Volume 7, Issue 1 (Journal of Clinical and Basic Research (JCBR) 2023)                   jcbr 2023, 7(1): 13-20 | Back to browse issues page

XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Dahiru M M. Recent advances in the therapeutic potential phytochemicals in managing diabetes. jcbr 2023; 7 (1) :13-20
URL: http://jcbr.goums.ac.ir/article-1-385-en.html
Department of Science Laboratory Technology, School of Science and Technology, Adamawa State Polytechnic, Yola , mubaraq93@gmail.com
Abstract:   (961 Views)
Diabetes and diabetic complications have been a global menace for a long time, putting a heavy burden on an individual, the health sector, and governments. Diabetic complications attributed to persistent hyperglycemia create a challenge in managing diabetes, considering the nature of the disease as a group of metabolic disorders. Various phytochemicals target different metabolic pathways and molecules through different mechanisms of action, acting individually or synergistically to achieve therapeutic goals. Phytochemicals such as alkaloids, saponins, glycosides, terpenoids, and flavonoids were reported to exert different anti-diabetic effects, including anti-hyperglycemic, anti-hyperlipidemic, anti-inflammatory, antioxidant, and insulinotropic activities. The present review focused on the recent advances in the therapeutic potentials of phytochemicals in managing diabetes and diabetic complications, emphasizing their in vitro and in vivo studies.
 
Full-Text [PDF 572 kb]   (235 Downloads)    
Article Type: Review | Subject: Biochemistry

References
1. ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, et al. Introduction and Methodology: Standards of Care in Diabetes-2023. Diabetes Care. 2023; 46(Suppl.1): S1-4. [View at Publisher] [DOI] [PMID] [Google Scholar]
2. American Diabetes Association Professional Practice Committee. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2022. Diabetes Care. 2022; 45(Suppl.1): S17-38. [View at Publisher] [DOI] [PMID] [Google Scholar]
3. American Diabetes Association. Diagnosis and Classification of Diabetes Mellitus. Diabetes Care. 2013;37(Supplement-1):S81-90. [View at Publisher] [DOI] [PMID] [Google Scholar]
4. Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119. [View at Publisher] [DOI] [PMID] [Google Scholar]
5. Dahiru MM, Nadro SM. A review of the Mechanisms of Action and Side Effects of Anti-diabetic Agents. Trends in Pharmaceutical Sciences. 2022;8(3):195-210. [View at Publisher] [DOI] [Google Scholar]
6. Dahiru MM, Badgal EB, Musa N. Phytochemistry, GS-MS analysis, and heavy metals composition of aqueous and ethanol stem bark extracts of Ximenia americana. GSC Biological and Pharmaceutical Sciences. 2022;21(3):145-56. [View at Publisher] [DOI] [Google Scholar]
7. Kurek J. editor. Introductory Chapter: Alkaloids - Their Importance in Nature and for Human Life. Rijeka: IntechOpen; 2019. p.Ch.1. [View at Publisher] [DOI] [Google Scholar]
8. Heinrich M, Mah J, Amirkia V. Alkaloids Used as Medicines: Structural Phytochemistry Meets Biodiversity-An Update and Forward Look. Molecules. 2021;26(7):1836. [View at Publisher] [DOI] [PMID] [Google Scholar]
9. Venkatesan GK, Kuppusamy A, Devarajan S, Kumar AKK. Review on medicinal potential of alkaloids and saponins. Pharamacologyonline. 2019;1:1-20. [View at Publisher] [Google Scholar]
10. Aryal B, Raut BK, Bhattarai S, Bhandari S, Tandan P, Gyawali K, et al. Potential Therapeutic Applications of Plant-Derived Alkaloids against Inflammatory and Neurodegenerative Diseases. Evid Based Complement Alternat Med. 2022;2022:1-18. [View at Publisher] [DOI] [PMID] [Google Scholar]
11. Behl T, Gupta A, Albratty M, Najmi A, Meraya AM, Alhazmi HA, et al. Alkaloidal Phytoconstituents for Diabetes Management: Exploring the Unrevealed Potential. Molecules. 2022;27(18):5851. [View at Publisher] [DOI] [PMID] [Google Scholar]
