Table of Contents - Issue
Recent articles
-
Potential Role of Flavonoids as Anti-diabetic Agents-A Comprehensive ReviewAuthor: Sridevi GopathyDOI: 10.21522/TIJPH.2013.SE.24.03.Art001
Potential Role of Flavonoids as Anti-diabetic Agents-A Comprehensive Review
Abstract:
Diabetes mellitus is a widespread and debilitating metabolic disorder marked by sustained elevated blood glucose levels, which can culminate in a range of severe complications if unmanaged. Flavonoids, polyphenolic chemicals originating from plants, have attracted a great deal of attention in the field of diabetes research because of their antidiabetic properties. These naturally occurring substances, which are 15 carbons in structure, are widely distributed in fruits, vegetables, and other plant-based diets, have been shown to offer a number of positive benefits, including the capacity to regulate many facets of insulin and glucose homeostasis. These compounds are classified into six major subclasses based on their structural differences. Numerous in vivo and in vitro studies have looked at the antidiabetic potential of flavonoids. Flavonoids have been found to modulate enzymes such as ά-glucosidase and ά-amylase, which are the key enzymes for the reduction of blood glucose levels. Emerging evidence suggests that flavonoids may exert their antidiabetic effects through their ability to modulate various cell signaling pathways involved in glucose metabolism, insulin sensitivity, and inflammation. It has been demonstrated that flavonoids contain anti-inflammatory and antioxidant qualities. These qualities are vital for reducing inflammation and oxidative stress, which are crucial factors to the onset of diabetes. This review aims providing comprehensive elucidation of the cellular and molecular mechanisms underlying the antidiabetic effects of flavonoids, considering their potential impact on various metabolic pathways involved in diabetes.
Potential Role of Flavonoids as Anti-diabetic Agents-A Comprehensive Review
References:
[1] Choudhury, H., Pandey, M., Hua, C. K., Mun, C. S., Jing, J. K., Kong, L., Ern, L. Y., Ashraf, N. A., Kit, S. W., Yee, T. S., Pichika, M. R., Gorain, B., & Kesharwani, P., 2018, An update on natural compounds in the remedy of diabetes mellitus: A systematic review. Journal of Traditional and Complementary Medicine, 8(3): 361–376. https://doi.org/10.1016/j.jtcme.2017.08.012
[2] Kharroubi, A. T., 2015, Diabetes mellitus: The epidemic of the century. World Journal of Diabetes, 6(6): 850. https://doi.org/10.4239/wjd.v6.i6.850
[3] International Diabetes Federation, 2021, IDF Diabetes Atlas, 10th edition. https://idf.org/about-diabetes/diabetes-facts-figures/
[4] Missoun, F., 2018, Antidiabetic bioactive compounds from plants. Medical Technologies Journal, 2(2): 199–214. https://doi.org/10.26415/2572-004x-vol2iss2p199-214
[5] Caro-Ordieres, T., Marín-Royo, G., Opazo-Ríos, L., Jiménez-Castilla, L., Moreno, J. A., Gómez-Guerrero, C., & Egido, J., 2020, The coming age of flavonoids in the treatment of diabetic complications. Journal of Clinical Medicine, 9(2): 346. https://doi.org/10.3390/jcm9020346
[6] AL-Ishaq, Abotaleb, Kubatka, Kajo, & Büsselberg, 2019, Flavonoids and their anti-diabetic effects: Cellular mechanisms and effects to improve blood sugar levels. Biomolecules, 9(9): 430. https://doi.org/10.3390/biom9090430
[7] Venugopala, K. N., Chaudhary, J., Sharma, V., Jain, A., Kumar, M., Sharma, D., & et al., 2022, Exploring the potential of flavonoids as efflorescing antidiabetic: An updated SAR and mechanistic based approach. Pharmacognosy Magazine, 18(80): 791-807. http://www.phcog.com/text.asp?2022/18/80/791/357236
[8] Bai, L., Li, X., He, L., Zheng, Y., Lu, H., Li, J., Zhong, L., Tong, R., Jiang, Z., Shi, J., & Li, J., 2019, Antidiabetic potential of flavonoids from traditional Chinese medicine: A review. The American Journal of Chinese Medicine, 47(05): 933–957. https://doi.org/10.1142/s0192415x19500496
[9] Sohretoglu, D., & Sari, S., 2019, Flavonoids as alpha-glucosidase inhibitors: Mechanistic approaches merged with enzyme kinetics and molecular modelling. Phytochemistry Reviews, 19(5): 1081–1092. https://doi.org/10.1007/s11101-019-09610-6
[10] Ke, R. Q., Wang, Y., Hong, S. H., & Xiao, L. X., 2023, Anti-diabetic effect of quercetin in type 2 diabetes mellitus by regulating the microRNA-92b-3p/EGR1 axis. Journal of Physiology and Pharmacology: An Official Journal of the Polish Physiological Society, 74(2). https://doi.org/10.26402/jpp.2023.2.03
[11] Ansari, P., Choudhury, S. T., Seidel, V., Rahman, A. B., Aziz, Md. A., Richi, A. E., Rahman, A., Jafrin, U. H., Hannan, J. M. A., & Abdel-Wahab, Y. H. A., 2022, Therapeutic potential of quercetin in the management of type-2 diabetes mellitus. Life, 12(8): 1146. https://doi.org/10.3390/life12081146
[12] Yang, Y., Chen, Z., Zhao, X., Xie, H., Du, L., Gao, H., & Xie, C., 2022, Mechanisms of Kaempferol in the treatment of diabetes: A comprehensive and latest review. Frontiers in Endocrinology, 13. https://doi.org/10.3389/fendo.2022.990299
[13] Kumari, G., Nigam, V. K., & Pandey, D. M., 2022, The molecular docking and molecular dynamics study of flavonol synthase and flavonoid 3’-monooxygenase enzymes involved for the enrichment of kaempferol. Journal of Biomolecular Structure and Dynamics, 41(6): 2478–2491. https://doi.org/10.1080/07391102.2022.2033324
[14] Qaddoori, M. H., & Al-Shmgani, H. S., 2023, Galangin-Loaded gold nanoparticles: Molecular mechanisms of antiangiogenesis properties in breast cancer. International Journal of Breast Cancer, 2023: 1–14. https://doi.org/10.1155/2023/3251211
[15] Al Duhaidahawi, D., Hasan, S. A., & Al Zubaidy, H. F. S., 2021, Flavonoids in the treatment of diabetes: Clinical outcomes and mechanism to ameliorate blood glucose levels. Current Diabetes Reviews, 17(6). https://doi.org/10.2174/1573399817666201207200346
[16] Mathew, M. G., Jeevanandan, G., Vishwanathaiah, S., Hamzi, K. A., Depsh, M. A. N., & Maganur, P. C., 2022, Parental and Child Outlook on the Impact of ECC on Oral Health-related Quality of Life: A Prospective Interventional Study. The Journal of Contemporary Dental Practice, 23(9), 877–882. https://doi.org/10.5005/jp-journals-10024-3397
[17] Jayaraman, S., Natarajan, S. R., Veeraraghavan, V. P., & Jasmine, S., 2023, Unveiling the anti-cancer mechanisms of calotropin: Insights into cell growth inhibition, cell cycle arrest, and metabolic regulation in human oral squamous carcinoma cells (HSC-3). Journal of oral biology and craniofacial research, 13(6), 704–713. https://doi.org/10.1016/j.jobcr.2023.09.002
[18] Liao, Y., Hu, X., Pan, J., & Zhang, G., 2022, Inhibitory mechanism of baicalein on acetylcholinesterase: Inhibitory interaction, conformational change, and computational simulation. Foods, 11(2): 168. https://doi.org/10.3390/foods11020168
[19] Chan, S.-H., Hung, C.-H., Shih, J.-Y., Chu, P.-M., Cheng, Y.-H., Tsai, Y.-J., Lin, H.-C., & Tsai, K.-L., 2016, Baicalein is an available anti-atherosclerotic compound through modulation of nitric oxide-related mechanism under oxLDL exposure. Oncotarget, 7(28): 42881–42891. https://doi.org/10.18632/oncotarget.10263
[20] Cazarolli, L., Zanatta, L., Alberton, E., Bonorino Figueiredo, M. S., Folador, P., Damazio, R., Pizzolatti, M., & Barreto Silva, F. R., 2008, Flavonoids: Prospective drug candidates. Mini-Reviews in Medicinal Chemistry, 8(13): 1429–1440. https://doi.org/10.2174/138955708786369564
[21] Vishwanathaiah, S., Maganur, P. C., Manoharan, V., Jeevanandan, G., Hakami, Z., Jafer, M. A., Khanagar, S., & Patil, S., 2022, Does Social Media have any Influence during the COVID-19 Pandemic? An Update. The Journal of Contemporary Dental Practice, 23(3), 327–330
[22] Hanchang, W., Wongmanee, N., Yoopum, S., & Rojanaverawong, W., 2022, Protective role of hesperidin against diabetes induced spleen damage: Mechanism associated with oxidative stress and inflammation. Journal of Food Biochemistry, 46(12). https://doi.org/10.1111/jfbc.14444
[23] He, W., Wang, Y., Yang, R., Ma, H., Qin, X., Yan, M., Rong, Y., Xie, Y., Li, L., Si, J., Li, X., & Ma, K., 2022, Molecular mechanism of naringenin against high-glucose-induced vascular smooth muscle cells proliferation and migration based on network pharmacology and transcriptomic analyses. Frontiers in Pharmacology, 13. https://doi.org/10.3389/fphar.2022.862709
[24] Prasad, B.S., & Srinivasan, K.K., 2023, α-Glucosidase Inhibition and Upregulation of PPARγ by Flavonoid Naringenin from Tinospora sinensis Stem, a Possible Mechanism of Antidiabetic Activity. In: Arunachalam, K., Yang, X., Puthanpura Sasidharan, S. (eds) Natural Product Experiments in Drug Discovery. Springer Protocols Handbooks. Humana, New York, NY. https://doi.org/10.1007/978-1-0716-2683-2_18
[25] Martín, M. Á., & Ramos, S., 2021, Impact of dietary flavanols on microbiota, immunity and inflammation in metabolic diseases. Nutrients, 13(3): 850. https://doi.org/10.3390/nu13030850
[26] Ahangarpour, A., Sayahi, M., & Sayahi, M., 2019, The antidiabetic and antioxidant properties of some phenolic phytochemicals: A review study. Diabetes & Metabolic Syndrome: Clinical Research & Reviews, 13(1): 854-857. https://doi.org/10.1016/j.dsx.2018.11.051
[27] Xu, Z., Ma, G., Zhang, Q., Chen, C., He, Y., Xu, L., Zhou, G., Li, Z., Yang, H., & Zhou, P., 2017, Inhibitory mechanism of epigallocatechin gallate on fibrillation and aggregation of amidated human islet amyloid polypeptide. ChemPhysChem, 18(12): 1611-1619. https://doi.org/10.1002/cphc.201700057
[28] Wang, W., Zhang, Y., Jin, W., Xing, Y., & Yang, A., 2018, Catechin weakens diabetic retinopathy by inhibiting the expression of NF-κB signaling pathway-mediated inflammatory factors. Annals of Clinical and Laboratory Science, 48(5): 594-600.
