Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter June 6, 2019

Trigonella foenum-graceum seed (Fenugreek) hydroalcoholic extract improved the oxidative stress status in a rat model of diabetes-induced memory impairment

  • Solaiman Bafadam , Farimah Beheshti , Tayebeh Khodabakhshi , Amir Asghari , Babak Ebrahimi , Hamid Reza Sadeghnia , Maryam Mahmoudabady ORCID logo , Saeed Niazmand and Mahmoud Hosseini EMAIL logo

Abstract

Background

The antidiabetic and antioxidant effects of Trigonella foenum-graceum have been suggested. The effects of hydroalcoholic extract of the plant seeds and metformin against the diabetes-induced memory impairment were investigated.

Materials and methods

The rats were treated: (1) control, (2) diabetic (3–6) and diabetic rats treated by 50, 100 and 200 mg/kg of the plant extract or metformin. The rats were diabetic by streptozotocin (STZ, 55 mg/kg). After the passive avoidance test, malondialdehyde (MDA), nitric oxide (NO) metabolites, total thiol (SH), catalase (CAT) and superoxide dismutase (SOD) were determined in the brain.

Results

In the diabetic group, at 3, 24 and 48 h after receiving a shock, the latency to enter the dark room was lower than for the controls (p < 0.001). All doses of the extract and metformin increased the latencies to enter the dark at 3 and 24 h after the shock treatment (p < 0.05–p < 0.001). Additionally, the two higher doses of the extract and metformin increased the latency at 48 h after the shock (p < 0.05–p < 0.001). Diabetes also elevated MDA and NO metabolites, while it reduced thiol, SOD and CAT in the hippocampal and cortical tissues (p < 0.001). Treatment of the diabetic animals by the highest dose of the extract and also metformin reduced the MDA and NO metabolites, while it improved thiols, SOD and CAT (p < 0.01–p < 0.001).

Conclusions

Based on our findings, metformin and the hydro-alcoholic extract from the T. foenum-graceum seed prevented memory deficits resulting from diabetes. Preventing oxidative damage in the brain may at least, in part, be responsible for the positive effects of the extract and metformin.

Acknowledgments

The authors would like to thank the Vice Presidency of Research, Mashhad University of Medical Sciences, for the financial support.

Author Statement

  1. Research funding: Mashhad University of Medical Sciences.

  2. Conflict of interest: We declare that we have no conflict of interest.

  3. Informed consent: Not applicable.

  4. Ethical approval: Animal procedures were carried out considering the Guide for the Care and Use of Laboratory Animals and were approved by the Committee on Animal Research of Mashhad University of Medical Sciences. The document number was IR.MUMS.REC.1396.120.

References

[1] Rathmann W, Giani G. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27:2568–9.10.2337/diacare.27.10.2568Search in Google Scholar PubMed

[2] Gispen WH, Biessels G-J. Cognition and synaptic plasticity in diabetes mellitus. Trends Neurosci. 2000;23:542–9.10.1016/S0166-2236(00)01656-8Search in Google Scholar PubMed

[3] McCall AL. The impact of diabetes on the CNS. Diabetes. 1992;41:557–70.10.2337/diab.41.5.557Search in Google Scholar PubMed

[4] Reaven GM, Thompson LW, Nahum D, Haskins E. Relationship between hyperglycemia and cognitive function in older NIDDM patients. Diabetes Care. 1990;13:16–21.10.2337/diacare.13.1.16Search in Google Scholar PubMed

[5] Ryan CM. Neurobehavioral complications of type I diabetes: examination of possible risk factors. Diabetes Care. 1988;11:86–93.10.2337/diacare.11.1.86Search in Google Scholar PubMed

[6] Stewart R, Liolitsa D. Type 2 diabetes mellitus, cognitive impairment and dementia. Diabet Med. 1999;16:93–112.10.1046/j.1464-5491.1999.00027.xSearch in Google Scholar PubMed

[7] Fukui K, Onodera K, Shinkai T, Suzuki S, Urano S. Impairment of learning and memory in rats caused by oxidative stress and aging, and changes in antioxidative defense systems. Ann N Y Acad Sci. 2001;928:168–75.10.1111/j.1749-6632.2001.tb05646.xSearch in Google Scholar PubMed

[8] Baydas G, Nedzvetskii VS, Nerush PA, Kirichenko SV, Yoldas T. Altered expression of NCAM in hippocampus and cortex may underlie memory and learning deficits in rats with streptozotocin-induced diabetes mellitus. Life Sci. 2003;73:1907–916.10.1016/S0024-3205(03)00561-7Search in Google Scholar PubMed