12. Mechanick JI, Marchetti A, Hegazi R, Hamdy O. Diabetes-Specific Nutrition Formulas in the Management of Patients with Diabetes and Cardiometabolic Risk. Nutrients. 2020;12(12):3616. [View at Publisher] [DOI] [PMID] [Google Scholar]
13. Azam K, Rasheed MA, Omer MO, Altaf I, Akhlaq A. Anti-hyperlipidemic and anti-diabetic evaluation of ethanolic leaf extract of Catharanthus roseus alone and in combination therapy. Brazilian Journal of Pharmaceutical Sciences. 2022;58. [View at Publisher] [DOI] [Google Scholar]
14. Ge Q, Chen L, Yuan Y, Liu L, Feng F, Lv P, et al. Network pharmacology-based dissection of the anti-diabetic mechanism of Lobelia chinensis. Front Pharmacol. 2020;11:347. [View at Publisher] [DOI] [PMID] [Google Scholar]
15. DelİOrman Orhan D, Orhan N, Demir Ö, KonuklugİL B. In Vitro Antidiabetic and Antioxidant Effects of Thirteen Marine Organisms From Mediterranean Sea. Farmacia. 2021;69(1):68-74. [View at Publisher] [DOI] [PMID] [Google Scholar]
16. Hassan SS, Muhammad I, Abbas SQ, Hassan M, Majid M, Jin HZ, et al. Stress driven discovery of natural products from actinobacteria with anti-oxidant and cytotoxic activities including docking and admet properties. Int J Mol Sci. 2021;22(21):11432. [View at Publisher] [DOI] [PMID] [Google Scholar]
17. Uvarani C, Jaivel N, Sankaran M, Chandraprakash K, Ata A, Mohan PS. Axially chiral biscarbazoles and biological evaluation of the constituents from Murraya koenigii. Fitoterapia. 2014;94:10-20. [View at Publisher] [DOI] [PMID] [Google Scholar]
18. Tiong SH, Looi CY, Arya A, Wong WF, Hazni H, Mustafa MR, et al. Vindogentianine, a hypoglycemic alkaloid from Catharanthus roseus (L.) G. Don (Apocynaceae). Fitoterapia. 2015;102:182-8. [View at Publisher] [DOI] [PMID] [Google Scholar]
19. Jan NU, Ali A, Ahmad B, Iqbal N, Adhikari A, Ali A, et al. Evaluation of antidiabetic potential of steroidal alkaloid of Sarcococca saligna. Biomedicine and Pharmacotherapy. 2018;100:461-6. [View at Publisher] [DOI] [PMID] [Google Scholar]
20. Patel OPS, Mishra A, Maurya R, Saini D, Pandey J, Taneja I, et al. Naturally occurring carbazole alkaloids from Murraya koenigii as potential antidiabetic agents. Journal of Natural Products. 2016;79(5):1276-84. [View at Publisher] [DOI] [PMID] [Google Scholar]
21. Patel MB, Mishra S. Isoquinoline alkaloids from Tinospora cordifolia inhibit rat lens aldose reductase. Phytotherapy Research. 2012;26(9):1342-7. [View at Publisher] [DOI] [PMID] [Google Scholar]
22. Cho H. Protein tyrosine phosphatase 1B (PTP1B) and obesity. Vitamins and Hormones. 2013;91:405-24. [View at Publisher] [DOI] [PMID] [Google Scholar]
23. Lankatillake C, Huynh T, Dias DA. Understanding glycaemic control and current approaches for screening antidiabetic natural products from evidence-based medicinal plants. Plant Methods. 2019;15(1):1-35. [View at Publisher] [DOI] [PMID] [Google Scholar]
24. Wiedemann M, Gurrola-Díaz CM, Vargas-Guerrero B, Wink M, García-López PM, Düfer M. Lupanine improves glucose homeostasis by influencing KATP channels and insulin gene expression. Molecules. 2015;20(10):19085-100. [View at Publisher] [DOI] [PMID] [Google Scholar]
25. Subramanian SP, Prasath GS. Antidiabetic and antidyslipidemic nature of trigonelline, a major alkaloid of fenugreek seeds studied in high-fat-fed and low-dose streptozotocin-induced experimental diabetic rats. Biomedicine & Preventive Nutrition. 2014;4(4):475-80. [View at Publisher] [DOI] [PMID] [Google Scholar]
26. Freitas Ld, Valli M, Dametto AC, Pennacchi PC, Andricopulo AD, Maria-Engler SS, et al. Advanced glycation end product inhibition by alkaloids from Ocotea paranapiacabensis for the prevention of skin aging. Journal of Natural Products. 2020;83(3):649-56. [View at Publisher] [DOI] [PMID] [Google Scholar]
27. Ye G, Huang C, Li J, Chen T, Tang J, Liu W, et al. Isolation, Structural Characterization and Antidiabetic Activity of New Diketopiperazine Alkaloids from Mangrove Endophytic Fungus Aspergillus sp. 16-5c. Marine Drugs [Internet]. 2021; 19(7). [View at Publisher] [DOI] [PMID] [Google Scholar]