[29] Wen, L., Wu, D., Tan, X., Zhong, M., Xing, J., Li, W., Li, D., & Cao, F., 2022, The Role of Catechins in regulating diabetes: An update review. Nutrients, 14(21): 4681. https://doi.org/10.3390/nu14214681
[30] Jayaraman, S., Raj Natarajan, S., Ponnusamy, B., Veeraraghavan, V. P., & Jasmine, S., 2023, Unlocking the potential of beta sitosterol: Augmenting the suppression of oral cancer cells through extrinsic and intrinsic signalling mechanisms. The Saudi Dental Journal, 35(8), 1007–1013. https://doi.org/10.1016/j.sdentj.2023.08.003
[31] Kuryłowicz, A., 2020, The role of isoflavones in type 2 diabetes prevention and treatment—a narrative review. International Journal of Molecular Sciences, 22(1): 218. https://doi.org/10.3390/ijms22010218
[32] Hussain, H., & Green, I. R., 2017, A patent review of the therapeutic potential of isoflavones (2012-2016). Expert Opinion on Therapeutic Patents, 27(10): 1135-1146. https://doi.org/10.1080/13543776.2017.1339791
[33] S, D. P. A., Solete, P., Jeevanandan, G., Syed, A. A., Almahdi, S., Alzhrani, M., Maganur, P. C., & Vishwanathaiah, S., 2023, Effect of Various Irrigant Activation Methods and Its Penetration in the Apical Third of Root Canal-In Vitro Study. European Journal of dentistry, 17(1), 57–61. https://doi.org/10.1055/s-0041-1742122.
[34] Abbasi, E., & Khodadadi, I., 2023, Antidiabetic effects of genistein: Mechanism of action. Endocrine, Metabolic & Immune Disorders - Drug Targets, 23(13): 1599-1610. https://doi.org/10.2174/1871530323666230516103420
[35] Zhang, N., Zhang, W., Guo, X., Liu, J., Li, S., Zhang, H., & Fan, B., 2022, Genistein protects against hyperglycemia and fatty liver disease in diet-induced prediabetes mice via activating hepatic insulin signaling pathway. Frontiers in Nutrition, 9. https://doi.org/10.3389/fnut.2022.1072044
[36] Liu, Y., Wang, Q., Wu, K., Sun, Z., Tang, Z., Li, X., & Zhang, B., 2022, Anthocyanins’ effects on diabetes mellitus and islet transplantation. Critical Reviews in Food Science and Nutrition, 63(33): 12102-12125. https://doi.org/10.1080/10408398.2022.2098464
[37] Pazhani, J., Jayaraman, S., Veeraraghavan, V. P., & Jasmine, S., 2024, Endothelin-1 axis as a therapeutic target in oral squamous cell carcinoma: Molecular insights. Journal of stomatology, oral and maxillofacial surgery, 125(6), 101792. Advance online publication. https://doi.org/10.1016/j.jormas.2024.101792
[38] Yang, L., Qiu, Y., Ling, W., Liu, Z., Yang, L., Wang, C., Peng, X., Wang, L., & Chen, J., 2020, Anthocyanins regulate serum adipsin and visfatin in patients with prediabetes or newly diagnosed diabetes: A randomized controlled trial. European Journal of Nutrition, 60(4): 1935-1944. https://doi.org/10.1007/s00394-020-02379-x
[39] Lai, D., Huang, M., Zhao, L., Tian, Y., Li, Y., Liu, D., Wu, Y., & Deng, F., 2019, Delphinidin-induced autophagy protects pancreatic β cells against apoptosis resulting from high-glucose stress via AMPK signaling pathway. Acta Biochimica et Biophysica Sinica, 51(12): 1242-1249. https://doi.org/10.1093/abbs/gmz126
[40] Ahmed, Q. U., Ali, A. H. M., Mukhtar, S., Alsharif, M. A., Parveen, H., Sabere, A. S. M., Nawi, M. S. Mohd., Khatib, A., Siddiqui, M. J., Umar, A., & Alhassan, A. M., 2020, Medicinal potential of isoflavonoids: Polyphenols that may cure diabetes. Molecules, 25(23): 5491. https://doi.org/10.3390/molecules25235491
Viewed PDF 160 48 -
Antidiabetic Activity of Allin Isolated from Allium Sativum: Role of P13K/AKT SignallingAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art002
Antidiabetic Activity of Allin Isolated from Allium Sativum: Role of P13K/AKT Signalling
Abstract:
This study presents a comprehensive molecular docking analysis aimed at elucidating the potential antidiabetic activity of allin, a compound isolated from Allium sativum (garlic). Through computational modelling, we investigated the binding interactions between allin and Rheb, revealing that allin exhibited the lowest binding affinities with a binding energy of -3.24 kcal/mol. The docking results unveiled a significant role of the P13K/AKT signalling pathway in mediating the antidiabetic effects of allin. The interaction between allin and Rheb was characterized by the establishment of a single hydrogen bond involving SER-16 and GDP-201. This interaction contributed to the formation of a binding pocket encompassing key residues such as PRO-37, ARG-15, SER-16, THR-88, LEU-123, GLU-126, and GDP-201. The molecular docking analysis sheds light on the intricate molecular mechanisms underlying the antidiabetic potential of allin, providing insights into its specific interactions with key signalling components. Furthermore, our findings suggest a potential modulation of the P13K/AKT signalling pathway by the allin, emphasizing its significance in the context of antidiabetic activity. The results of this study suggest contributes valuable information to the understanding of the molecular basis of allin's therapeutic potential and provides a foundation for further experimental validations and exploration of its application in diabetes management.
Antidiabetic Activity of Allin Isolated from Allium Sativum: Role of P13K/AKT Signalling
References:
[1] Bayan, L., Koulivand, P. H., & Gorji, A., 2014, Garlic: A review of its medicinal properties. Phytotherapy Research. PMID: 25050296[2] El-Saber Batiha, G., Magdy Beshbishy, A.,Wasef, G. L., Elewa, Y. H. A., Al-Sagan, A. A., Abd El-Hack, M. E., Taha, A. E., M Abd-Elhakim, Y., & Prasad Devkota, H., 2020, Chemical Constituents and Pharmacological Activities of Garlic. Phytochemistry Reviews. 10.3390/nu12030872[3] Jiang, Y., Yue, R., Liu, G., & Liu, J., 2022, Garlic (Allium sativum L.) in diabetes and its complications: Recent advances in mechanisms of action. 10.1080/10408398.2022.2153793[4] Thomson, M., Al-Amin, Z.M., Al-Qattan, K.K., Shaban, L.H., & Ali, M., 2008, Anti-diabetic and hypolipidaemic properties of garlic (Allium sativum) in streptozotocin-induced diabetic rats. International Journal of Diabetes and Metabolism. 10.1159/000497643[5] Eidi, A., Eidi, M., & Esmaeili, E., 2006, Antidiabetic effect of garlic (Allium sativum L.) in normal and streptozotocin-induced diabetic rats. 10.1016/j.phymed.2005.09.010[6] Sreevarun, M., Ajay, R., Suganya, G., Rakshagan, V., Bhanuchander, V., & Suma, K., 2023, Formulation, Configuration, and Physical Properties of Dental Composite Resin Containing a Novel 2π + 2π Photodimerized Crosslinker - Cinnamyl Methacrylate: An In Vitro Research. The Journal of Contemporary Dental Practice, 24(6), 364–371. https://doi.org/10.5005/jp-journals-10024-3480[7] Baluchnejadmojarad, T., & Rohgani, M., 2003, Endothelium-dependent and -independent effect of aqueous extract of garlic on vascular reactivity on diabetic rats. 10.1016/s0367-326x(03)00158-8[8] Ashraf, R., Khan, RA., & Ashraf, I., 2011, Garlic (Allium sativum) supplementation with standard antidiabetic agent provides better diabetic control in type 2 diabetes patients. 24(4):565-70[9] Guo, Y., & Heindl L. M., 2023, The Role of Nutrition in the Prevention and Intervention of Type 2 Diabetes. 10.3389/fbioe.2020.575442[10] Alam, M. K., Alqhtani, N. R., Alnufaiy, B., Alqahtani, A. S., Elsahn, N. A., Russo, D., Di Blasio, M., Cicciù, M., & Minervini, G. (2024). A systematic review and meta-analysis of the impact of resveratrol on oral cancer: potential therapeutic implications. BMC Oral Health, 24(1), 412. https://doi.org/10.1186/s12903-024-04045-8[11] https://pubchem.ncbi.nlm.nih.gov/compound/6167[12] Yadalam, P. K., Arumuganainar, D., Ronsivalle, V., Di Blasio, M., Badnjevic, A., Marrapodi, M. M., Cervino, G., & Minervini, G., 2024, Prediction of interactomic hub genes in PBMC cells in type 2 diabetes mellitus, dyslipidemia, and periodontitis. BMC Oral Health, 24(1), 385. https://doi.org/10.1186/s12903-024-04041-y[13] Vo Van, L., Pham, EC., Nguyen, CV., Duong, NTN., Vi Le Thi, T., & Truong, TN., 2022, In vitro and in vivo antidiabetic activity, isolation of flavonoids, and in silico molecular docking of stem extract of Merremia tridentata (L). 10.1016/j.biopha.2021.112611[14] Jayaraman, S., Raj Natarajan, S., Ponnusamy, B., Veeraraghavan, V. P., & Jasmine, S., 2023, Unlocking the potential of beta sitosterol: Augmenting the suppression of oral cancer cells through extrinsic and intrinsic signalling mechanisms. The Saudi Dental Journal, 35(8), 1007–1013. https://doi.org/10.1016/j.sdentj.2023.08.003Huang, X., Liu, G., Guo, J., & Su, Z., 2018, The PI3K/AKT pathway in obesity and type 2 diabetes. 2018, 10.7150/ijbs.27173[15] Camaya, I., Donnelly, S., & Obrien, B., 2022, Targeting the PI3K/Akt signalling pathway in pancreatic β‐cells to enhance their survival and function: An emerging therapeutic strategy for type 1 diabetes. 2022, 10.1111/1753-0407.13252[16] Miao, R., Fang, X., Wei, J., Wu, H., Wang, X., & Tian, J., 2022, Akt: A Potential Drug Target for Metabolic Syndrome https://doi.org/10.3389/fphys.2022.822333[17] Pazhani, J., Jayaraman, S., Veeraraghavan, V. P., & Jasmine, S., 2024, Endothelin-1 axis as a therapeutic target in oral squamous cell carcinoma: Molecular insights. Journal of stomatology, oral and maxillofacial surgery, 125(6), 101792. Advance online publication. https://doi.org/10.1016/j.jormas.2024.101792[18] El-Hachem, N., Haibe-Kains, B., Khalil, A., Kobeissy, F. H., & Nemer, G., 2017, Auto Dock and AutoDockTools for Protein-Ligand Docking: Beta-Site Amyloid Precursor Protein Cleaving Enzyme 1(BACE1) as a Case Study. methmolbio-1598-391-17, 10.1007/978-1-4939-6952-420[19] Selvaraj, J., Vishnupriya, V., Sardar, H., Balakrishna, J. P., Rex, J., Mohan, S. K., Vijayalakshmi, P., Ponnulakshmi, R., 2020, Molecular docking analysis of COX-2 for potential inhibitors. 10.6026/97320630016753, 753–758Viewed PDF 118 26 -
Identification of the Role of Ajoene from Aegle marmelos Correa for its Anti-Diabetic Action: Role of NRF2/KEAP-1 SignalingAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art003
Identification of the Role of Ajoene from Aegle marmelos Correa for its Anti-Diabetic Action: Role of NRF2/KEAP-1 Signaling
Abstract:
Diabetes mellitus is a chronic metabolic disorder that affects millions of people worldwide. The management of diabetes is a significant for new and effective therapeutic agents. Ajoene, a compound found in garlic, has been shown to have anti-diabetic properties. The Nrf2/Keap-1 signaling pathway has been identified as a potential target for the treatment of diabetes. The aim of this study is to investigate the role of ajoene from Aegle Marmelos Correa in the activation of the Nrf2/Keap-1 signaling pathway and its potential as an anti-diabetic agent. In silico analysis was performed using molecular docking and molecular dynamics simulations to investigate the interaction between ajoene and the Nrf2/Keap-1 signaling pathway. The molecular docking studies revealed that ajoene binds to the Nrf2/Keap-1 complex with high affinity, indicating a potential interaction between ajoene and Cul3 was establishment of a single hydrogen bond involving ILE258. This interaction contributed to the formation of a binding pocket encompassing key residues such as LEU-253, ILE-258, VAL-260, LEU-266, LEU-292. These findings suggest that ajoene may activate the Nrf2/Keap-1 signaling pathway, leading to the upregulation of antioxidant genes and the inhibition of oxidative stress, which are known to contribute to the development of diabetes. The results of this study suggest that ajoene from Aegle Marmelos Correa may have potential as anti-diabetic agent through its activation of the Nrf2/Keap-1 signaling pathway. The specific interaction between ajoene and Cul3, characterized by the establishment of a single hydrogen bond involving ILE258, contributes to the formation of a binding pocket encompassing key residues.
