Glycemic and Biochemical Effects of Polyphenol-Rich Fraction of <i>Parkia biglobosa</i> Leaves in Wistar rats Experimentally Induced with Diabetes

http://www.doi.org/10.26538/tjnpr/v7i10.40

Authors

  • Obioma U. Njoku Department of Biochemistry, University of Nigeria, Nsukka.
  • Christian C. Chibuogwu Department of Biochemistry, University of Nigeria, Nsukka.
  • Okwesili F.C. Nwodo Department of Biochemistry, University of Nigeria, Nsukka.

Keywords:

Liver function, Renal function, glucose tolerance, polyphenol-rich, antioxidant, Parkia biglobosa

Abstract

This study evaluated the effect of polyphenol-rich fraction of Parkia biglobosa leaves (PBF) in rats experimentally induced with diabetes. Thirty albino rats divided into five groups (n=6) were used for this study. Except for group one, the rats were first maintained on high high-calorie diet for eight weeks followed by streptozotocin injection (40 mg/kg). Groups 1 and 2 (normal and diabetic controls respectively) received 1 ml of distilled water throughout the experiment. Group 3 received 50 mg/kg of glibenclamide while groups 4 and 5 received 200 and 400 mg/kg of PBF respectively. After 4 weeks of treatment, after an overnight fast, blood was collected from the rats for haematological and biochemical analyses. Body weight and fasting blood glucose measurements were taken weekly. The in vitro antioxidant analysis revealed good ferric reducing power, and DPPH radical-quenching activity (IC50 = 0.47mg/ml). The PBF groups had markedly (p<0.05) reduced blood glucose and showed better glucose tolerance compared to the untreated group. The PBF groups also had marked (p<0.05) diminution of liver enzymes’ activities (ALP, ALT, AST) as well as markedly reduced bilirubin levels relative to the diabetic control. Urea, creatinine, and K+ levels were markedly (p<0.05) reduced in the PBF groups relative to the untreated, while the PBF groups had markedly high Na+ levels in comparison to the diabetic control. This result justifies the use of the leaves locally for disease management and could serve as a cheaper and low-toxicity alternative in the management of diabetes and associated complications.

Author Biography

Christian C. Chibuogwu, Department of Biochemistry, University of Nigeria, Nsukka.

Institute for Drug-Herbal Medicine-Excipients Research and Development, University of Nigeria, Nsukka.

References

Singh A, Kukreti R, Saso L, Kukreti S. Pathways and Type 2 Diabetes. Mol. 2022; 27: 950–969.

Agyemang C, Meeks K, Beune E, Owusu-Dabo E, Mockenhaupt FP, Addo J. Obesity and type 2 diabetes in sub-Saharan Africans - Is the burden in today's Africa similar to African migrants in Europe? The RODAM study. BMC Med. 2016; 14(1): 166.

Godman B, Basu D, Pillay Y, Mwita JC, Rwegerera GM, Anand Paramadhas BD, Tiroyakgosi C, Okwen PM, Niba LL, Nonvignon J, Sefah I, Oluka M, Guantai AN, Kibuule D, Kalemeera F, Mubita M, Fadare J, Ogunleye OO, Distiller LA, Rampamba EM, Wing J, Mueller D, Alfadl A, Amu AA, Matsebula Z, Kalungia A, Zaranyika T, Masuka N, Wale J, Hill R, Kurdi A, Timoney A, Campbell S, Meyer J.C. Review of ongoing activities and challenges to improve the care of patients with Type 2 Diabetes across Africa and the implications for the future. Front. Pharm. 2020; 11: 108

Pastakia SD, Pekny CR, Manyara SM, Fischer L. Diabetes in sub-Saharan Africa - from policy to practice to progress: targeting the existing gaps for future care for diabetes. Diab Met Syn and Obes. 2017; 10: 247–263.

Atun R, Davies JI, Gale EAM, Barnighausen T, Beran D, Kengne AP. Diabetes in sub-Saharan Africa: from clinical care to health policy. Lanc Diab Endoc, 2017; 5(8): 622–667.

Mapa-Tassou C, Katte JC, Mba-Maadjhou C, Mbanya JC. Economic impact of diabetes in Africa. Curr Diab Rep. 2019; 19(2): 5.