28. Li C-J, Chen P-N, Li H-J, Mahmud T, Wu D-L, Xu J, et al. Potential Antidiabetic Fumiquinazoline Alkaloids from the Marine-Derived Fungus Scedosporium apiospermum F41-1. Journal of Natural Products. 2020;83(4):1082-91. [View at Publisher] [DOI] [PMID] [Google Scholar]
29. Zhang H, Hui J, Yang J, Deng J, Fan D. Eurocristatine, a plant alkaloid from Eurotium cristatum, alleviates insulin resistance in db/db diabetic mice via activation of PI3K/AKT signaling pathway. European Journal of Pharmacology. 2020;887:173557. [View at Publisher] [DOI] [PMID] [Google Scholar]
30. Bai J, Zhang S, Cao J, Sun H, Mang Z, Shen WL, et al. Hernandezine, a natural herbal alkaloid, ameliorates type 2 diabetes by activating AMPK in two mouse models. Phytomedicine. 2022;105:154366. [View at Publisher] [DOI] [PMID] [Google Scholar]
31. Dou Y, Huang R, Li Q, Liu Y, Li Y, Chen H, et al. Oxyberberine, an absorbed metabolite of berberine, possess superior hypoglycemic effect via regulating the PI3K/Akt and Nrf2 signaling pathways. Biomedicine and Pharmacotherapy. 2021;137:111312. [View at Publisher] [DOI] [PMID] [Google Scholar]
32. Zang Y. Pharmacological Activities of Coumarin Compounds in Licorice: A Review. Natural Product Communications. 2020;15(9):1934578X20953954. [View at Publisher] [DOI] [PMID] [Google Scholar]
33. Yang Y, Laval S, Yu B. Chapter Two - Chemical Synthesis of Saponins. In: Baker DC, editor. Advances in Carbohydrate Chemistry and Biochemistry. 79: Academic Press; 2021. p. 63-150. [View at Publisher] [DOI] [PMID] [Google Scholar]
34. Amraei S, Ahmadi S. Recent studies on antimicrobial and anticancer activities of saponins: A mini-review. Nano Micro Biosystems. 2022;1(1):22-6. [View at Publisher] [DOI] [PMID] [Google Scholar]
35. Kareem O, Ali T, Dar LA, Mir SA, Rashid R, Nazli N, et al. Positive Health Benefits of Saponins from Edible Legumes: Phytochemistry and Pharmacology. Edible Plants in Health and Diseases. 2022:279-98. [View at Publisher] [DOI] [PMID] [Google Scholar]
36. Ashour A, Abed El Aziz M, Melad A. A review on saponins from medicinal plants: chemistry, isolation, and determination. Journal of Nanomedicine Research. 2019;7:282-8. [View at Publisher] [DOI] [PMID] [Google Scholar]
37. Choudhary N, Khatik LG, Suttee A. The Possible Role of Saponin in Type-II Diabetes- A Review. Current Diabetes Reviews. 2021;17(2):107-21. https://doi.org/10.2174/1573399816666200516173829 [View at Publisher] [DOI] [PMID] [Google Scholar]
38. Zhou Y, Xu B. New insights into anti-diabetes effects and molecular mechanisms of dietary saponins. Critical Reviews in Food Science and Nutrition. 2022:1-26. [View at Publisher] [DOI] [PMID] [Google Scholar]
39. Jiang S, Xu L, Xu Y, Guo Y, Wei L, Li X, et al. Antidiabetic effect of Momordica charantia saponins in rats induced by high-fat diet combined with STZ. Electronic Journal of Biotechnology. 2020;43:41-7. [View at Publisher] [DOI] [PMID] [Google Scholar]
40. 4Keller AC, He K, Brillantes A-M, Kennelly EJ. A characterized saponin-rich fraction of Momordica charantia shows antidiabetic activity in C57BLK/6 mice fed a high fat diet. Phytomedicine Plus. 2021;1(4):100134. [View at Publisher] [DOI] [PMID] [Google Scholar]
41. Ejelonu OC, Elekofehinti OO, Adanlawo IG, Kundu R. TGR5 potentiates GLP-1 secretion and cause pancreatic islet regeneration in response to Tithonia diversifolia saponin-rich extract in diabetic model mice. Phytomedicine Plus. 2022;2(1):100203. [View at Publisher] [DOI] [PMID] [Google Scholar]
42. An S, Niu D, Wang T, Han B, He C, Yang X, et al. Total Saponins Isolated from Corni Fructus via Ultrasonic Microwave-Assisted Extraction Attenuate Diabetes in Mice. Foods [Internet]. 2021;10(3). [View at Publisher] [DOI] [PMID] [Google Scholar]
43. Parveen A, Farooq MA, Kyunn WW. A New Oleanane Type Saponin from the Aerial Parts of Nigella sativa with Anti-Oxidant and Anti-Diabetic Potential. Molecules [Internet]. 2020;25(9). [View at Publisher] [DOI] [PMID] [Google Scholar]