Identification of the Role of Ajoene from Aegle marmelos Correa for its Anti-Diabetic Action: Role of NRF2/KEAP-1 Signaling
References:
[1] Gupta, R. K., Kesari, A. N., Watal, G., Murthy, P. S., Chandra, R., & Tandon, V., 2005, Hypoglycemic and antidiabetic effect of aqueous extract of leaves of Annona squamosa (L.) in experimental animals. Current Science, 1246-1254
[2] Bhatti, R., Sharma, S., Kumar, A., & Singh, A., 2013, Anti adipogenic activity of Aegle marmelos Correa. Pharmacognosy Magazine, 9(34), 132
[3] Hattori, A., Yamada, N., Nishikawa, T., Fukuda, H., & Fujino, T., 2005, Antidiabetic effects of ajoene in genetically diabetic KK-Ay mice. Journal of Nutritional Science and Vitaminology, 51(5), 382-384
[4] Kesari, A. N., Gupta, R. K., Singh, S. K., Diwakar, S., & Watal, G., 2006, Hypoglycemic and antihyperglycemic activity of Aegle marmelos seed extract in normal and diabetic rats. Journal of Ethnopharmacology, 107(3), 374-379
[5] Tebay, L. E., Robertson, H., Durant, S. T., Vitale, S. R., Penning, T. M., Dinkova-Kostova, A. T., & Hayes, J. D., 2015, Mechanisms of activation of the transcription factor Nrf2 by redox
[6] Kensler, T. W., Wakabayashi, N., & Biswal, S., 2007, Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu. Rev. Pharmacol. Toxicol., 47, 89-116
[7] Narendhirakannan, R. T., Subramanian, S., & Kandaswamy, M., 2006, Biochemical evaluation of antidiabetogenic properties of some commonly used Indian plants on streptozotocin-induced diabetes in experimental rats. Clinical and Experimental Pharmacology and Physiology, 33(12), 1150-1157
[8] International Diabetes Federation., 2019, IDF Diabetes Atlas, 9th edn. Brussels, Belgium: International Diabetes Federation
[9] Kamalakkann, N., Rajadurai, M., & Prince, P. S., 2003, Effect of Aegle marmelos fruits on normal and streptozotocin-diabetic Wistar rats. Journal of Medicinal Food, 6(2), 93-98
[10] Hattori, A., Yamada, N., Nishikawa, T., Fukuda, H., & Fujino, T., 2005, Antidiabetic effects of ajoene in genetically diabetic KK-Ay mice. Journal of Nutritional Science and Vitaminology, 51(5), 382-384
[11] Zheng, H., Whitman, S. A., Wu, W., Wondrak, G. T., Wong, P. K., Fang, D., & Zhang, D. D., 2011, Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy. Diabetes, 60(11), 3055-3066
[12] Mathew, M. G., Jeevanandan, G., Vishwanathaiah, S., Hamzi, K. A., Depsh, M. A. N., & Maganur, P. C., (2022). Parental and Child Outlook on the Impact of ECC on Oral Health-related Quality of Life: A Prospective Interventional Study. The journal of Contemporary Dental Practice, 23(9), 877–882. https://doi.org/10.5005/jp-journals-10024-3397
[13] Itoh, K., Tong, K. I., & Yamamoto, M., 2004, Molecular mechanism activating Nrf2-Keap1 pathway in regulation of adaptive response to electrophiles. Free Radical Biology and Medicine, 36(10), 1208-1213
[14] Tan, Y., Ichikawa, T., Li, J., Si, Q., Yang, H., Chen, X., & Cui, T., 2011, Diabetic downregulation of Nrf2 activity via ERK contributes to oxidative stress-induced insulin resistance in cardiac cells in vitro and in vivo. Diabetes, 60(2), 625-633
[15] Paulose, C. S., Dakshinamurti, K., Packer, S., & Stephens, N. L., 1988, Sympathetic stimulation and hypertension in the Nrf2-knockout mouse. Hypertension, 53(4), 659-665
[16] Krishnan, R. P., Pandiar, D., & Ramani, P., 2023, Sclerosing Variant of Adenoid Cystic Carcinoma - A Case Report on the Role of Sclerosis in the Prognostic Outcome. Annals of Maxillofacial Surgery, 13(2), 248–251. https://doi.org/10.4103/ams.ams_116_23
[17] Kesari, A. N., Gupta, R. K., Singh, S. K., Diwakar, S., & Watal, G., 2006, Hypoglycemic and antihyperglycemic activity of Aegle marmelos seed extract in normal and diabetic rats. Journal of Ethnopharmacology, 107(3), 374-379
[18] https://pubchem.ncbi.nlm.nih.gov/compound/5386591
[19] Gopalakrishnan, U., Felicita, A. S., Qureshi, T., Muruganandhan, J., Hassan, A. A. A., El-Shamy, F. M., Osman, H. A., Medabesh, A. A., & Patil, S., 2022, Effect of Fluoridated Mouthwashes on Corrosion Property of Orthodontic Appliances: A Narrative Review. The Journal of Contemporary Dental Practice, 23(4), 460–466.
[20] Ulasov, A. V., Rosenkranz, A. A., Georgiev, G. P., & Soboleva, A. S., 2022, Nrf2/Keap1/ARE signaling: Towards specific regulation doi: 10.1016/j.lfs.2021.120111
[21] Furukawa, M., He, Y.J., Borchers, C., & Xiong, Y., 2003, Targeting of protein ubiquitination by BTB-Cullin 3-Roc1 ubiquitin ligases DOI: 10.1038/ncb1056
[22] Jayaraman, S., Natarajan, S. R., Veeraraghavan, V. P., & Jasmine, S., 2023, Unveiling the anti-cancer mechanisms of calotropin: Insights into cell growth inhibition, cell cycle arrest, and metabolic regulation in human oral squamous carcinoma cells (HSC-3). Journal of oral biology and craniofacial research, 13(6), 704–713. https://doi.org/10.1016/j.jobcr.2023.09.002
[23] Tiwari, R., Mishra, S., Danaboina, G., Pratap Singh Jadaun, G., Kalaivani, M., Kalaiselvan, V., Dhobi, M., & Raghuvanshi, R.S., 2023, Comprehensive chemo-profiling of coumarins enriched extract derived from Aegle marmelos (L.) Correa fruit pulp, as an anti-diabetic and anti-inflammatory agent Doi: 10.1016/j.jsps.2023.101708.
[24] Sharma, P., Joshi , T., Mathpal , S., Chandra, S., & Tamta, S., 2021, In silico identification of antidiabetic target for phytochemicals of A. marmelos and mechanistic insights by molecular dynamics simulations. Doi:10.1080/07391102.2021.1944910
Viewed PDF 85 11 -
Acacetin Interacts with Glycolytic Enzymes and Inhibits DiabetesAuthor: Ponnulakshmi RajagopalDOI: 10.21522/TIJPH.2013.SE.24.03.Art004
Acacetin Interacts with Glycolytic Enzymes and Inhibits Diabetes
Abstract:
Diabetes mellitus, a chronic metabolic disorder, has been a global health concern with rising prevalence. The search for novel therapeutic agents, especially from natural sources, remains a priority. Acacetin, a flavonoid found in various plants, has shown potential in various biological activities. However, its role in diabetes management, particularly its interaction with glycolytic enzymes, has been less explored. This study aimed to investigate the interaction of acacetin with glycolytic enzymes and its potential as a therapeutic agent for diabetes management. A comprehensive molecular docking analysis was employed to explore the binding affinity of acacetin to glycolytic enzymes, including hexokinase, phosphofructokinase, and pyruvate kinase. The study utilized advanced computational tools and techniques to simulate the interaction dynamics. The binding energy, interaction sites, and stability of the acacetin-enzyme complex were evaluated. Acacetin exhibited significant binding affinity towards all three glycolytic enzymes, with notable stability in the enzyme active sites. The binding energies indicated a strong interaction, suggesting potential inhibitory effects on the enzymes. The interaction was characterized by both hydrogen bonding and hydrophobic interactions, contributing to the stability of the complexes. The molecular docking analysis suggests that acacetin interacts effectively with key glycolytic enzymes, potentially inhibiting their activity. This interaction could impede the glycolytic pathway, which is crucial in diabetes pathophysiology. Therefore, acacetin emerges as a promising candidate for diabetes management, warranting further in-vitro and in-vivo studies to explore its therapeutic potential and mechanism of action.
