A, Hafeman DG, Hoekstra W. Selenium: biochemical role as a component of glutathione peroxidase. Science. 1973;179(4073):588–590. doi:10.1126/science.179.4073.588
  • Kum-Tatt L, Tan I-K. A new colorimetric method for the determination of glutathione in erythrocytes. Clinica Chimica Acta. 1974;53(2):153–161. doi:10.1016/0009-8981(74)90093-X
  • Shagirtha K, Bashir N, MiltonPrabu S. Neuroprotective efficacy of hesperetin against cadmium induced oxidative stress in the brain of rats. Toxicol Ind Health. 2017;33(5):454–468. doi:10.1177/0748233716665301
  • Shimizu T, Kondo K, Hayaishi O. Role of prostaglandin endoperoxides in the serum thiobarbituric acid reaction. Arch Biochem Biophys. 1981;206(2):271–276. doi:10.1016/0003-9861(81)90091-6
  • Miranda HF, Puig MM, Prieto JC, Pinardi G. Synergism between paracetamol and nonsteroidal anti-inflammatory drugs in experimental acute pain. Pain. 2006;121(1–2):22–28. doi:10.1016/j.pain.2005.11.012
  • Jońca J, Żuk M, Wasąg B, et al. New insights into butyrylcholinesterase activity assay: serum dilution factor as a crucial parameter. PLoS One. 2015;10(10):e0139480. doi:10.1371/journal.pone.0139480
  • Pagel P, Blome J, Wolf HU. High-performance liquid chromatographic separation and measurement of various biogenic compounds possibly involved in the pathomechanism of Parkinson’s disease. J Chromatogr B Biomed Sci Appl. 2000;746(2):297–304. doi:10.1016/S0378-4347(00)00348-0
  • Shyjan AW, Cena V, Klein DC, Levenson R. Differential expression and enzymatic properties of the Na+, K (+)-ATPase alpha 3 isoenzyme in rat pineal glands. Proc Natl Acad Sci. 1990;87(3):1178–1182. doi:10.1073/pnas.87.3.1178
  • Marcus D, Hanwell DEC, Lonie DC, et al. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. J Cheminform. 2012;4:17.
  • Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31(2):455–461. doi:10.1002/jcc.21334
  • Lawal B, Liu Y-L, Mokgautsi N, et al. Pharmacoinformatics and preclinical studies of NSC765690 and NSC765599, potential STAT3/CDK2/4/6 inhibitors with antitumor activities against NCI60 human tumor cell lines. Biomedicines. 2021;9(1):92. doi:10.3390/biomedicines9010092
  • Wu S-Y, Lin K-C, Lawal B, Wu ATH, Wu C-Z. MXD3 as an onco-immunological biomarker encompassing the tumor microenvironment, disease staging, prognoses, and therapeutic responses in multiple cancer types. Comput Struct Biotechnol J. 2021;19:4970–4983. doi:10.1016/j.csbj.2021.08.047
  • Lawal B, Lee C-Y, Mokgautsi N, et al. mTOR/EGFR/iNOS/MAP2K1/FGFR/TGFB1 are druggable candidates for N-(2,4-difluorophenyl)-2′,4′-difluoro-4-hydroxybiphenyl-3-carboxamide (NSC765598), with consequent anticancer implications. Front Oncol. 2021;11:932. doi:10.3389/fonc.2021.656738
  • Olugbodi JO, Samaila K, Lawal B, et al. Computational and preclinical evidence of anti-ischemic properties of l-carnitine-rich supplement via stimulation of anti-inflammatory and antioxidant events in testicular torsed rats. Oxid Med Cell Longev. 2021;2021:5543340. doi:10.1155/2021/5543340
  • Visualizer DS. BIOVIA, Dassault Systèmes, BIOVIA Workbook, Release 2020; BIOVIA Pipeline Pilot, Release 2020. San Diego: Dassault Systèmes; 2020.
  • Orhan I, Kartal M, Tosun F, Şener B. Screening of various phenolic acids and flavonoid derivatives for their anticholinesterase potential. Zeitschrift für Naturforschung C. 2007;62(11–12):829–832. doi:10.1515/znc-2007-11-1210
  • Inácio Lunkes G, Stefanello F, Sausen Lunkes D, Maria Morsch V, Schetinger MRC, Gonçalves JF. Serum cholinesterase activity in diabetes and associated pathologies. Diabetes Res Clin Pract. 2006;72(1):28–32. doi:10.1016/j.diabres.2005.08.009
  • Pari L, Murugavel P. Diallyl tetrasulfide improves cadmium induced alterations of acetylcholinesterase, ATPases and oxidative stress in brain of rats. Toxicology. 2007;234(1–2):44–50. doi:10.1016/j.tox.2007.01.021
  • Papoutsis K, Zhang J, Bowyer MC, Brunton N, Gibney ER, Lyng J. Fruit, vegetables, and mushrooms for the preparation of extracts with α-amylase and α-glucosidase inhibition properties: a review. Food Chem. 2021;338:128119. doi:10.1016/j.foodchem.2020.128119
  • Asayama K, Nakane T, Uchida N, Hayashibe H, Dobashi K, Nakazawa S. Serum antioxidant status in streptozotocin-induced diabetic rat. Hormone Metabol Res. 1994;26(07):313–315. doi:10.1055/s-2007-1001693
  • Garg MC, Ojha S, Bansal D. Antioxidant status of streptozotocin diabetic rats. Indian J Exp Biol. 1996;34(3):264–266.