44. Samaddar S, Jha DK, Koneri R. Optimization of pancreatic islet isolation from rat and evaluation of islet protective potential of a saponin isolated from fruits of Momordica dioica. Journal of Applied Pharmaceutical Science. 2020;10(7):089-99. [View at Publisher] [DOI] [PMID] [Google Scholar]
45. Wang Q, Wu X, Shi F, Liu Y. Comparison of antidiabetic effects of saponins and polysaccharides from Momordica charantia L. in STZ-induced type 2 diabetic mice. Biomedicine and Pharmacotherapy. 2019;109:744-50. [View at Publisher] [DOI] [PMID] [Google Scholar]
46. Kamal R, Kharbach M, Heyden YV, Yu H, Bouklouze A, Cherrah Y, et al. In Vitro & In Vivo Anti-Hyperglycemic Potential of Saponins Cake and Argan Oil from Argania spinosa. Foods [Internet]. 2021;10(5). [View at Publisher] [DOI] [PMID] [Google Scholar]
47. Boussoussa H, Khacheba I, Berramdane T, Maamri A, Bendahgane H, Yousfi M. In vitro antidiabetic effect of saponins and phenolic extracts from fruits and seeds of Algerian cypress tree: Cupressus sempervirens L. Current Enzyme Inhibition. 2018;14(2):92-6. [View at Publisher] [DOI] [PMID] [Google Scholar]
48. Lundqvist LCE, Rattigan D, Ehtesham E, Demmou C, Östenson C-G, Sandström C. Profiling and activity screening of Dammarane-type triterpen saponins from Gynostemma pentaphyllum with glucose-dependent insulin secretory activity. Scientific Reports. 2019;9(1):627. [View at Publisher] [DOI] [PMID] [Google Scholar]
49. Xiong H, Zhang S, Zhao Z, Zhao P, Chen L, Mei Z. Antidiabetic activities of entagenic acid in type 2 diabetic db/db mice and L6 myotubes via AMPK/GLUT4 pathway. Journal of Ethnopharmacology. 2018;211:366-74. [View at Publisher] [DOI] [PMID] [Google Scholar]
50. Pandey AR, Ahmad S, Singh SP, Mishra A, Bisen AC, Sharma G, et al. Furostanol saponins from Asparagus racemosus as potential hypoglycemic agents. Phytochemistry. 2022;201:113286. [View at Publisher] [DOI] [PMID] [Google Scholar]
51. Al Sharif M, Alov P, Diukendjieva A, Vitcheva V, Simeonova R, Krasteva I, et al. Molecular determinants of PPARγ partial agonism and related in silico/in vivo studies of natural saponins as potential type 2 diabetes modulators. Food and Chemical Toxicology. 2018;112:47-59. [View at Publisher] [DOI] [PMID] [Google Scholar]
52. Salah El Dine R, Abdallah HM, Kandil ZA, Zaki AA, Khan SI, Khan IA. PPARα and γ Activation Effects of New Nor-triterpenoidal Saponins from the Aerial Parts of Anabasis articulata. Planta Medica. 2019;85(04):274-81. Epub 10/25. En. [View at Publisher] [DOI] [PMID] [Google Scholar]
53. Dubey K, Dubey R, Gupta RA, Gupta AK. Anti-diabetic and antioxidant potential of saponin extract of leaves of Ziziphus mauritiana. Journal of Drug Delivery and Therapeutics. 2019;9(2-A):75-7. [View at Publisher] [DOI] [PMID] [Google Scholar]
54. Chai Y, Luo J, Bao Y. Effects of Polygonatum sibiricum saponin on hyperglycemia, gut microbiota composition and metabolic profiles in type 2 diabetes mice. Biomedicine and Pharmacotherapy. 2021;143:112155. [View at Publisher] [DOI] [PMID] [Google Scholar]
55. Zhu X, Li C, Zhu Y. Saponins extracted from Asparagus officinalis L. by products exerts hypoglycemic effect in streptozotocin induced type 2 diabetic rats. IOP Conference Series: Earth and Environmental Science. 2020;559(1):012002. [View at Publisher] [DOI] [PMID] [Google Scholar]
56. Guo X, Sun W, Luo G, Wu L, Xu G, Hou D, et al. Panax notoginseng saponins alleviate skeletal muscle insulin resistance by regulating the IRS1-PI3K-AKT signaling pathway and GLUT4 expression. FEBS Open Bio. 2019;9(5):1008-19. [View at Publisher] [DOI] [PMID] [Google Scholar]
57. Gangasani JK, Pemmaraju DB, Murthy USN, Rengan AK, Naidu VGM. Chapter 4 - Chemistry of herbal biomolecules. In: Mandal SC, Nayak AK, Dhara AK, editors. Herbal Biomolecules in Healthcare Applications: Academic Press; 2022. p.63-79. [View at Publisher] [DOI] [PMID] [Google Scholar]