Acacetin Interacts with Glycolytic Enzymes and Inhibits Diabetes
References:
[1] Roden M., & Shulman G.I., 2019, The integrative biology of type 2 diabetes. Nature. 576:51–60. doi: 10.1038/s41586-019-1797-8.[2] Mali, A. V., Bhise, S. S., Hegde, M. V., & Katyare, S. S., 2016, Altered Erythrocyte Glycolytic Enzyme Activities in Type-II Diabetes. Indian J Clin Biochem. 2016 Jul; 31[3]: 321–325. PMID: 27382204. doi: 10.1007/s12291-015-0529-6.[3] Rabbani, N., Xue, M., & Thornalley, P. J., 2022, Hexokinase-2-Linked Glycolytic Overload and Unscheduled Glycolysis—Driver of Insulin Resistance and Development of Vascular Complications of Diabetes. doi: 10.3390/ijms23042165.[4] Gordin, D., Shah, H., Shinjo, T., Louis, R. S., Qi, W., Park, K., Paniagua, S. M., Pober, D. M., Wu, I., Bahnam, V., Brissett, M. J., Tinsley, L. J., Dreyfuss, J. M., Pan, H., Dong, Y., Niewczas, M. A., Amenta, P., Sadowski, T., Kannt, A., Keenan, H. A., & King, G. L., 2019, Characterization of Glycolytic Enzymes and Pyruvate Kinase M2 in Type 1 and 2 Diabetic Nephropathy. Diabetes Care. Jul; 42[7]: 1263–1273. PMID: 31076418. doi: 10.2337/dc18-2585.[5] Guo, X., Li, H., Xu, H., Woo, S., Dong, H., Lu, F., Lange, A. J., Wu, C., 2012, Glycolysis in the control of blood glucose homeostasis. Acta Pharmaceutica Sinica B Volume 2, Issue 4, Pages 358-367. https://doi.org/10.1016/j.apsb.2012.06.002.[6] Haythorne, H., Rohm, M., Bunt, M. V. D., Brereton, M. F., Tarasov, A. I., Blacker, T. S., Sachse, G., Santos, M. S. D., Exposito, R. T., Davis, S., Baba, O., Fischer, R., Duchen, M. R., Rorsman, P., MacRae, J. I., & Ashcroft, F. M., 2019, Diabetes causes marked inhibition of mitochondrial metabolism in pancreatic β-cells, 2019. https://doi.org/10.1038/s41467-019-10189-x.[7] Liu, Q. J., Yuan, W., Yang, P., Liu Q. J., Yuan, W., Yang, P., & Shao, C., 2023, Role of glycolysis in diabetic atherosclerosis. World J Diabetes 14[10]: 1478-1492. DOI: 10.4239/wjd.v14.i10.1478. PMID: 37970130.[8] Haythorne, E., Lloyd, M., Tickle, J. W., Tarasov, A. I., Sandbrink, J., Portillo, I., Exposito, R. T., Sachse, G, Cyranka, M., Rohm, R., Rorsman, P., McCullagh, J., & Ashcroft, F. M., 2022, Altered glycolysis triggers impaired mitochondrial metabolism and mTORC1 activation in diabetic β-cells. Nature communications. https://doi.org/10.1038/s41467-022-34095-x.[9] Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., Goodsell, D. S. and Olson, A. J., 2009, Autodock4 and AutoDockTools4: automated docking with selective receptor flexiblity. J. Computational Chemistry 2009, 16: 2785-91.[10] Meng, X. Y., Zhang, H. X., Mezei, M., & Cui, M., 2011, Molecular Docking: A powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des. 2011 Jun 1; 7[2]: 146–157. doi: 10.2174/157340911795677602. PMID: 21534921[11] Varghese, R. M., Kumar, S. A., & Selvaraj, Y., 2023, Assessment of Soft Tissue, Airway Dimension and Hyoid Bone Position in Class II Patients Treated by PowerScope Class 2 Corrector. The Journal of Contemporary Dental Practice, 24(5), 308–313. https://doi.org/10.5005/jp-journals-10024-3485.[12] Iizuka, K., 2023, Recent Progress on Fructose Metabolism—Chrebp, Fructolysis, and Polyol Pathway. Nutrients 2023, 15[7], 1778; https://doi.org/10.3390/nu15071778.[13] Pirovich, D. B., Da’dara, A. A., & Skelly, P. J., 2021, Multifunctional Fructose 1,6-Bisphosphate Aldolase as a Therapeutic Target. Front Mol Biosci. 2021; 8: 719678. PMID: 34458323. doi: 10.3389/fmolb.2021.719678.[14] Varghese, R.M., Subramanian, A.K., Maliael, M.T., 2023, PowerScope™ for Class II Malocclusions: A Systematic Review and Meta-analysis. World Journal of Dentistry,14(7):639–647.[15] Hu, H., Zhao, M., Li, Z., Nie, H., He, J., Chen, Z., Yuan, J., Guo, H., Zhang, X., Yang, H., Wu, T., & He, M., 2022, Plasma miR-193b-3p Is Elevated in Type 2 Diabetes and Could Impair Glucose Metabolism. Front Endocrinol [Lausanne]. 2022; 13: 814347. PMID: 35712251. doi: 10.3389/fendo.2022.814347.[16] Lazarev, V. F., Guzhova, I. V., & Margulis, B. A., 2020, Glyceraldehyde-3-phosphate Dehydrogenase Is a Multifaceted Therapeutic Target. Pharmaceutics 2020, 12[5], 416; https://doi.org/10.3390/pharmaceutics12050416.[17] Sowmithra Devi, S., Sundari, S.,2023, Occlusal Contact Changes With Traumatic Occlusion After Orthodontic Treatment: A Prospective Study. Journal of Advanced Oral Research. 14(2):134-142. doi:10.1177/23202068231190202.[18] Kanwar, M., & Kowluru, R. A., 2009, Role of glyceraldehyde 3-phosphate dehydrogenase in the development and progression of diabetic retinopathy. Diabetes. 2009 Jan;58[1]:227-34. doi: 10.2337/db08-1025. PMCID: PMC2606877.[19] Bouterse, S. M., Mohammad, G., & Kowluru, R. A., 2010, Glyceraldehyde-3-Phosphate Dehydrogenase in Retinal Microvasculature: Implications for the Development and Progression of Diabetic Retinopathy. Retinal Cell Biology.[20] Didiasova, M., Schaefer, L., & Wygrecka, M., 2019, When Place Matters: Shuttling of Enolase-1 Across Cellular Compartments. Cell Dev. Biol., 26 April 2019. Molecular and Cellular Pathology, Volume 7 - 2019 | https://doi.org/10.3389/fcell.2019.00061[21] Xie, J., Du, R., Li , Q., & Li, L., 2024, The relationship between neuron-specific enolase, high sensitivity C reactive protein, and diabetic peripheral neuropathy in Chinese patients with type 2 diabetes: A prospective nested case–control analysis. Volume 44, Pages 190-199.[22] Rajala, A., Soni, K., & Rajala, R. V. S., 2020, Metabolic and Non-metabolic Roles of Pyruvate Kinase M2 Isoform in Diabetic Retinopathy. Scientific Reports volume 10, Article number: 7456.[23] Tu, C., Wang, L., & Wei, L., 2022, The Role of PKM2 in Diabetic Microangiopathy. Diabetes Metab Syndr Obes. 2022; 15: 1405–1412. PMID: 35548702. doi: 10.2147/DMSO.S366403.[24] Lee, I. K., 2014, The Role of Pyruvate Dehydrogenase Kinase in Diabetes and Obesity. Diabetes & Metabolism Journal 2014;38[3]:181-186. DOI: https://doi.org/10.4093/dmj.2014.38.3.181.[25] Park, Y. S., Han, J. H., Park, J. H., Choi, J. S., Kim, S. H., & Kim, H. S., 2023, Pyruvate Kinase M2: A New Biomarker for the Early Detection of Diabetes-Induced Nephropathy. Int. J. Mol. Sci. 24[3],2683; https://doi.org/10.3390/ijms24032683.Viewed PDF 107 10 -
Studies on the Molecular Interaction of Colchicine with Antioxidant Signaling Molecules and Identification Antidiabetic Activity: Evidences through In-silico AnalysisAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art005
Studies on the Molecular Interaction of Colchicine with Antioxidant Signaling Molecules and Identification Antidiabetic Activity: Evidences through In-silico Analysis
Abstract:
Colchicine, a naturally occurring alkaloid, has garnered attention for its potential anti-diabetic properties. This study delves into the molecular interactions between colchicine and antioxidant signaling molecules, aiming to uncover its therapeutic potential in managing diabetes. The primary aim of this research is to investigate the intricate molecular interactions between colchicine and key antioxidant signaling molecules. Additionally, through in-silico analysis, the study seeks to identify the antidiabetic activity of colchine. In this study, molecular docking simulations were employed to explore the binding affinities and interactions of colchicine with antioxidant signaling molecules like superoxide dismutase, catalase, glutathione peroxidase, Peroxiredoxin and Hemeoxygenase. The computational analysis was carried out using state-of-the-art software tools, allowing for a comprehensive assessment of potential binding energies. Furthermore, an in-silico analysis was conducted to predict colchicine’s ability to modulate key pathways related to diabetes. The findings reveal that colchicine exhibits strong binding affinities with antioxidant enzymes, suggesting its potential as an antioxidant agent. This study provides valuable insight into the molecular interactions between colchicine and antioxidant signaling molecules. The promising binding affinities and potential antidiabetic activity identified through in-silico analysis highlight colchicine as a candidate for further investigation as a therapeutic agent for diabetes. Further in vitro and in vivo experiments are warranted to validate these in-silico finding and unlock the full potential of colchicine in diabetes management.
Studies on the Molecular Interaction of Colchicine with Antioxidant Signaling Molecules and Identification Antidiabetic Activity: Evidences through In-silico Analysis
References:
[1] Naris, S., 2022, Significant Role of Free Radicals in Diabetes Mellitus. Oxidants and Antioxidants in Medical Science. Oxidants and Antioxidants in Medical Science, 11(8), 1-2.[2] Drummond, G. R., Selemidis, S., Griendling K., & Sobey, C. G., 2011, Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nature Reviews Drug Discovery, 10(6),453–471. doi: 10.1038/nrd3403.[3] Johansen, J., Harris, A. K., Rychly, D. J., & Ergul, A., 2005, Oxidative stress and the use of antioxidants in diabetes: linking basic science to clinical practice. Cardiovascular Diabetology,4(1), p. 5. doi: 10.1186/1475-2840-4-5.[4] Pacher, P., Beckman, J. S., & Liaudet, L., 2007, Nitric oxide and peroxynitrite in health and disease. Physiological Reviews, 87(1),315–424. doi: 10.1152/physrev.00029.2006.[5] Zhang, D. X., & Gutterman, D.D., 2007, Mitochondrial reactive oxygen species-mediated signaling in endothelial cells. American Journal of Physiology-Heart and Circulatory Physiology, 292(5), H2023–H2031. doi: 10.1152/ajpheart.01283.2006.[6] Goldstein, B.J., Mahadev, K., Wu, X., Zhu, L., & Motoshima, H., 2005, Role of Insulin-Induced Reactive Oxygen Species in the Insulin Signaling Pathway. Antioxid Redox Signal, 7(7-8), 1021–1031. doi: 10.1089/ars.2005.7.1021.[7] Harsha, L., & Subramanian, A. K., 02022, Comparative Assessment of pH and Degree of Surface Roughness of Enamel When Etched with Five Commercially Available Etchants: An In Vitro Study. The Journal of Contemporary Dental Practice, 23(2), 181–185.[8] Nrf2. 2021, The IUPHAR/BPS Guide to Pharmacology. https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=2605&familyId=694&familyType=TF.[9] He, L., He, T., Farrar, S., Ji, L., Liu, T., & Ma, X., 2017, Antioxidants Maintain Cellular Redox Homeostasis by Elimination of Reactive Oxygen Species. Cellular Physiology and Biochemistry, 44(2),532-553. doi: 10.1159/000485089.91.[10] Antonyuk, S.V., Strange, R.W., Marklund, S.L., & Hasnain, S.S., 2009, The Structure of Human Extracellular Copper-Zinc Superoxide Dismutase at 1.7 A Resolution: Insights into Heparin and Collagen Binding. Journal of Molecular Biology, 388, 310. PubMed: 19289127. DOI: https://doi.org/10.1016/j.jmb.2009.03.026.[11] Eletsky, A., Pulavarthi, S.V.S.R.K., Lee, D., Kohan, E., Janjua, H., Xiao, R., Acton, T.B., Everette, J.K., Montelione, G.T., & Szyperski, T., 2012, Solution NMR Structure of the DNA-Binding Domain of Human NF-E2-Related Factor 2, Northeast Structural Genomics onsortium(NESG)TargetHR3520O. https://doi.org/10.2210/pdb2LZ1/pdb.[12] Kavanagh, K.L., Johansson, C., Smee, C., Gileadi, O., Von Delft, F., & Oppermann,U., 2005, Crystal structure of the selenocysteine to glycine mutant of human glutathione peroxidase 1. https://doi.org/10.2210/pdb2F8A/pdb.[13] Pilka, E.S., Kavanagh, K.L., Cooper, C., von Delft, F., Sundstrom, M., Arrowsmith, C.A., Weigelt, J., Edwards, A., & Oppermann, U., 2007, Crystal structure of human peroxiredoxin 4 (thioredoxin peroxidase). https://doi.org/10.2210/pdb2PN8/pdb[14] Neralla, M., M, H., Preethi, A., Selvakumar, S. C., & Sekar, D., 2024, Expression levels of microRNA-7110 in oral squamous cell carcinoma. Minerva dental and oral science, 73(3), 155–160. https://doi.org/10.23736/S2724-6329.23.04801-5.[15] Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., Goodsell, & D. S. and Olson, A. J., 2009, Autodock4 and AutoDockTools4: automated docking with selective receptor flexiblity. Journal of Computational Chemistry,16, 2785-2791.