Abbas G, Al Harrasi A, Hussain H, Hamaed A, Supuran CT. The management of diabetes mellitus-imperative role of natural products against dipeptidyl peptidase-4, α-glucosidase and sodium-dependent glucose co-transporter 2 (SGLT2). Bioorg Chem. 2019; 86: 305–315.

Koura K, Ganglo JC, Assogbadjo AE, Agbangla C. Ethnic differences in use values and use patterns of Parkia biglobosa in Northern Benin. J. Ethnobio and Ethnomed. 2015; 7: 42.

Nyadanu D, Amoah RA, Obeng B, Kwarteng A, Akromah R, Aboagye L, Adu-Dapaah H. Ethnobotany and analysis of food components of African locust bean (Parkia biglobosa (Jacq.) Benth.) in the transitional zone of Ghana: implications for domestication, conservation and breeding of improved varieties. Gen Res Crop Evol. 2017; 64: 1231–1240

Komolafe K, Akinmoladun AC, Komolafe TR, Olaleye MT, Akindahunsi AA, Rocha JBT. African locust bean (Parkia biglobosa, Jacq Benth) leaf extract affects mitochondrial redox chemistry and inhibits angiotensin-converting enzyme in vitro. Clin Phytosci. 2017; 3: 19.

Ekperikpe US, Owolabi OJ, Olapeju BI. Effects of Parkia biglobosa aqueous seed extract on some biochemical, haematological and histopathological parameters in streptozotocin-induced diabetic rats. J. Ethnopharm. 2018

https://doi.org/10.1016/j.jep.2018.09.016.

Ogunyinka BI, Oyinloye BE, Osunsanmi FO, Kolanisi U, Opoku AR, Kappo AP. Protein isolate from Parkia biglobosa seeds improves dyslipidaemia and cardiac oxidative stress in streptozotocin-induced diabetic rats. Antiox. 2019; 8: 481

Panche AN, Diwan AD, Chandra SR. Flavonoids: An overview. J Nutr Sc. 2016; 5: 1-15.

Fei J, Liang B, Jiang C, Ni H, Wang L. Luteolin inhibits IL-1β-induced inflammation in rat chondrocytes and attenuates osteoarthritis progression in a rat model. Biomed & Pharmacother 2019; 109: 1586–1592.

Chibuogwu CC, Asomadu RO, Okagu IU, Nkwocha CC, Amadi BC. "Attenuation of glycation and biochemical aberrations in fructose‐loaded rats by polyphenol‐rich ethyl acetate fraction of Parkia biglobosa (Jacq.) Benth. (Mimosaceae) leaves". Clin Phytosci, 2021; 7: 41

Lee SY, Mediani A, Ismail IS, Abas M, Abas F. Antioxidants and α-glucosidase inhibitors from Neptunia oleracea fractions using 1H NMR-based metabolomics approach and UHPLC-MS/MS analysis. BMC Compl and Alt Med. 2019; 19: 7

Alachaher FZ, Dali S, Dida N, Krouf D. Comparison of phytochemical and antioxidant properties of extracts from flaxseed (Linum usitatissimum) using different solvents. Inter Food Res J. 2018; 25(1): 75-82.

Poovitha S, Parani M. In vitro and in vivo α-amylase and α-glucosidase inhibiting activities of the protein extracts from two varieties of bitter gourd (Momordica charantia L.). BMC Complem and Alt Med. 2016; 16(1); 1-8.

Okoduwa SIR, Umar IA, James DB, Inuwa HM. Appropriate insulin level in selecting fortified diet-fed, streptozotocin-treated rat model of type 2 diabetes for anti-diabetic studies. PLoS ONE: 2017; 12(1): e0170971.

Reitman S, Frankel S. A colorimetric method for the determination of serum glutamic oxaloacetic and glutamic pyruvic transaminases. Am J Clin Path. 1957; 28: 56–63.

Jendrassik J, Grof P. In vitro determination of total and direct bilirubin in serum or plasma. Biochem. 1938; 6: 269–275

Cheesbrough M. Measurement of serum or plasma creatinine and urea. In: District Laboratory Practice in Tropical Countries, 2nd edn. Cambridge University Press, Cambridge. 2005; pp. 333–340.