[9] Hawkins CL, Davies MJ. Generation and propagation of radical reactions on proteins. Biochim Biophys Acta. 2001;1504:196–219.10.1016/S0005-2728(00)00252-8Search in Google Scholar PubMed

[10] Srinivasan K. Fenugreek (Trigonella foenum-graecum): a review of health beneficial physiological effects. Food Rev Int. 2006;22:203–24.10.1080/87559120600586315Search in Google Scholar

[11] Kassem A, Al-Aghbari A, Molham A-H, Al-Mamary M. Evaluation of the potential antifertility effect of fenugreek seeds in male and female rabbits. Contraception. 2006;73:301–6.10.1016/j.contraception.2005.08.020Search in Google Scholar PubMed

[12] Belaïd-Nouira Y, Bakhta H, Bouaziz M, Flehi-Slim I, Haouas Z, Cheikh HB. Study of lipid profile and parieto-temporal lipid peroxidation in AlCl3 mediated neurotoxicity. Modulatory effect of fenugreek seeds. Lipids Health Dis. 2012;11:16.10.1186/1476-511X-11-16Search in Google Scholar PubMed

[13] Satheeshkumar N, Mukherjee PK, Bhadra S, Saha B. Acetylcholinesterase enzyme inhibitory potential of standardized extract of Trigonella foenum graecum L. and its constituents. Phytomedicine. 2010;17:292–5.10.1016/j.phymed.2009.06.006Search in Google Scholar PubMed

[14] Belaïd-Nouira Y, Bakhta H, Samoud S, Trimech M, Haouas Z, Ben Cheikh H. A novel insight on chronic AlCl3 neurotoxicity through IL-6 and GFAP expressions: modulating effect of functional food fenugreek seeds. Nutr Neurosci. 2013;16:218–24.10.1179/1476830512Y.0000000048Search in Google Scholar PubMed

[15] Belaid-Nouira Y, Bakhta H, Haouas Z, Flehi-Slim I, Neffati F, Najjar MF, et al. Fenugreek seeds, a hepatoprotector forage crop against chronic AlCl3 toxicity. BMC Vet Res. 2013;9:22.10.1186/1746-6148-9-22Search in Google Scholar PubMed

[16] Naghibi SM, Hosseini M, Khani F, Rahimi M, Vafaee F, Rakhshandeh H, et al. Effect of aqueous extract of Crocus sativus L. on morphine-induced memory impairment. Adv Pharmacol Sci. 2012;2012:494367.10.1155/2012/494367Search in Google Scholar PubMed

[17] Farrokhi E, Hosseini M, Beheshti F, Vafaee F, Hadjzadeh MA-R, Dastgheib SS. Brain tissues oxidative damage as a possible mechanism of deleterious effects of propylthiouracil-induced hypothyroidism on learning and memory in neonatal and juvenile growth in rats. Basic Clin Neurosci. 2014;5:285–94.Search in Google Scholar PubMed

[18] Khodabandehloo F, Hosseini M, Rajaei Z, Soukhtanloo M, Farrokhi E, Rezaeipour M. Brain tissue oxidative damage as a possible mechanism for the deleterious effect of a chronic high dose of estradiol on learning and memory in ovariectomized rats. Arq Neuropsiquiatr. 2013;71:313–9.10.1590/0004-282X20130027Search in Google Scholar PubMed

[19] Ellman GL. Tissue sulfhydryl groups. Arch Biochem Biophys. 1959;82:70–7.10.1016/0003-9861(59)90090-6Search in Google Scholar PubMed

[20] Beheshti F, Hosseini M, Shafei MN, Soukhtanloo M, Ghasemi S, Vafaee F, et al. The effects of Nigella sativa extract on hypothyroidism-associated learning and memory impairment during neonatal and juvenile growth in rats. Nutr Neurosci. 2017;20:49–59.10.1179/1476830514Y.0000000144Search in Google Scholar PubMed

[21] Madesh M, Balasubramanian KA. Microtiter plate assay for superoxide dismutase using MTT reduction by superoxide. Indian J Biochem Biophys 1998;35:184–8.Search in Google Scholar PubMed

[22] Asghari A, Hosseini M, Beheshti F, Shafei MN, Mehri S. Inducible nitric oxide inhibitor aminoguanidine, ameliorated oxidative stress, interleukin-6 concentration and improved brain-derived neurotrophic factor in the brain tissues of neonates born from titanium dioxide nanoparticles exposed rats. J Matern Fetal Neonatal Med. 2018:1–12.10.1080/14767058.2018.1480602Search in Google Scholar PubMed