58. Bartnik M, Facey PC. Chapter 8 - Glycosides. In: Badal S, Delgoda R, editors. Pharmacognosy. Boston: Academic Press; 2017. p.101-61. [View at Publisher] [DOI] [PMID] [Google Scholar]
59. Pałasz A, Cież D, Trzewik B, Miszczak K, Tynor G, Bazan B. In the Search of Glycoside-Based Molecules as Antidiabetic Agents. Topics in Current Chemistry. 2019;377(4):19. [View at Publisher] [DOI] [PMID] [Google Scholar]
60. Salehi B, Ata A, V. Anil Kumar N, Sharopov F, Ramirez-Alarcon K, Ruiz-Ortega A, et al. Antidiabetic potential of medicinal plants and their active components. Biomolecules. 2019;9(10):551. [View at Publisher] [DOI] [PMID] [Google Scholar]
61. Myint KZ, Chen J-m, Zhou Z-y, Xia Y-m, Lin J, Zhang J. Structural dependence of antidiabetic effect of steviol glycosides and their metabolites on streptozotocin-induced diabetic mice. Journal of the Science of Food and Agriculture. 2020;100(10):3841-9. [View at Publisher] [DOI] [PMID] [Google Scholar]
62. Kurek JM, Krejpcio Z. The functional and health-promoting properties of Stevia rebaudiana Bertoni and its glycosides with special focus on the antidiabetic potential - A review. Journal of Functional Foods. 2019;61:103465. [View at Publisher] [DOI] [PMID] [Google Scholar]
63. Abdel Motaal A, Salem HH, Almaghaslah D, Alsayari A, Bin Muhsinah A, Alfaifi MY, et al. Flavonol Glycosides: In Vitro Inhibition of DPPIV, Aldose Reductase and Combating Oxidative Stress are Potential Mechanisms for Mediating the Antidiabetic Activity of Cleome droserifolia. Molecules [Internet]. 2020;25(24). [View at Publisher] [DOI] [PMID] [Google Scholar]
64. Perera WH, Shivanagoudra SR, Pérez JL, Kim DM, Sun Y, K. Jayaprakasha G, et al. Anti-Inflammatory, Antidiabetic Properties and In Silico Modeling of Cucurbitane-Type Triterpene Glycosides from Fruits of an Indian Cultivar of Momordica charantia L. Molecules [Internet]. 2021; 26(4). [View at Publisher] [DOI] [PMID] [Google Scholar]
65. Luyen NT, Binh PT, Tham PT, Hung TM, Dang NH, Dat NT, et al. Wedtrilosides A and B, two new diterpenoid glycosides from the leaves of Wedelia trilobata (L.) Hitchc. with α-amylase and α-glucosidase inhibitory activities. Bioorganic Chemistry. 2019;85:319-24. [View at Publisher] [DOI] [PMID] [Google Scholar]
66. Rampadarath A, Balogun FO, Pillay C, Sabiu S. Identification of Flavonoid C-Glycosides as Promising Antidiabetics Targeting Protein Tyrosine Phosphatase 1B. Journal of Diabetes Research. 2022;2022:6233217. [View at Publisher] [DOI] [PMID] [Google Scholar]
67. Kurek JM, Król E, Krejpcio Z. Steviol Glycosides Supplementation Affects Lipid Metabolism in High-Fat Fed STZ-Induced Diabetic Rats. Nutrients [Internet]. 2021;13(1). [View at Publisher] [DOI] [PMID] [Google Scholar]
68. Mohammed HS, Abdel-Aziz MM, Abu-Baker MS, Saad AM, Mohamed MA, Ghareeb MA. Antibacterial and potential antidiabetic activities of flavone C-glycosides isolated from Beta vulgaris subspecies cicla L. var. flavescens (Amaranthaceae) cultivated in Egypt. Current Pharmaceutical Biotechnology. 2019;20(7):595-604. [View at Publisher] [DOI] [PMID] [Google Scholar]
69. Zaidan UH, Mohamad Zen NI, Amran NA, Shamsi S, Gani SSA. Biochemical evaluation of phenolic compounds and steviol glycoside from Stevia rebaudiana extracts associated with in vitro antidiabetic potential. Biocatalysis and Agricultural Biotechnology. 2019;18:101049. [View at Publisher] [DOI] [PMID] [Google Scholar]
70. Shenoy RS, Prashanth KVH, Manonmani HK. In Vitro Antidiabetic Effects of Isolated Triterpene Glycoside Fraction from Gymnema sylvestre. Evidence-Based Complementary and Alternative Medicine. 2018;2018:7154702. [View at Publisher] [DOI] [PMID] [Google Scholar]
71. Dzydzan O, Brodyak I, Sokół-Łętowska A, Kucharska AZ, Sybirna N. Loganic Acid, an Iridoid Glycoside Extracted from Cornus mas L. Fruits, Reduces of Carbonyl/Oxidative Stress Biomarkers in Plasma and Restores Antioxidant Balance in Leukocytes of Rats with Streptozotocin-Induced Diabetes Mellitus. Life [Internet]. 2020;10(12). [View at Publisher] [DOI] [PMID] [Google Scholar]