[16] Smith, J. A., & Park, S., 2021, Superoxide Dismutase 1 (SOD1). In Handbook of Amyotrophic Lateral Sclerosis, (pp. 19-31), Springer.[17] Younus, H., 2018, Therapeutic potentials of superoxide dismutase. International Journal of Health Sciences (Qassim). 12(3): 88-93.[18] Lei, X.G., Zhu, J.H., Cheng, W.H., Bao, Y., Ho, Y.S., Reddi, A.R., Holmgren, A., & Arnér, E.S., 2016, Paradoxical roles of antioxidant enzymes: basic mechanisms and health implications. Physiol Rev, 96:307–364. DOI: 10.1152/physrev.00010.2014.[19] Lewis, P., Stefanovic, N., Pete, J., Calkin, A.C., Giunti, S., Thallasbonke, V., Jandeleitdahm, K.A., Allen, T.J., Kola, I., & Cooper, M.E., 2007, Lack of the antioxidant enzyme glutathione peroxidase-1 accelerates atherosclerosis in diabetic apolipoprotein E–deficient mice. Circulation, 115,2178–2187. doi: 10.1161/CIRCULATIONAHA.106.664250.[20] Ramalingam, K., Yadalam, P. K., Ramani, P., Krishna, M., Hafedh, S., Badnjević, A., Cervino, G., & Minervini, G., 2024, Light gradient boosting-based prediction of quality of life among oral cancer-treated patients. BMC Oral Health, 24(1), 349. https://doi.org/10.1186/s12903-024-04050-x.[21] Yamamoto, T., Suzuki, T., Kobayashi, A., Wakabayashi, J., Maher, J., Motohashi, H., & Yamamoto, M., 2008, Physiological significance of reactive cysteine residues of Keap1 in determining Nrf2 activity. Molecular and Cellular Biology, 28,2758–2770. doi: 10.1128/MCB.01704-07.[22] Stancill, J.S., & Corbett, J.A., 2021, The Role of Thioredoxin/Peroxiredoxin in the β-Cell Defense Against Oxidative Damage.Front Endocrinol (Lausanne),12,718235. doi: 10.3389/fendo.2021.718235.[23] Elmarakby, A.A., Faulkner, J., Baban, B., & Sullivan, J.C., 2012, Induction of Hemeoxygenase-1 Reduces Renal Oxidative Stress and Inflammation in Diabetic Spontaneously Hypertensive Rats. International Journal of Hypertension, 957235. doi:10.1155/2012/957235.[24] Elmarakby, A.A., Faulkner, J., Baban, B., Saleh, M.A., & Sullivan, J.C., 2012, Induction of hemeoxygenase-1 reduces glomerular injury and apoptosis in diabetic spontaneously hypertensive rats. American Journal of Physiology-Renal Physiology, 302(7), F791–F800. doi: 10.1152/ajprenal.00472.2011: 10.1152/ajprenal.00472.2011.Viewed PDF 95 6 -
Studies on the Antioxidant Role of Β-Sitosterol on Neuronal Cell Line (IMR32) In Vitro: Role of NRF2-Keap PathwayAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art006
Studies on the Antioxidant Role of Β-Sitosterol on Neuronal Cell Line (IMR32) In Vitro: Role of NRF2-Keap Pathway
Abstract:
β-Sitosterol is a phytosterol which is universally present in most of the plant seeds and nuts. Its anti-inflammatory property has been studied in vitro in cell line studies and some in vivo studies have also been conducted but few studies have been done to establish its antioxidant property. In this study, we have attempted to explore the antioxidant property through the NRF2-KEAP pathway in the IMR 32 neuronal cell line. Initially, cell viability was checked for varying concentrations of sitosterol using an MTT assay. There was a decline in viability for increased concentration and time factors. Reactive oxygen species (ROS) assay revealed the tendency of sitosterol to inhibit lipid peroxides and hydrogen peroxides. An antioxidant activity assay depicted activating antioxidant enzymes, superoxide dismutase, and nonenzymatic antioxidants, such as reduced glutathione. In the gene expression analysis using Real Time-PCR NRF2 and KEAP expression was found to be observed maximum for concentration ranges of 10 µM concentration of β-Sitosterol. Current studies have substantiated the antioxidant property of β-Sitosterol through the NRF2- KEAP signalling pathway.
Studies on the Antioxidant Role of Β-Sitosterol on Neuronal Cell Line (IMR32) In Vitro: Role of NRF2-Keap Pathway
References:
[1]. Zhang, P., Liu, N., Xue, M., Zhang, M., Liu, W., Xu, C., Fan, Y., Meng, Y., Zhang, Q., & Zhou, Y., 2023, Anti-Inflammatory and Antioxidant Properties of β-Sitosterol in Copper Sulfate-Induced Inflammation in Zebrafish (Danio rerio). Antioxidants (Basel, Switzerland), 12(2), 391. https://doi.org/10.3390/antiox12020391[2]. Sun, Y., Gao, L., Hou, W., & Wu, J., 2020, β-Sitosterol Alleviates Inflammatory Response via Inhibiting the Activation of ERK/p38 and NF-κB Pathways in LPS-Exposed BV2 Cells. BioMed Research International, 2020, 7532306. https://doi.org/10.1155/2020/7532306[3]. Kaplanski, G., 2018, Interleukin-18: Biological Properties and Role in Disease Pathogenesis. Immunological Reviews, 281(1), 138–153. https://doi.org/10.1111/imr.12616[4]. Prathipa, S., Shanmuga Sundaram, K. S., Geetha Rani., Krithika Chandrasekar., Ramajayam Govindan., Mahesh Kumar, P., Jaideep Mahendra., Ponnulakshmi,R., 2023, Phytosterols and its Neuroprotective Effect – An Updated Review, European Chemical Bulletin, J. Doi:10.48047/ecb/2023.12.si4.701[5]. Zheng, Y., Zhao, J., Chang, S., Zhuang, Z., Waimei, S., Li, X., Chen, Z., Jing, B., Zhang, D., & Zhao, G., 2023, β-Sitosterol Alleviates Neuropathic Pain by Affect Microglia Polarization through Inhibiting TLR4/NF-κB Signaling Pathway. Journal of Neuroimmune Pharmacology: The Official Journal of the Society on NeuroImmune Pharmacology, 18(4), 690–703. https://doi.org/10.1007/s11481-023-10091-w[6]. Baskar, A. A., Al Numair, K. S., Gabriel Paulraj, M., Alsaif, M. A., Muamar, M. A., & Ignacimuthu, S., 2012, β-sitosterol Prevents Lipid Peroxidation And Improves Antioxidant Status and Histoarchitecture in rats with 1, 2-Dimethylhydrazine-Induced Colon Cancer. Journal of Medicinal Food, 15(4), 335–343. https://doi.org/10.1089/jmf.2011.1780[7]. Gupta, R., Sharma, A. K., Dobhal, M. P., Sharma, M. C., & Gupta, R. S., 2011, Antidiabetic and Antioxidant Potential of β-Sitosterol in Streptozotocin-Induced Experimental Hyperglycemia. Journal of Diabetes, 3(1), 29–37. https://doi.org/10.1111/j.1753-0407.2010.00107.x[8]. Kaspar, J. W., Niture, S. K., & Jaiswal, A. K., 2009, Nrf2:INrf2 (Keap1) Signaling In Oxidative Stress. Free Radical Biology & Medicine, 47(9), 1304–1309. https://doi.org/10.1016/j.freeradbiomed.2009.07.035[9]. Ye, J. Y., Li, L., Hao, Q. M., Qin, Y., & Ma, C. S., 2020, β-Sitosterol Treatment Attenuates Cognitive Deficits And Prevents Amyloid Plaque Deposition In Amyloid Protein Precursor/Presenilin 1 Mice. The Korean Journal Of Physiology & Pharmacology : official Journal of the Korean Physiological Society and The Korean Society of Pharmacology, 24(1), 39–46. https://doi.org/10.4196/kjpp.2020.24.1.39[10]. Wang, J., Wu, F., & Shi, C., 2013, Substitution of Membrane Cholesterol with β-Sitosterol Promotes Nonamyloidogenic Cleavage of Endogenous Amyloid Precursor Protein. Neuroscience, 247, 227–233. https://doi.org/10.1016/j.neuroscience.2013.05.022[11]. Prince, M., Bryce, R., Albanese, E., Wimo, A., Ribeiro, W., & Ferri, C. P., 2013, The global prevalence of Dementia: A Systematic Review and Metaanalysis. Alzheimer's & Dementia: The Journal of the Alzheimer's Association, 9(1), 63–75.e2. https://doi.org/10.1016/j.jalz.2012.11.007[12]. Tiwari, A., & Jain, R. K., 2023, Comparative Evaluation of White Spot Lesion incidence between NovaMin, Probiotic, And Fluoride containing Dentifrices during Orthodontic treatment Using Laser Fluorescence - A Prospective Randomized Controlled Clinical Trial. Clinical and Investigative Orthodontics, 82(2), 75–82. https://doi.org/10.1080/27705781.2023.2190950[13]. Pratiwi, R., Nantasenamat, C., Ruankham, W., Suwanjang, W., Prachayasittikul, V., Prachayasittikul, S., & Phopin, K., 2021, Mechanisms and Neuroprotective Activities of Stigmasterol Against Oxidative Stress-Induced Neuronal Cell Death via Sirtuin Family. Frontiers in Nutrition, 8, 648995. https://doi.org/10.3389/fnut.2021.648995[14]. Tomolonis, J. A., Xu, X., Dholakia, K. H., Zhang, C., Guo, L., Courtney, A. N., Wang, S., Balzeau, J., Barragán, G. A., Tian, G., Di Pierro, E. J., & Metelitsa, L. S., 2023, Interaction Between Tumor Cell TNFR2 and Monocyte Membrane-Bound TNF-α Triggers Tumorigenic Inflammation in Neuroblastoma. Journal for Immunotherapy of Cancer, 11(3), e005478. https://doi.org/10.1136/jitc-2022-005478[15]. Sreenivasagan, S., Subramanian, A. K., Mohanraj, K. G., & Kumar, R. S., 2023, Assessment of Toxicity of Green Synthesized Silver Nanoparticle-coated Titanium Mini-implants with Uncoated Mini-implants: Comparison in an Animal Model Study. The Journal of Contemporary Dental Practice, 24(12), 944–950. https://doi.org/10.5005/jp-journals-10024-3577.[16]. Shi, C., Wu, F., Zhu, X. C., & Xu, J., 2013, Incorporation of Beta-Sitosterol Into The Membrane Increases Resistance To Oxidative Stress And Lipid Peroxidation via Estrogen Receptor-Mediated PI3K/GSK3beta Signaling. Biochimica et Biophysica Acta, 1830(3), 2538–2544. https://doi.org/10.1016/j.bbagen.2012.12.012[17]. Neralla, M., M, H., Preethi, A., Selvakumar, S. C., & Sekar, D., 2024, Expression levels of microRNA-7110 in oral squamous cell carcinoma. Minerva dental and oral science, 73(3), 155–160. https://doi.org/10.23736/S2724-6329.23.04801-5[18]. Ayaz, M., Junaid, M., Ullah, F., Subhan, F., Sadiq, A., Ali, G., Ovais, M., Shahid, M., Ahmad, A., Wadood, A., El-Shazly, M., Ahmad, N., & Ahmad, S., 2017, Anti-Alzheimer's Studies on β-Sitosterol Isolated from Polygonum hydropiper L. Frontiers in Pharmacology, 8, 697. https://doi.org/10.3389/fphar.2017.00697[19]. Babu, S., & Jayaraman, S., 2020, An update on β-sitosterol: A Potential Herbal Nutraceutical for Diabetic Management. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, 131, 110702. https://doi.org/10.1016/j.biopha.2020.110702[20]. Jayaraman, S., Devarajan, N., Rajagopal, P., Babu, S., Ganesan, S. K., Veeraraghavan, V. P., Palanisamy, C. P., Cui, B., Periyasamy, V., & Chandrasekar, K., 2021, β-Sitosterol Circumvents Obesity Induced Inflammation and Insulin Resistance by down-Regulating IKKβ/NF-κB and JNK Signaling Pathway in Adipocytes of Type 2 Diabetic Rats. Molecules (Basel, Switzerland), 26(7), 2101. https://doi.org/10.3390/molecules26072101Viewed PDF 81 7 -
Molecular Basis behind the Neuroprotective Potential of Beta Sitosterol in Lipopolysaccharide-Induced Wistar Albino RatsAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art007
Molecular Basis behind the Neuroprotective Potential of Beta Sitosterol in Lipopolysaccharide-Induced Wistar Albino Rats
Abstract:
Neurodegenerative disorders are on the rise globally. β-Sitosterol shows potential therapeutic benefits, but its neuroprotective mechanisms remain largely unexplored. This study aimed to assess the neuroprotective effects of β-Sitosterol on pro-inflammatory (NFκB) and antioxidant (NRF-2/KEAP-1) pathways in an in vivo in LPS-induced neurodegeneration model in albino rats. The rats were divided into four groups: normal control, LPS-induced, LPS-induced treated with β-Sitosterol (20 mg/kg/day for 4 weeks), and normal treated with β-Sitosterol. Neurotransmitters (dopamine and serotonin) and antioxidant enzymes (GSH and CAT) were measured by ELISA, and gene expression of NFκB, NRF-2, KEAP-1, IL-6, and IL-18 was assessed by Real-Time RT-PCR. Histopathology of brain tissues was performed. LPS induction significantly decreased neurotransmitters and antioxidant enzymes and upregulated NFκB while downregulating NRF-2 and KEAP-1 mRNA expression. β-Sitosterol treatment normalized these levels (p<0.05) and reduced hyperchromatic pyknotic changes in neuronal nuclei observed in LPS-induced rats. Normal rats treated with β-Sitosterol showed no significant alterations, indicating its safety. These findings suggest β-Sitosterol can reduce neuroinflammation by modulating antioxidant signaling, providing a potential therapeutic approach for neurodegenerative diseases.