[23] Ghasemi S, Moradzadeh M, Hosseini M, Beheshti F, Sadeghnia HR. Beneficial effects of Urtica dioica on scopolamine-induced memory impairment in rats: protection against acetylcholinesterase activity and neuronal oxidative damage. Drug Chem Toxicol. 2018:1–9.10.1080/01480545.2018.1463238Search in Google Scholar

[24] Salmani H, Hosseini M, Beheshti F, Baghcheghi Y, Sadeghnia HR, Soukhtanloo M, et al. Angiotensin receptor blocker, losartan ameliorates neuroinflammation and behavioral consequences of lipopolysaccharide injection. Life Sci. 2018;203:161–70.10.1016/j.lfs.2018.04.033Search in Google Scholar PubMed

[25] Strachan M, Frier B, Deary I. Type 2 diabetes and cognitive impairment. Diabet Med. 2003;20:1–2.10.1046/j.1464-5491.2003.00855.xSearch in Google Scholar PubMed

[26] Sommerfield AJ, Deary IJ, Frier BM. Acute hyperglycemia alters mood state and impairs cognitive performance in people with type 2 diabetes. Diabetes Care. 2004;27:2335–40.10.2337/diacare.27.10.2335Search in Google Scholar PubMed

[27] Biessels G-J, Kamal A, Urban IJ, Spruijt BM, Erkelens DW, Gispen WH. Water maze learning and hippocampal synaptic plasticity in streptozotocin-diabetic rats: effects of insulin treatment. Brain Res. 1998;800:125–35.10.1016/S0006-8993(98)00510-1Search in Google Scholar PubMed

[28] Biessels GJ, Staekenborg S, Brunner E, Brayne C, Scheltens P. Risk of dementia in diabetes mellitus: a systematic review. Lancet Neurol. 2006;5:64–74.10.1016/S1474-4422(05)70284-2Search in Google Scholar PubMed

[29] Tamaddonfard E, Farshid AA, Asri-Rezaee S, Javadi S, Khosravi V, Rahman B, et al. Crocin improved learning and memory impairments in streptozotocin-induced diabetic rats. Iran J Basic Med Sci. 2013;16:91–100.Search in Google Scholar PubMed

[30] Parinandi NL, Thompson EW, Schmid HH. Diabetic heart and kidney exhibit increased resistance to lipid peroxidation. Biochim Biophys Acta. 1990;1047:63–9.10.1016/0005-2760(90)90261-USearch in Google Scholar PubMed

[31] Kakkar R, Kalra J, Mantha SV, Prasad K. Lipid peroxidation and activity of antioxidant enzymes in diabetic rats. Mol Cell Biochem. 1995;151:113–9.10.1007/BF01322333Search in Google Scholar PubMed

[32] Baydas G, Canatan H, Turkoglu A. Comparative analysis of the protective effects of melatonin and vitamin E on streptozocin-induced diabetes mellitus. J Pineal Res. 2002;32:225–30.10.1034/j.1600-079X.2002.01856.xSearch in Google Scholar PubMed

[33] Baydas G, Sonkaya E, Tuzcu M, Yasar A, Donder E. Novel role for gabapentin in neuroprotection of central nervous system in streptozotocine-induced diabetic rats. Acta Pharmacol Sin. 2005;26:417–22.10.1111/j.1745-7254.2005.00072.xSearch in Google Scholar PubMed

[34] Çelik S, Baydaş G, Yılmaz Ö. Influence of vitamin E on the levels of fatty acids and MDA in some tissues of diabetic rats. Cell Biochem Funct. 2002;20:67–71.10.1002/cbf.936Search in Google Scholar PubMed

[35] Van Dam PS, Gispen WH, Bravenboer B, Van Asbeck BS, Erkelens DW, Marx JJ. The role of oxidative stress in neuropathy and other diabetic complications. Diabetes Metab Rev. 1995;11:181–92.10.1002/dmr.5610110303Search in Google Scholar PubMed

[36] Baynes JW. Role of oxidative stress in development of complications in diabetes. Diabetes. 1991;40:405–12.10.2337/diab.40.4.405Search in Google Scholar PubMed

[37] Majithiya JB, Balaraman R. Metformin reduces blood pressure and restores endothelial function in aorta of streptozotocin-induced diabetic rats. Life Sci. 2006;78:2615–24.10.1016/j.lfs.2005.10.020Search in Google Scholar PubMed