Tietz NW. Textbook of Clinical Chemistry. (2nd Ed). Saunders Company, Philadelphia. 1994; p. 751

Ross JA, Kasum CM. Dietary flavonoids: Bioavailability, metabolic effects, and safety. Ann Rev Nut. 2002; 22: 19-34.

Chukwuma IF, Nworah FN, Apeh VO, Omeje KO, Nweze EJ, Asogwa CD, Ezeorba TPC. Phytochemical Characterization, Functional Nutrition, and Anti-Diabetic Potentials of Leptadenia hastata (pers) Decne Leaves: In Silico and In Vitro Studies. Bioinfor Bio Ins. 2022; 16. https://doi.org/10.1177/11779322221115436

Ibrahim MA, Habila JD, Koorbanally NA, Islam MS. Butanol fraction of Parkia biglobosa (Jacq.) G. Don leaves enhance pancreatic β-cell functions, stimulates insulin secretion and ameliorates other type 2 diabetes-associated complications in rats. J. Ethnopharm. 2016; 183: 103–111.

Kikuchi A, Takamura T. Where does liver fat go? A possible molecular link between fatty liver and diabetes. J. Diab Investig. 2017; 8: 152–154.

Snel M, Jonker JT, Schoones J, Lamb H, De Roos A, Pijl H, Jazet IM. Ectopic Fat and Insulin Resistance: Pathophysiology and Effect of Diet and Lifestyle Interventions. Inter. J. Endocrin. 2012; 18

Eyth E, Basit H, Smith CJ. Glucose tolerance test. In: StatPearls (Internet). StatPearls Publishing, Florida, USA. 2021.

Narasimhan A, Chinnaiyan M, Karundevi B. Ferulic acid exerts its antidiabetic effect by modulating insulin-signaling molecules in the liver of high-fat diet and fructose-induced type-2 diabetic adult male rat. Appl Physio Nutr and Metab. 2015; 40(8): 769–781.

Peng J, Li Q, Li K, Zhu L, Lin X, Lin X, Shen Q, Li G, Xie X. Quercetin improves glucose and lipid metabolism of diabetic rats: involvement of Akt signalling and SIRT1. J. Diab Res. 2017; 3417306.

Cheng DM, Kuhn P, Poulev A, Rojo LE, Lila MA, Raskin I. In vivo and in vitro antidiabetic effects of aqueous cinnamon extract and cinnamon polyphenol-enhanced food matrix. Food Chem. 2012; 135: 2994–3002.

Sherwani SI, Khan HA, Ekhzaimy A, Masood A, Sakharkar MK. Significance of HbA1c test in diagnosis and prognosis of diabetic patients. Biomark Insig. 2016; 11: 95–104.

Indyk D, BronowickaSzydełko A, Gamian A, Kuzan A. Advanced glycation end products and their receptors in serum of patients with type 2 diabetes. Sci Rep. 2021; 11: 13264

Nazrul M, Bhuiyan I, Mitsuhash S, Sigetomi K, Ubukata M. Quercetin inhibits advanced glycation end product formation via chelating metal ions, trapping methylglyoxal, and trapping reactive oxygen species. Biosci Biotech and Biochem. 2017; 81: 882-890

Kang BPS, Frencher S, Reddy V, Kessler A, Malhotra A, Meggs LG. High glucose promotes mesangial cell apoptosis by oxidant-dependent mechanism,” Am. J. Physiolo-Ren Physiol. 2015; 284(3): 455-466.

Baloyi CM, Khathi A, Sibiya NH, Ngubane PS. The haematological effects of oleanolic acid in streptozotocin-induced diabetic rats: effects on selected markers. J Diab Res. 2019; 6753541.

Livshits L, Barshtein G, Arbell D, Gural A, Levin C, Guizouarn H. Do we store packed red blood cells under “quasi-diabetic” conditions? Biomol. 2021; 11: 992.

Abdelmageed ME, Shehatou GS, Suddek GM, Salem HA. Protocatechuic acid improves hepatic insulin resistance and restores vascular oxidative status in type-2 diabetic rats. Environ Toxicol and Pharmacol. 2021; 83: 103577.