72. Zhu K, Meng Z, Tian Y, Gu R, Xu Z, Fang H, et al. Hypoglycemic and hypolipidemic effects of total glycosides of Cistanche tubulosa in diet/streptozotocin-induced diabetic rats. Journal of Ethnopharmacology. 2021;276:113991. [View at Publisher] [DOI] [PMID] [Google Scholar]
73. Huang Z-Y, Jia S-S, Jia A, Huang J-W, Yuan K. Antidiabetic potential of the total flavone glycoside from okra fruit in type 2 diabetic rats. Pharmacognosy Magazine. 2018;14(58):482-8. [View at Publisher] [DOI] [PMID] [Google Scholar]
74. Sun H, Tan J, Lv W, Li J, Wu J, Xu J, et al. Hypoglycemic triterpenoid glycosides from Cyclocarya paliurus (Sweet Tea Tree). Bioorganic Chemistry. 2020;95:103493. [View at Publisher] [DOI] [PMID] [Google Scholar]
75. Isah MB, Tajuddeen N, Umar MI, Alhafiz ZA, Mohammed A, Ibrahim MA. Chapter 7 - Terpenoids as Emerging Therapeutic Agents: Cellular Targets and Mechanisms of Action against Protozoan Parasites. In: Atta ur R, editor. Studies in Natural Products Chemistry. 59: Elsevier; 2018. p. 227-50. [View at Publisher] [DOI] [PMID] [Google Scholar]
76. Yang W, Chen X, Li Y, Guo S, Wang Z, Yu X. Advances in Pharmacological Activities of Terpenoids. Natural Product Communications. 2020;15(3):1934578X20903555. [View at Publisher] [DOI] [PMID] [Google Scholar]
77. Kim T, Song B, Cho KS, Lee I-S. Therapeutic potential of volatile terpenes and terpenoids from forests for inflammatory diseases. International journal of molecular sciences. 2020;21(6):2187. [View at Publisher] [DOI] [PMID] [Google Scholar]
78. Deepak Kumar D, Chandra Kishore T, Anil Kumar S, Vaibhav T. Revisiting the Medicinal Value of Terpenes and Terpenoids. In: Vijay Singh M, Hanuman Prasad P, Sunita Kumari M, editors. Revisiting Plant Biostimulants. Rijeka: IntechOpen; 2022. p.Ch.5. [View at Publisher] [DOI] [PubMed] [Google Scholar]
79. Putta DS, Yarla Ns, Kilari E, Surekha C, Aliev G, Basavaraju D, et al. Therapeutic Potentials of Triterpenes in Diabetes and its Associated Complications. Current Topics in Medicinal Chemistry. 2016;16:Epub ahead of print. [View at Publisher] [DOI] [PMID] [Google Scholar]
80. Song B-R, Alam MB, Lee S-H. Terpenoid-Rich Extract of Dillenia indica L. Bark Displays Antidiabetic Action in Insulin-Resistant C2C12 Cells and STZ-Induced Diabetic Mice by Attenuation of Oxidative Stress. Antioxidants [Internet]. 2022;11(7). [View at Publisher] [DOI] [PMID] [Google Scholar]
81. SmithaGrace SR, Chandran G, Chauhan JB. Terpenoids: An Activator of "Fuel-Sensing Enzyme AMPK" with Special Emphasis on Antidiabetic Activity. In: Ozturk M, Hakeem KR, editors. Plant and Human Health, Volume 2: Phytochemistry and Molecular Aspects. Cham: Springer International Publishing; 2019. p.227-44. [View at Publisher] [DOI] [PMID] [Google Scholar]
82. Germoush MO, Elgebaly HA, Hassan S, Kamel EM, Bin-Jumah M, Mahmoud AM. Consumption of Terpenoids-Rich Padina pavonia Extract Attenuates Hyperglycemia, Insulin Resistance and Oxidative Stress, and Upregulates PPARγ in a Rat Model of Type 2 Diabetes. Antioxidants [Internet]. 2020; 9(1). [View at Publisher] [DOI] [PMID] [Google Scholar]
83. Gomaa AA, Makboul RM, El-Mokhtar MA, Abdel-Rahman EA, Ahmed IA, Nicola MA. Terpenoid-rich Elettaria cardamomum extract prevents Alzheimer-like alterations induced in diabetic rats via inhibition of GSK3β activity, oxidative stress and pro-inflammatory cytokines. Cytokine. 2019;113:405-16. [View at Publisher] [DOI] [PMID] [Google Scholar]
84. Valdés M, Calzada F, Mendieta-Wejebe JE, Merlín-Lucas V, Velázquez C, Barbosa E. Antihyperglycemic Effects of Annona diversifolia Safford and Its Acyclic Terpenoids: α-Glucosidase and Selective SGLT1 Inhibitiors. Molecules [Internet]. 2020;25(15). [View at Publisher] [DOI] [PMID] [Google Scholar]