  • Paul A, Anandabaskar N, Mathaiyan J, Raj GM, editors. Introduction to Basics of Pharmacology and Toxicology. Volume 2 : Essentials of Systemic Pharmacology: From Principles to Practice. Singapore: Springer Nature Singapore Pte Ltd; 2021. doi:10.1007/978-981-33-6009-9S
  • Ramakrishnan R, Namasivayam A. Norepinephrine and epinephrine levels in the brain of alloxan diabetic rats. Neurosci Lett. 1995;186(2–3):200–202. doi:10.1016/0304-3940(95)11315-N
  • Chen Y, Xu J, Chen Y. Regulation of neurotransmitters by the gut microbiota and effects on cognition in neurological disorders. Nutrients. 2021;13(6):2099. doi:10.3390/nu13062099
  • Okesola MA, Ajiboye BO, Oyinloye BE, et al. Effect of Solanum macrocarpon Linn leaf aqueous extract on the brain of an alloxan-induced rat model of diabetes. J Int Med Res. 2020;48(6):300060520922649. doi:10.1177/0300060520922649
  • Bagi Z, Erdei N, Papp Z, Édes I, Koller A. Up-regulation of vascular cyclooxygenase-2 in diabetes mellitus. Pharmacol Rep. 2006;58:52.
  • Pop-Busui R, Kellogg AP, Cheng HT. Cyclooxygenase-2 pathway as a potential therapeutic target in diabetic peripheral neuropathy. Curr Drug Targets. 2008;9(1):68–76. doi:10.2174/138945008783431691
  • Lees JA, Saito M, Vidal M, et al. The retinoblastoma protein binds to a family of E2F transcription factors. Mol Cell Biol. 1993;13(12):7813–7825. doi:10.1128/mcb.13.12.7813-7825.1993
  • Wrzos HF, Cruz A, Polavarapu R, Shearer D, Ouyang A. Nitric oxide synthase (NOS) expression in the myenteric plexus of streptozotocin-diabetic rats. Dig Dis Sci. 1997;42(10):2106–2110. doi:10.1023/A:1018830820537
  • Babu PVA, Sabitha KE, Shyamaladevi CS. Green tea impedes dyslipidemia, lipid peroxidation, protein glycation and ameliorates Ca2+-ATPase and Na+/K+-ATPase activity in the heart of streptozotocin-diabetic rats. Chem Biol Interact. 2006;162(2):157–164. doi:10.1016/j.cbi.2006.05.020
  • Ajayi AM, Adedapo AD, Badaki VB, Oyagbemi AA, Adedapo AA. Chrysophyllum albidum fruit ethanol extract ameliorates hyperglycaemia and elevated blood pressure in streptozotocin-induced diabetic rats through modulation of oxidative stress, NF-κB and PPAR-γ. Biomed Pharmacother. 2021;141:111879. doi:10.1016/j.biopha.2021.111879
  • Kunte KB, Kuna Y. Neuroprotective effect of Bacopa monniera on memory deficits and ATPase system in Alzheimer’s disease (AD) induced mice. J Sci Innov Res. 2013;2(4):719–735.
  • Slatter D, Bolton C, Bailey A. The importance of lipid-derived malondialdehyde in diabetes mellitus. Diabetologia. 2000;43(5):550–557. doi:10.1007/s001250051342
  • Oyinloye BE, Adenowo AF, Osunsanmi FO, Ogunyinka BI, Nwozo SO, Kappo AP. Aqueous extract of Monodora myristica ameliorates cadmium-induced hepatotoxicity in male rats. SpringerPlus. 2016;5(1):1–7. doi:10.1186/s40064-016-2228-z
  • Kitchen DB, Decornez H, Furr JR, Bajorath J. Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discov. 2004;3(11):935–949. doi:10.1038/nrd1549
  • Chen J-H, Wu ATH, Lawal B, et al. Identification of cancer hub gene signatures associated with immune-suppressive tumor microenvironment and ovatodiolide as a potential cancer immunotherapeutic agent. Cancers. 2021;13(15):3847. doi:10.3390/cancers13153847
  • Yeh Y-C, Lawal B, Hsiao M, Huang T-H, Huang C-YF. Identification of NSP3 (SH2D3C) as a prognostic biomarker of tumor progression and immune evasion for lung cancer and evaluation of organosulfur compounds from Allium sativum L. as therapeutic candidates. Biomedicines. 2021;9(11):1582. doi:10.3390/biomedicines9111582
  • Wu ATH, Lawal B, Wei L, Wen Y-T, Tzeng DTW, Lo W-C. Multiomics identification of potential targets for Alzheimer disease and antrocin as a therapeutic candidate. Pharmaceutics. 2021;13(10):1555. doi:10.3390/pharmaceutics13101555
  • Lawal B, Kuo Y-C, Tang S-L, et al. Transcriptomic-based identification of the immuno-oncogenic signature of cholangiocarcinoma for HLC-018 multi-target therapy exploration. Cells. 2021;10(11):2873. doi:10.3390/cells10112873
  • Lawal B, Kuo YC, Sumitra MR, Wu ATH, Huang HS. In vivo pharmacokinetic and anticancer studies of HH-N25, a selective inhibitor of Topoisomerase I, and hormonal signaling for treating breast cancer. J Inflamm Res. 2021;14:4901–4913. doi:10.2147/JIR.S329401