Symon, T., Gaxiola-Robles, R., Hernández-Camacho, C. and Zenteno-Savín, T. Oxidative stress indicators in leukocytes from humans and Bottlenose Dolphins in response to a pro-inflammatory challenge. Free Rad Biol and Med. 2020; 159(48): 378-400.

Nagareddy PR, Murphy A J, Stirzaker RA, Hu Y, Yu S, Miller RG, Ramkhelawon B, Distel E, Westerterp M, Huang LS. Hyperglycemia promotes myelopoiesis and impairs the resolution of atherosclerosis. Cell Metab. 2013; 17: 695–708.

Giese IM, Schilloks MC, Degroote RL, Weigand M, Renner S, Wolf E, Hauck SM, Deeg CA. Chronic hyperglycemia drives functional impairment of lymphocytes in diabetic INSC94Y transgenic pigs. Front Immun. 2021; 11: 607473.

Muller YD, Golshatan D, Ehirchiou D, Wyss JC, Giovannoni L, Meier R, Serre-Beinier V, Yung GP, Morel P, Buhler LH, Seebach JD. Immunosuppressive effects of streptozotocin-induced diabetes result in absolute lymphopenia and a relative increase of T-regulatory cells. Diabetes. 2011; 60: 2331-2340.

Bosevski M, Stojanovska L, Apostolopoulos V. Inflammatory biomarkers: impact for diabetes and diabetic vascular disease. Acta Biochimi et Biophys Sin. 2015; 47(12): 1029–1031

Huang B, Liu J, Ma D, Chen G, Wang W, Fu S. Myricetin prevents dopaminergic neurons from undergoing neuroinflammation-mediated degeneration in a lipopolysaccharide-induced Parkinson’s disease model. J Funct Foods. 2018; 45: 452–461.

Xing J, Yu Z, Zhang X, Li W, Gao D, Wang J, Wang W. Epicatechin alleviates inflammation in lipopolysaccharide-induced acute lung injury in mice by inhibiting the p38 MAPK signaling pathway. Intern Immunopharm. 2019; 66: 146–153.

Ogar I, Egbung GE, Nna VU, Atangwho IJ, Itam EH. Hyptis verticillata attenuates dyslipidemia, oxidative stress and hepato-renal damage in streptozotocin-induced diabetic rats. Life Sci. 2019; 219: 283-293.

Neuschwander-Tetri BA. Non-alcoholic fatty liver disease. BMC Med. 2017; 15(1): 45.

Pei K, Ou J, Huang J, Ou S. p-Coumaric acid and its conjugates: Dietary sources, pharmacokinetic properties and biological activities. J Sci of Food & Agric. 2016; 96: 2952–2962.

Wang AW, Song L, Miao J. “Baicalein attenuates angiotensin II-induced cardiac remodeling via inhibition of AKT/mTOR, ERK1/2, NF-κB, and calcineurin signaling pathways in mice”. Ame J Hyperten. 2015; 28(4): 518–526.

Gross JL, de Azevedo MJ, Silveiro SP, Canani LH, Caramori ML, Zelmanovitz T. Diabetic nephropathy: Diagnosis, prevention, and treatment. Diab Care. 2005; 28: 164-176.

Musso CG, Gregori JA, Jauregui JR, Macías Núñez JF. Creatinine, urea, uric acid, water and electrolytes renal handling in the healthy oldest old. World J Nephr. 2012; 1(5): 123–126.

Liamis G, Liberopoulos E, Barkas F, Elisaf M. Diabetes mellitus and electrolyte disorders. W J Clin Cases. 2014; 2(10): 488-496.

Siboto A, Sibiya N, Khathi A, Ngubane P. The effects of Momordica balsamina methanolic extract on kidney function in STZ-induced diabetic rats: effects on selected metabolic markers. J Diab Res. 2018; 7341242.

Published

2023-10-29

How to Cite

Njoku, O. U., Chibuogwu, C. C., & Nwodo, O. F. (2023). Glycemic and Biochemical Effects of Polyphenol-Rich Fraction of <i>Parkia biglobosa</i> Leaves in Wistar rats Experimentally Induced with Diabetes: http://www.doi.org/10.26538/tjnpr/v7i10.40. Tropical Journal of Natural Product Research (TJNPR), 7(10), 4998–5004. Retrieved from https://tjnpr.org/index.php/home/article/view/2865