[38] Sena CM, Matafome P, Louro T, Nunes E, Fernandes R, Seiça RM. Metformin restores endothelial function in aorta of diabetic rats. Br J Pharmacol. 2011;163:424–37.10.1111/j.1476-5381.2011.01230.xSearch in Google Scholar PubMed PubMed Central

[39] Bonnefont-Rousselot D, Raji B, Walrand S, Gardes-Albert M, Jore D, Legrand A, et al. An intracellular modulation of free radical production could contribute to the beneficial effects of metformin towards oxidative stress. Metabolism. 2003;52:586–9.10.1053/meta.2003.50093Search in Google Scholar PubMed

[40] Faure P, Rossini E, Wiernsperger N, Richard MJ, Favier A, Halimi S. An insulin sensitizer improves the free radical defense system potential and insulin sensitivity in high fructose-fed rats. Diabetes. 1999;48:353–7.10.2337/diabetes.48.2.353Search in Google Scholar PubMed

[41] Stepensky D, Friedman M, Raz I, Hoffman A. Pharmacokinetic-pharmacodynamic analysis of the glucose-lowering effect of metformin in diabetic rats reveals first-pass pharmacodynamic effect. Drug Metab Dispos. 2002;30:861–8.10.1124/dmd.30.8.861Search in Google Scholar PubMed

[42] Goyal S, Gupta N, Chatterjee S. Investigating therapeutic potential of Trigonella foenum-graecum L. as our defense mechanism against several human diseases. J Toxicol. 2016;2016:1250387.10.1155/2016/1250387Search in Google Scholar PubMed PubMed Central

[43] Shetty K. Biotechnology to harness the benefits of dietary phenolics: focus on Lamiaceae. Asia Pac J Clin Nutr. 1997;6:162–71.Search in Google Scholar PubMed

[44] Basch E, Ulbricht C, Kuo G, Szapary P, Smith M. Therapeutic applications of fenugreek. Altern Med Rev. 2003;8:20–27.Search in Google Scholar PubMed

[45] Satheeshkumar N, Mukherjee PK, Bhadra S, Saha BP. Acetylcholinesterase enzyme inhibitory potential of standardized extract of Trigonella foenum graecum L. and its constituents. Phytomedicine. 2010;17:292–5.10.1016/j.phymed.2009.06.006Search in Google Scholar PubMed

[46] Khursheed R, Rizwani GH, Sultana V, Ahmed M, Kamil A. Antidepressant effect and categorization of inhibitory activity of monoamine oxidase type A and B of ethanolic extract of seeds of Trigonella foenum graecum Linn. Pak J Pharm Sci. 2014;27:1419–25.Search in Google Scholar

[47] Kandhare AD, Bodhankar SL, Mohan V, Thakurdesai PA. Effect of glycosides based standardized fenugreek seed extract in bleomycin-induced pulmonary fibrosis in rats: Decisive role of Bax, Nrf2, NF-kappaB, Muc5ac, TNF-alpha and IL-1beta. Chem Biol Interact. 2015;237:151–65.10.1016/j.cbi.2015.06.019Search in Google Scholar PubMed

[48] Fuller S, Stephens JM. Diosgenin, 4-hydroxyisoleucine, and fiber from fenugreek: mechanisms of actions and potential effects on metabolic syndrome. Adv Nutr. 2015;6:189–97.10.3945/an.114.007807Search in Google Scholar PubMed PubMed Central

[49] Prema A, Thenmozhi AJ, Manivasagam T, Essa MM, Akbar MD, Akbar M. Fenugreek seed powder nullified aluminium chloride induced memory loss, biochemical changes, abeta burden and apoptosis via regulating Akt/GSK3beta signaling pathway. PLoS One. 2016;11:e0165955.10.1371/journal.pone.0165955Search in Google Scholar PubMed PubMed Central

[50] Saini D, Dhingra AK, Chopra B, Parle M. Psychopharmacological investigation of the nootropic potential of Trigonella foenum Linn in mice. Asian J Pharm Clin Res. 2011;4:76–84.Search in Google Scholar

[51] Genet S, Kale RK, Baquer NZ. Alterations in antioxidant enzymes and oxidative damage in experimental diabetic rat tissues: effect of vanadate and fenugreek (Trigonella foenum graecum). Mol Cell Biochem. 2002;236:7–12.10.1023/A:1016103131408Search in Google Scholar PubMed

Received: 2018-11-12
Accepted: 2019-02-14
Published Online: 2019-06-06

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 26.4.2024 from https://www.degruyter.com/document/doi/10.1515/hmbci-2018-0074/html
Scroll to top button