85. Ahmed QU, Hasan M, Siddiqui MJ. ID 82. In vitro Adipogenic Potential and Glucose Uptake Stimulatory Effect of the Terpenoids Isolated from Tetracera indica Merr. Journal of Pharmacy & Bioallied Sciences. 2020;12. [View at Publisher] [DOI] [PubMed] [Google Scholar]
86. Sallau AB, Yakubu RN, Abdullahi SM, Salihu A, Boniface BY. In vitro effect of terpenoids - rich extract of momordica charantia on alpha glucosidase activity. Vitae. 2018;25:148-53. [View at Publisher] [DOI] [PMID] [Google Scholar]
87. Anitha K, Mohana Lakshmi S, Satyanarayana SV. Antidiabetic, lipid lowering and antioxidant potentiating effect of Canavalia species in high fat diet-streptozotocin induced model. Advances in Traditional Medicine. 2020;20(4):609-18. [View at Publisher] [DOI] [PMID] [Google Scholar]
88. 88.Nair ANS, Nair RVR, Nair APR, Nair AS, Thyagarajan S, Johnson AJ, et al. Antidiabetes constituents, cycloartenol and 24-methylenecycloartanol, from Ficus krishnae. PloS One. 2020;15(6):e0235221. [View at Publisher] [DOI] [PMID] [Google Scholar]
89. Shen N, Wang T, Gan Q, Liu S, Wang L, Jin B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chemistry. 2022;383:132531. [View at Publisher] [DOI] [PMID] [Google Scholar]
90. Donadio G, Mensitieri F, Santoro V, Parisi V, Bellone ML, De Tommasi N, et al. Interactions with microbial proteins driving the antibacterial activity of flavonoids. Pharmaceutics. 2021;13(5):660. [View at Publisher] [DOI] [PMID] [Google Scholar]
91. Roy A, Datta S, Bhatia KS, Jha P, Prasad R. Role of plant derived bioactive compounds against cancer. South African Journal of Botany. 2021. [View at Publisher] [DOI] [PMID] [Google Scholar]
92. Shamsudin NF, Ahmed QU, Mahmood S, Shah SA, Sarian MN, Khattak MM, et al. Flavonoids as Antidiabetic and Anti-Inflammatory Agents: A Review on Structural Activity Relationship-Based Studies and Meta-Analysis. International Journal of Molecular Sciences [Internet]. 2022; 23(20). [View at Publisher] [DOI] [PMID] [Google Scholar]
93. Dokumacioglu E, Iskender H, Musmul A. Effect of hesperidin treatment on α-Klotho/FGF-23 pathway in rats with experimentally-induced diabetes. Biomedicine and Pharmacotherapy. 2019;109:1206-10. [View at Publisher] [DOI] [PMID] [Google Scholar]
94. Singh AK, Raj V, Keshari AK, Rai A, Kumar P, Rawat A, et al. Isolated mangiferin and naringenin exert antidiabetic effect via PPARγ/GLUT4 dual agonistic action with strong metabolic regulation. Chemico-Biological Interactions. 2018;280:33-44. [View at Publisher] [DOI] [PMID] [Google Scholar]
95. Al-Dosari DI, Ahmed MM, Al-Rejaie SS, Alhomida AS, Ola MS. Flavonoid Naringenin Attenuates Oxidative Stress, Apoptosis and Improves Neurotrophic Effects in the Diabetic Rat Retina. Nutrients [Internet]. 2017;9(10). [View at Publisher] [DOI] [PMID] [Google Scholar]
96. Hameed A, Hafizur RM, Hussain N, Raza SA, Rehman M, Ashraf S, et al. Eriodictyol stimulates insulin secretion through cAMP/PKA signaling pathway in mice islets. European Journal of Pharmacology. 2018;820:245-55. [View at Publisher] [DOI] [PMID] [Google Scholar]
97. Lv P, Yu J, Xu X, Lu T, Xu F. Eriodictyol inhibits high glucose-induced oxidative stress and inflammation in retinal ganglial cells. Journal of Cellular Biochemistry. 2019;120(4):5644-51. [View at Publisher] [DOI] [PMID] [Google Scholar]
98. Wang N, Yi WJ, Tan L, Zhang JH, Xu J, Chen Y, et al. Apigenin attenuates streptozotocin-induced pancreatic β cell damage by its protective effects on cellular antioxidant defense. In Vitro Cellular & Developmental Biology - Animal. 2017;53(6):554-63. [View at Publisher] [DOI] [PMID] [Google Scholar]
99. Al-Ishaq RK, Abotaleb M, Kubatka P, Kajo K, Büsselberg D. Flavonoids and Their Anti-Diabetic Effects: Cellular Mechanisms and Effects to Improve Blood Sugar Levels. Biomolecules [Internet]. 2019;9(9). [View at Publisher] [DOI] [PMID] [Google Scholar]