Molecular Basis behind the Neuroprotective Potential of Beta Sitosterol in Lipopolysaccharide-Induced Wistar Albino Rats
References:
[1]. Uttara, B., Singh, A. V., Zamboni, P., & Mahajan, R. T., 2009, Oxidative Stress and Neurodegenerative Diseases: A Review of Upstream and Downstream Antioxidant Therapeutic Options. Current Neuropharmacology, 7(1), 65–74. https://doi.org/10.2174/157015909787602823.
[2]. S, D. P. A., Solete, P., Jeevanandan, G., Syed, A. A., Almahdi, S., Alzhrani, M., Maganur, P. C., & Vishwanathaiah, S., 2023, Effect of Various Irrigant Activation Methods and Its Penetration in the Apical Third of Root Canal-In Vitro Study. European Journal of dentistry, 17(1), 57–61. https://doi.org/10.1055/s-0041-1742122.
[3]. Prathipa S., Shanmuga, S., Geetha Rani, K.S., Krithika, Chandrasekar., Ramajayam Govindan., Mahesh Kumar, P., Jaideep Mahendra & Ponnulakshmi, R., 2023, Phytosterols and its Neuroprotective Effect – An Updated Review. European Chemical Bulletin.j. DOI:10.48047/ecb/2023.12.si4.701.
[4]. Kaspar, J. W., Niture, S. K., & Jaiswal, A. K., 2009, Nrf2:INrf2 (Keap1) Signaling in Oxidative Stress. Free Radical Biology & Medicine, 47(9), 1304–1309. https://doi.org/10.1016/j.freeradbiomed.2009.07.035.
[5]. Zhao, J., Bi, W., Xiao, S., Lan, X., Cheng, X., Zhang, J., Lu, D., Wei, W., Wang, Y., Li, H., Fu, Y., & Zhu, L., 2019, Neuroinflammation Induced by Lipopolysaccharide Causes Cognitive Impairment in Mice. Scientific Reports, 9(1), 5790. https://doi.org/10.1038/s41598-019-42286-8.
[6]. Gao, W., Guo, L., Yang, Y., Wang, Y., Xia, S., Gong, H., Zhang, B. K., & Yan, M. , 2022, Dissecting the Crosstalk Between Nrf2 and NF-κB Response Pathways in Drug-Induced Toxicity. Frontiers in Cell and Developmental Biology, 9, 809952. https://doi.org/10.3389/fcell.2021.809952.
[7]. Juárez Olguín, H., Calderón Guzmán, D., Hernández García, E., & Barragán Mejía, G. , 2016, The Role of Dopamine and its Dysfunction as a Consequence of Oxidative Stress. Oxidative Medicine and Cellular Longevity, 2016, 9730467. https://doi.org/10.1155/2016/9730467
[8]. Yin, Y., Liu, X., Liu, J., Cai, E., Zhao, Y., Li, H., Zhang, L., Li, P., & Gao, Y., 2018, The Effect of Beta-Sitosterol and its Derivatives on Depression by the Modification of 5-HT, DA and GABA-Ergic Systems in Mice. RSC Advances, 8(2), 671–680. https://doi.org/10.1039/c7ra11364a
[9]. Sedlak, J., & Lindsay, R.H., 1968, Analytical Biochemistry,25, 192-205.
[10]. Takahara, S., Hamilton, H.B., Neel, J.V., Kobara,T.Y., Ogura, Y., & Nishimura, E.T., 1960, Journal of Clinical Investigation, 39, 610-619.
[11]. Vishwanathaiah, S., Maganur, P. C., Manoharan, V., Jeevanandan, G., Hakami, Z., Jafer, M. A., Khanagar, S., & Patil, S., 2022, Does Social Media have any Influence during the COVID-19 Pandemic? An Update. The Journal of Contemporary Dental Practice, 23(3), 327–330.
[12]. Lee, D. Y., Song, M. Y., & Kim, E. H., 2021, Role of Oxidative Stress and Nrf2/KEAP1 Signaling in Colorectal Cancer: Mechanisms and Therapeutic Perspectives with Phytochemicals. Antioxidants (Basel, Switzerland), 10(5), 743. https://doi.org/10.3390/antiox10050743
[13]. Babu, S., Krishnan, M., Rajagopal, P., Periyasamy, V., Veeraraghavan, V., Govindan, R., & Jayaraman, S., 2020, Beta-sitosterol Attenuates Insulin Resistance in Adipose Tissue via IRS-1/Akt Mediated Insulin Signaling in High Fat Diet and Sucrose Induced Type-2 Diabetic Rats. European Journal of Pharmacology, 873, 173004. https://doi.org/10.1016/j.ejphar.2020.173004
[14]. Saha, S., Buttari, B., Panieri, E., Profumo, E., & Saso, L., 2020, An Overview of Nrf2 Signaling Pathway and its Role in Inflammation. Molecules (Basel, Switzerland), 25(22), 5474. https://doi.org/10.3390/molecules25225474.
[15]. Sun, Y., Gao, L., Hou, W., & Wu, J., 2020, β-Sitosterol Alleviates Inflammatory Response via Inhibiting the Activation of ERK/p38 and NF-κB Pathways in LPS-Exposed BV2 Cells. BioMed Research International, 2020, 7532306. https://doi.org/10.1155/2020/7532306
[16]. Francois, A., Terro, F, Janet, T., Rioux Bilan, A., Paccalin, M., & Page, G.,2013, Involvement of Interleukin-1Beta in the Autophagic Process of Microglia: Relevance to Alzheimer’s Disease, Journal of Neuroinflammation,10,151.
[17]. Mathew, M. G., Jeevanandan, G., Vishwanathaiah, S., Hamzi, K. A., Depsh, M. A. N., & Maganur, P. C., 2022, Parental and Child Outlook on the Impact of ECC on Oral Health-related Quality of Life: A Prospective Interventional Study. The Journal of Contemporary Dental Practice, 23(9), 877–882. https://doi.org/10.5005/jp-journals-10024-3397.
[18]. Teleanu, R. I., Niculescu, A. G., Roza, E., Vladâcenco, O., Grumezescu, A. M., & Teleanu, D. M., 2022, Neurotransmitters-Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. International Journal of Molecular Sciences, 23(11), 5954. https://doi.org/10.3390/ijms23115954
[19]. Tagliamonte, A., Biggio, G., Vargiu, L., & Gessa, G. L., 1973, Free Tryptophan in Serum Controls Brain Tryptophan Level and Serotonin Synthesis. Life sciences. Pt. 2: Biochemistry, General and Molecular Biology, 12(6), 277–287. https://doi.org/10.1016/0024-3205(73)90361-5.
[20]. Babu, S., & Jayaraman, S., 2020, An Update on β-sitosterol: A Potential Herbal Nutraceutical for Diabetic Management. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, 131, 110702. https://doi.org/10.1016/j.biopha.2020.110702.
[21]. Jayaraman, S., Devarajan, N., Rajagopal, P., Babu, S., Ganesan, S. K., Veeraraghavan, V. P., Palanisamy, C. P., Cui, B., Periyasamy, V., & Chandrasekar, K. (2021). β-Sitosterol Circumvents Obesity Induced Inflammation and Insulin Resistance by Down-Regulating IKKβ/NF-κB and JNK Signaling Pathway in Adipocytes of Type 2 Diabetic Rats. Molecules (Basel, Switzerland), 26(7), 2101. https://doi.org/10.3390/molecules26072101
Viewed PDF 89 7 -
Possible Interventional Anticancer Therapy by Phytomedicines - A ReviewAuthor: Sridevi GopathyDOI: 10.21522/TIJPH.2013.SE.24.03.Art008
Possible Interventional Anticancer Therapy by Phytomedicines - A Review
Abstract:
Cancer is the rapid proliferation that causes abnormal cells which metastasize to distant tissues. This aberrant signalling mechanism disrupts the regulation of cell proliferation and persistence, ultimately becoming the primary cause of mortality worldwide. The need for novel medications for the treatment and prevention of this deadly disease is constantly rising. Herbal therapies have significance for both preventing and treating a variety of malignancies. Anticancer medications have been discovered and developed from many herbal medicines by the presence of their bioactive phytochemicals such as phenolics, alkaloids, flavonoids, carotenoids, and other secondary metabolites. These herbal products are said to have less toxic side effects when compared to modern treatment strategies. Therapeutic medicinal herbs suppress the progression of cancerous cells by influencing the action of particular enzymes and hormones. The bioactive phytochemicals obstruct cancerous cell multiplication, promote apoptosis of malignant cells, enforce the necrosis of tumors, and inhibit their translocation. They also exert their action by enhancing the number of leukocytes and platelets, promoting the reverse transformation from tumor cells back to usual cells, and they similarly prevent carcinogenesis of regular cells. This review paper enlightens the significance of herbal medicines as anticancer agents and explains, in brief, the mechanism of action and the effects of the herbal bioactive compound. This review helps to explore the potential therapeutic plants as a basis for the discovery of chemotherapy medications.