100. Yao Z, Gu Y, Zhang Q, Liu L, Meng G, Wu H, et al. Estimated daily quercetin intake and association with the prevalence of type 2 diabetes mellitus in Chinese adults. European Journal of Nutrition. 2019;58(2):819-30. [View at Publisher] [DOI] [PMID] [Google Scholar]
101. Bule M, Abdurahman A, Nikfar S, Abdollahi M, Amini M. Antidiabetic effect of quercetin: A systematic review and meta-analysis of animal studies. Food and Chemical Toxicology. 2019;125:494-502. [View at Publisher] [DOI] [PMID] [Google Scholar]
102. Eitah HE, Maklad YA, Abdelkader NF, Gamal el Din AA, Badawi MA, Kenawy SA. Modulating impacts of quercetin/sitagliptin combination on streptozotocin-induced diabetes mellitus in rats. Toxicology and Applied Pharmacology. 2019;365:30-40. [View at Publisher] [DOI] [PMID] [Google Scholar]
103. Ghorbani A. Mechanisms of antidiabetic effects of flavonoid rutin. Biomedicine and Pharmacotherapy. 2017;96:305-12. [View at Publisher] [DOI] [PMID] [Google Scholar]
104. Alkhalidy H, Moore W, Wang Y, Luo J, McMillan RP, Zhen W, et al. The Flavonoid Kaempferol Ameliorates Streptozotocin-Induced Diabetes by Suppressing Hepatic Glucose Production. Molecules [Internet]. 2018; 23(9). [View at Publisher] [DOI] [PMID] [Google Scholar]
105. Sharma D, Gondaliya P, Tiwari V, Kalia K. Kaempferol attenuates diabetic nephropathy by inhibiting RhoA/Rho-kinase mediated inflammatory signalling. Biomedicine and Pharmacotherapy. 2019;109:1610-9. [View at Publisher] [DOI] [PMID] [Google Scholar]
106. Althunibat OY, Al Hroob AM, Abukhalil MH, Germoush MO, Bin-Jumah M, Mahmoud AM. Fisetin ameliorates oxidative stress, inflammation and apoptosis in diabetic cardiomyopathy. Life Sciences. 2019;221:83-92. [View at Publisher] [DOI] [PMID] [Google Scholar]
107. Baek Y, Lee MN, Wu D, Pae M. Luteolin Improves Insulin Resistance in Postmenopausal Obese Mice by Altering Macrophage Polarization (FS12-01-19). Current Developments in Nutrition. 2019;3(Supplement_1):nzz049.FS012-001-019. [View at Publisher] [DOI] [Google Scholar]
108. Liu Y, Han J, Zhou Z, Li D. Tangeretin inhibits streptozotocin-induced cell apoptosis via regulating NF-κB pathway in INS-1 cells. Journal of Cellular Biochemistry. 2019;120(3):3286-93. [View at Publisher] [DOI] [PMID] [Google Scholar]
109. Hsu C-C, Lin MH, Cheng J-T, Wu MC. Diosmin, a Citrus Nutrient, Activates Imidazoline Receptors to Alleviate Blood Glucose and Lipids in Type 1-Like Diabetic Rats. Nutrients [Internet]. 2017;9(7). [View at Publisher] [DOI] [PMID] [Google Scholar]
110. Yang Z, Huang W, Zhang J, Xie M, Wang X. Baicalein improves glucose metabolism in insulin resistant HepG2 cells. European Journal of Pharmacology. 2019;854:187-93. [View at Publisher] [DOI] [PMID] [Google Scholar]
111. Ma L, Li XP, Ji HS, Liu YF, Li EZ. Baicalein protects rats with diabetic cardiomyopathy against oxidative stress and inflammation injury via phosphatidylinositol 3-kinase (PI3K)/AKT pathway. Medical science monitor: international medical journal of experimental and clinical research. 2018;24:5368. [View at Publisher] [DOI] [PMID] [Google Scholar]
112. Yin H, Huang L, Ouyang T, Chen L. Baicalein improves liver inflammation in diabetic db/db mice by regulating HMGB1/TLR4/NF-κB signaling pathway. International Immunopharmacology. 2018;55:55-62. [View at Publisher] [DOI] [PMID] [Google Scholar]
113. Chattopadhyay RR. A comparative evaluation of some blood sugar lowering agents of plant origin. Journal of ethnopharmacology. 1999 Nov 30;67(3):367-72. [View at Publisher] [DOI] [PMID] [Google Scholar]
114. Yao XG, Chen F, Li P, Quan L, Chen J, Yu L, Ding H, Li C, Chen L, Gao Z, Wan P. Natural product vindoline stimulates insulin secretion and efficiently ameliorates glucose homeostasis in diabetic murine models. Journal of ethnopharmacology. 2013 Oct 28;150(1):285-97. [View at Publisher] [DOI] [PMID] [Google Scholar]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 4.0 | Journal of Clinical and Basic Research

Designed & Developed by : Yektaweb

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0).