Possible Interventional Anticancer Therapy by Phytomedicines - A Review
References:
[1] Ferlay, J., Ervik, M., Lam, F., Colombet, M., Mery, L., Piñeros, M., 2021, Global Cancer Observatory: Cancer Today. Lyon: International Agency for Research on Cancer.[2] Krishnan Sathishkuma, Meesha Chaturvedi, Priyanka Das, Stephen, S., & Prashant Mathur., 2022, Cancer incidence estimates for 2022 & projection for 2025: Result from National Cancer Registry Programme, India. The Indian Journal of Medical Research, 156(4&5):598-607.[3] Roy, Arpita & Ahuja, Shruti & Bharadvaja, Navneeta., 2017, A Review on Medicinal Plants against Cancer. Journal of Plant Sciences and Agricultural Research, Volume-2 No.1:008.[4] Montbriand, MJ., 2004, Herbs or natural products that decrease cancer growth part one of a four-part series. Oncol Nurs Forum, 31(4): E75-90.[5] DeVita, VT., Lawrence, TS., Hellmans, Rosenberg, SA., 2004, Cancer: Principles & Practice of Oncology (8th Edn).[6] Madhusudan, S., Middleton, MR., 2005, The emerging role of DNA repair proteins as predictive, prognostic and therapeutic targets in cancer. Cancer Treat Rev, 31(8):603-17. doi: 10.1016/j.ctrv.2005.09.006.[7] Khanam, Sofia, & Prakash, Aman., 2021, An overview of medicinal plants as anticancer agents. IP International Journal of Comprehensive and Advanced Pharmacology, 6. 53-62.[8] Bijauliya, Rohit, & Alok, Shashi, & Singh, Man & Mishra, Shanti Bhushan., 2017, A comprehensive review on cancer and anticancer herbal drugs. International Journal of Pharmaceutical Sciences and Research, 8. 2740-2761.[9] Wenying Ren, Zhenhua Qiao, Hongwei Wang, Lei Zhu, Li Zhang., 2003, “Flavonoids: Promising anticancer agents,” Medical Research Reviews, Volume 23, Issue 4 Pg. 519-534[10] Tait, Stephen & Green, Douglas., 2012, Mitochondria and cell signalling. Journal of cell science. 125. 807-15.[11] Yadav, N., Chandra, D., 2014, Mitochondrial and post-mitochondrial survival signalling in cancer. Mitochondrion, 16:18-25.[12] Hanahan, D., Weinberg, R.A., 2011, Hallmarks of cancer: the next generation, Cell, 144,646–674.[13] Elkord, Eyad & Alcantar-Orozco, Erik & Dovedi, Simon & Tran, Dat & Hawkins, Robert & Gilham, David., 2010, T regulatory cells in cancer: Recent advances and therapeutic potential. Expert opinion on biological therapy, 10:1573-86. 10.1517/14712598.2010.529126.[14] Sena, Laura & Chandel, Navdeep, Sena, LA., Chandel, NS., 2012, Physiological roles of mitochondrial reactive oxygen species. Mol Cell, 48:158-167, 10.1016/j.molcel.2012.09.025.[15] Wallace, Douglas., 2012, Mitochondria and Cancer. Nature Reviews Cancer, 12. 685-698. 10.1038/nrc3365.[16] Ray, P., Paul, & Huang, Bo-Wen, & Tsuji, Yoshiaki., 2012, Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cellular signalling, 24. 981-90. 10.1016/j.cellsig.2012.01.008.[17] Van Waes, C., 2007, Nuclear factor-kappaB in development, prevention, and therapy of cancer. Clin Cancer Res, 13(4):1076-82.[18] Mondal, A., Gandhi, A., Fimognari, C., Atanasov, AG., Bishayee, A., 2019, Alkaloids for cancer prevention and therapy: Current progress and future perspectives. Eur J Pharmacol, 5; 858:172472.[19] Huang, Wu-Yang, & Cai, Yi-Zhong, & Zhang, Yanbo., 2010, Natural Phenolic Compounds from Medicinal Herbs and Dietary Plants: Potential Use for Cancer Prevention. Nutrition and cancer, 62. 1-20. 10.1080/01635580903191585.[20] Sreevarun, M., Ajay, R., Suganya, G., Rakshagan, V., Bhanuchander, V., & Suma, K., 2023, Formulation, Configuration, and Physical Properties of Dental Composite Resin Containing a Novel 2π + 2π Photodimerized Crosslinker - Cinnamyl Methacrylate: An In Vitro Research. The Journal of Contemporary Dental Practice, 24(6), 364–371. https://doi.org/10.5005/jp-journals-10024-3480[21] Thomson, Martha, & Ali, Muslim., 2003, Garlic (Allium sativum): A Review of its Potential Use as an Anti-Cancer Agent. Current cancer drug targets, 3. 67-81. 10.2174/1568009033333736.[22] Claudia, Cerella, Christiane, Scherer, Silvia Cristofanon, Estelle Henry, Mario Dicato & Marc Diederich., 2009, Cell cycle arrest in early mitosis and induction of caspase-dependent apoptosis in U937 cells by diallyltetrasulfide (Al2S4). Apoptosis, 14, 641–654.[23] Noble, RL., 1990, The discovery of the vinca alkaloids-chemotherapeutic agents against cancer. Biochem Cell Biol, 68(12):1344-51.[24] Bruneton, J., 1993. Pharmacognosy, phytochemistry, medicinal plants (No. Ed. 2, pp. xii+-915).[25] Alam, M. K., Alqhtani, N. R., Alnufaiy, B., Alqahtani, A. S., Elsahn, N. A., Russo, D., Di Blasio, M., Cicciù, M., & Minervini, G. (2024). A systematic review and meta-analysis of the impact of resveratrol on oral cancer: potential therapeutic implications. BMC Oral Health, 24(1), 412. https://doi.org/10.1186/s12903-024-04045-8.[26] Elumalai, P., Gunadharini, DN., Senthilkumar, K., Banudevi, S., Arunkumar, R., Benson, CS., Sharmila, G., Arunakaran, J., 2012, Induction of apoptosis in human breast cancer cells by nimbolide through extrinsic and intrinsic pathway. Toxicol Lett, 215(2):131-42.[27] Yadalam, P. K., Arumuganainar, D., Ronsivalle, V., Di Blasio, M., Badnjevic, A., Marrapodi, M. M., Cervino, G., & Minervini, G., 2024, Prediction of interactomic hub genes in PBMC cells in type 2 diabetes mellitus, dyslipidemia, and periodontitis. BMC Oral Health, 24(1), 385. https://doi.org/10.1186/s12903-024-04041-y..[28] Cragg, GM., Newman, DJ., 2005, Plants as a source of anti-cancer agents. J Ethnopharmacol, 100(1-2):72-9.[29] Bala, A., Mukherjee, P. K., Braga, F. C., & Matsabisa, M. G., 2018, Comparative inhibition of MCF-7 breast cancer cell growth, invasion and angiogenesis by Cannabis sativa L. sourced from sixteen different geographic locations. South African Journal of Botany, 119, 154–162.[30] Malhotra, P., Casari, I., & Falasca, M., 2021, Therapeutic potential of cannabinoids in combination cancer therapy. Advances in Biological Regulation, 79, 100774.[31] Pereira, Manisha, & Haniadka, Raghavendra, & Chacko, Palatty, Princy, & Baliga, Shrinath., 2011, Zingiber officinale Roscoe (ginger) as an adjuvant in cancer treatment: A review. Journal of B.U. ON: official journal of the Balkan Union of Oncology, 16. 414-24.[32] Prasad, S., Tyagi, A.K., 2015, Ginger and its constituents: Role in prevention and treatment of gastrointestinal cancer. Gastroenterology Research and Practice, 142979, 1–11.[33] Debnath, R., Chatterjee, N., Das, S., Mishra, S., Bose, D., Banerjee, S., Das, S., Saha, KD., Ghosh, D., Maiti, D., 2019, Bromelain with peroxidase from pineapple are more potent to target leukemia growth inhibition—a comparison with only bromelain. Toxicol in Vitro, 55:24–32.[34] Kargutkar, S., Brijesh, S., 2018, Anti-inflammatory evaluation and characterization of leaf extract of Ananas comosus. Infammopharmacology, 26:469–477.[35] Zari, A.T., Hakeem, K.R., 2021, Anticancer Properties of Eugenol: A Review. Molecules, 26, 7407.[36] Fangjun, L., Zhijia, Y., 2018, Tumor suppressive roles of eugenol in human lung cancer cells. Thorac. Cancer, 9, 25–29.[37] Zongchao Hong, Yi Lu, Chongwang Ran, Peili Tang, Ju Huang, Yanfang Yang, Xueyun Duan, Hezhen Wu., 2021, The bioactive ingredients in Actinidia chinensis Planch. Inhibit liver cancer by inducing apoptosis. Journal of Ethnopharmacology, Volume 281, 114553, ISSN 0378-8741.[38] Motohashi, N., Shirataki, Y., Kawase, M., Tani, S., Sakagami, H., 2002, Cancer prevention and therapy with kiwifruit in Chinese folklore medicine: a study of kiwifruit extracts. J Ethnopharmacol, 81, 357–364,[39] Pecere, T., Gazzola, MV., Mucignat, C., Parolin, C., Vecchia, FD., Cavaggioni, A., Basso, G., Diaspro, A., Salvato, B., Carli, M., Palù, G., 2000, Aloe-emodin is a new type of anticancer agent with selective activity against neuroectodermal tumors. Cancer Res, 60(11):2800-4. PMID: 10850417.[40] Manivannan, Umadevi, & Kumar, & Bhowmik, Debjit & Duraivel, S., 2013, Traditionally Used Anticancer Herbs in India. J. Med. Plants Res, 1. 56-74.Viewed PDF 92 10 -
Anti-diabetic Potential Mechanisms of Phytomedicines – A ReviewAuthor: Sridevi GopathyDOI: 10.21522/TIJPH.2013.SE.24.03.Art009
Anti-diabetic Potential Mechanisms of Phytomedicines – A Review
Abstract:
Diabetic mellitus is an endocrine disorder characterized by hyperglycemia, polyphagia, polyuria, and polydipsia. In this condition, the cells and tissues are unable to utilize glucose for energy due to inadequate insulin secretion. The complications of the disease include diabetic retinopathy, diabetic neuropathy, and diabetic nephropathy that affect the eyes, nerves, kidneys and stroke, renal failure, and heart attacks are other serious consequences of diabetes. The conventional modern medicines for treatment are oral hypoglycemic drugs, sulfonylureas and glinides, metformin and thiazolidinedione, dipeptidyl peptidase-4 (DPP4) inhibitors, and injections such as GLP-1 agonists. Due to the presence of a lot of side effects, the modern world is now turning to bioactive chemical components synthesized from plants. Today, drugs derived from herbs or plants are widely used because of the exploitation of specific compounds and their therapeutic actions. Various phytochemicals have notable and significant mechanisms for reducing the blood glucose level. These natural agents can have a protective and therapeutic effect on diabetes mellitus through cellular mechanisms such as the regeneration of pancreatic β cells, antioxidative stress, and intracellular signalling transduction pathways. The present study aims to review the mechanisms of various phytochemicals that play a role in antidiabetic activity. The possible mechanisms by which the antidiabetic herbs act are α-glucosidase inhibitors, PPAR activators, free radical scavengers, HMG Co suppressors, regenerators of beta cells, and cause an increase in insulin secretion and glycogen synthesis in glycemic control.
Anti-diabetic Potential Mechanisms of Phytomedicines – A Review
References:
[1] American Diabetes Association, 2012, Diagnosis and Classification of Diabetes Mellitus. Diabetes Care, 36(Supplement_1), S67–S74. https://doi.org/10.2337/dc13-S067
[2] International Diabetes Federation, 2021, IDF Diabetes Atlas, 10th edition. Brussels, Belgium: https://www.diabetesatlas.org/.
[3] Kakkar, R., 2016, Rising burden of Diabetes-Public Health Challenges & way out. Nepal Journal of Epidemiology, 6(2), 557–559. https://doi.org/10.3126/nje.v6i2.15160
[4] Ali, M. K., Narayan, K. M., & Tandon, N. 2010, Diabetes & coronary heart disease: current perspectives. The Indian journal of medical research, 132(5), 584–597.
[5] Singh, N., Armstrong, D. G., & Lipsky, B. A., 2005, Preventing foot ulcers in patients with diabetes. JAMA, 293(2), 217. https://doi.org/10.1001/jama.293.2.217
[6] Levetan, C., 2007, Oral antidiabetic agents in type 2 diabetes. Current Medical Research and Opinion, 23(4), 945–952. https://doi.org/10.1185/030079907x178766
[7] Inzucchi, S. E., 2002, Oral antihyperglycemic therapy for type 2 diabetes. JAMA, 287(3), 360. https://doi.org/10.1001/jama.287.3.360
[8] Mendoza, N., & Silva, E. M. E., 2018, Introduction to phytochemicals: Secondary metabolites from plants with active principles for pharmacological importance. In Phytochemicals - Source of Antioxidants and Role in Disease Prevention. InTech.http://dx.doi.org/10.5772/intechopen.78226
[9] Vinayagam, R., Xiao, J., & Xu, B., 2017, An insight into anti-diabetic properties of dietary phytochemicals. Phytochemistry Reviews, 16(3), 535–553. https://doi.org/10.1007/s11101-017-9496-2
[10] Singh, V. K., Umar, S., Ansari, S. A., & Iqbal, M., 2008, Gymnema sylvestre for Diabetics. Journal of Herbs, Spices & Medicinal Plants, 14(1–2), 88–106. https://doi.org/10.1080/10496470802341508
[11] Gulab S. Thakur., 2012, Gymnema sylvestre: An Alternative Therapeutic Agent for Management of Diabetes. Journal of Applied Pharmaceutical Science, 2(12), 001-006. https://doi.org/10.7324/japs.2012.21201
[12] Persaud, S., Al-Majed, H., Raman, A., & Jones, P., 1999, Gymnema sylvestre stimulates insulin release in vitro by increased membrane permeability. Journal of Endocrinology, 163(2), 207-212. https://doi.org/10.1677/joe.0.1630207
[13] AlAttas, S. A., Zahran, F. M., & Turkistany, S. A., 2016, Nigella sativa and its active constituent thymoquinone in oral health. Saudi Medical Journal, 37(3), 235–244. https://doi.org/10.15537/smj.2016.3.13006
[14] Varghese, R. M., Kumar, S. A., & Selvaraj, Y., 2023, Assessment of Soft Tissue, Airway Dimension and Hyoid Bone Position in Class II Patients Treated by PowerScope Class 2 Corrector. The Journal of Contemporary Dental Practice, 24(5), 308–313. https://doi.org/10.5005/jp-journals-10024-3485
[15] Atangwho, I. J., Ebong, P. E., Eyong, E. U., Williams, I. O., Eteng, M. U., & Egbung, G. E., 2009, Comparative chemical composition of leaves of some antidiabetic medicinal plants: Azadirachta indica, Vernonia amygdalina and Gongronema latifolium. African Journal of Biotechnology, 8(18), 4685-4689.
[16] Djeujo, F. M., Stablum, V., Pangrazzi, E., Ragazzi, E., & Froldi, G., 2023, Luteolin and Vernodalol as Bioactive Compounds of Leaf and Root Vernonia amygdalina Extracts: Effects on α-Glucosidase, Glycation, ROS, Cell Viability, and In Silico ADMET Parameters. Pharmaceutics, 15(5), 1541. https://doi.org/10.3390/pharmaceutics15051541
[17] Baquer, N. Z., Kumar, P., Taha, A., Kale, R., Cowsik, S., & McLean, P., 2011, Metabolic and molecular action of Trigonella foenum-graecum (fenugreek) and trace metals in experimental diabetic tissues. Journal of Biosciences, 36(2), 383–396. https://doi.org/10.1007/s12038-011-9042-0
[18] Sowmithra Devi, S., Sundari, S.,2023, Occlusal Contact Changes With Traumatic Occlusion After Orthodontic Treatment: A Prospective Study. Journal of Advanced Oral Research. 14(2):134-142. doi:10.1177/23202068231190202
[19] Kahramanoğlu, İ., Chen, C., Chen, J., & Wan, C., 2019, Chemical Constituents, Antimicrobial Activity, and Food Preservative Characteristics of Aloe vera Gel. Agronomy, 9(12), 831. https://doi.org/10.3390/agronomy9120831
[20] Tanaka, M., Misawa, E., Ito, Y., Habara, N., Nomaguchi, K., Yamada, M., … Higuchi, R., 2006, Identification of five phytosterols from aloe vera gel as anti-diabetic compounds. Biological and Pharmaceutical Bulletin, 29(7), 1418–1422. https://doi.org/10.1248/bpb.29.1418
[21] Kumar, D., Mitra, A., & M, M., 2011, Azadirachtolide: An anti-diabetic and hypolipidemic effects from Azadirachta indica leaves. Pharmacognosy Communications, 1(1), 78–84. https://doi.org/10.5530/pc.2011.1.5
[22] Ponnusamy, S., Haldar, S., Mulani, F., Zinjarde, S., Thulasiram, H., & RaviKumar, A., 2015. Gedunin and Azadiradione: Human Pancreatic Alpha-Amylase Inhibiting Limonoids from Neem (Azadirachta indica) as Anti-Diabetic Agents. PLOS ONE, 10(10), e0140113. https://doi.org/10.1371/journal.pone.0140113
[23] Aabideen, Z. U., Mumtaz, M. W., Akhtar, M. T., Raza, M. A., Mukhtar, H., Irfan, A., Saari, N., 2021, Cassia fistula Leaves; UHPLC-QTOF-MS/MS Based Metabolite Profiling and Molecular Docking Insights to Explore Bioactives Role towards Inhibition of Pancreatic Lipase. Plants, 10(7), 1334. https://doi.org/10.3390/plants10071334
[24] Adisakwattana, S., 2017, Cinnamic acid and its derivatives: Mechanisms for prevention and management of diabetes and its complications. Nutrients, 9(2), 163. https://doi.org/10.3390/nu9020163
[25] Roy, J. R., Janaki, C. S., Jayaraman, S., Periyasamy, V., Balaji, T., Vijayamalathi, M., & Veeraraghavan, V. P., 2022, Carica papaya Reduces Muscle Insulin Resistance via IR/GLUT4 Mediated Signaling Mechanisms in High Fat Diet and Streptozotocin-Induced Type-2 Diabetic Rats. Antioxidants, 11(10), 2081. https://doi.org/10.3390/antiox11102081
[26] Ansari, P., Flatt, P. R., Harriott, P., Hannan, J. M. A., & Abdel-Wahab, Y. H. A., 2021, Identification of Multiple Pancreatic and Extra-Pancreatic Pathways Underlying the Glucose-Lowering Actions of Acacia arabica Bark in Type-2 Diabetes and Isolation of Active Phytoconstituents. Plants, 10(6), 1190. https://doi.org/10.3390/plants10061190
[27] Srinivasan, S., Sathish, G., Jayanthi, M., Muthukumaran, J., Muruganathan, U., & Ramachandran, V., 2013, Ameliorating effect of eugenol on hyperglycemia by attenuating the key enzymes of glucose metabolism in streptozotocin-induced diabetic rats. Molecular and Cellular Biochemistry, 385(1–2), 159–168. https://doi.org/10.1007/s11010-013-1824-2
[28] Adebanke, O., Babatunde, A., Franklyn, I., Keleeko, A., Joseph, O., & Olubanke, O., 2023, Free radical scavenging activity, pancreatic lipase and a-amylase inhibitory assessment of ethanolic leaf extract of Phyllanthus amarus. Plant Science Today, 10(2), 20–26. https://doi.org/10.14719/pst.1809
[29] Ahmad, N., Hasan, N., Ahmad, Z., Zishan, M., & Zohrameena, S., 2016, MOMORDICA CHARANTIA: FOR TRADITIONAL USES AND PHARMACOLOGICAL ACTIONS. Journal of Drug Delivery and Therapeutics, 6(2), 40-44. https://doi.org/10.22270/jddt.v6i2.1202
[30] Yan, L., Vaghari-Tabari, M., Malakoti, F., Moein, S., Qujeq, D., Yousefi, B., & Asemi, Z., 2022, Quercetin: An effective polyphenol in alleviating diabetes and diabetic complications. Critical Reviews in Food Science and Nutrition, 63(28), 9163–9186. https://doi.org/10.1080/10408398.2022.2067825
[31] Varghese, R.M., Subramanian, A.K., Maliael, M.T., 2023, PowerScope™ for Class II Malocclusions: A Systematic Review and Meta-analysis. World Journal of Dentistry,14(7):639–647
[32] Bozkurt, O., Kocaadam-Bozkurt, B., & Yildiran, H., 2022, Effects of curcumin, a bioactive component of turmeric, on type 2 diabetes mellitus and its complications: An updated review. Food & Function, 13(23), 11999–12010. https://doi.org/10.1039/d2fo02625b
[33] Majeed, M., Mundkur, L., Paulose, S., & Nagabhushanam, K., 2022, Novel Emblica officinalis extract containing β-glucogallin vs. metformin: A randomized, open-label, comparative efficacy study in newly diagnosed type 2 diabetes mellitus patients with dyslipidemia. Food & Function, 13(18),9523–9531. https://doi.org/10.1039/d2fo01862d
[34] Mathiyazhagan, J., & Kodiveri Muthukaliannan, G., 2020, The role of mTOR and oral intervention of combined Zingiberofficinale Terminalia chebula extract in type 2 diabetes rat models. Journal of Food Biochemistry, 44(7). https://doi.org/10.1111/jfbc.13250
[35] Mahindrakar, K. V., & Rathod, V. K., 2020, Antidiabetic potential evaluation of aqueous extract of wasteSyzygium cuminiseed kernel’s byin vitroα-amylase and α-glucosidase inhibition. Preparative Biochemistry & Biotechnology, 51(6), 589–598. https://doi.org/10.1080/10826068.2020.1839908
[36] Rasool, S., Al Meslmani, B., & Alajlani, M., 2023, Determination of hypoglycemic, hypolipidemic and nephroprotective effects of berberis calliobotrys in alloxan-induced diabetic rats. Molecules, 28(8), 3533. https://doi.org/10.3390/molecules2808353
[37] Etsassala, N. G. E. R., Badmus, J. A., Marnewick, J. L., Egieyeh, S., Iwuoha, Emmanuel. I., Nchu, F., & Hussein, A. A., 2022, Alpha-Glucosidase and Alpha-Amylase Inhibitory Activities, Molecular Docking, and Antioxidant Capacities of Plectranthus ecklonii Constituents. Antioxidants, 11(2), 378. https://doi.org/10.3390/antiox11020378
[38] Maher, S., Choudhary, M. I., Saleem, F., Rasheed, S., Waheed, I., Halim, S. A., … Ahmad, S., 2020, Isolation of Antidiabetic Withanolides from Withania coagulans Dunal and Their In Vitro and In Silico Validation. Biology, 9(8), 197. https://doi.org/10.3390/biology9080197
[39] Hsu, J.-H., Yang, C.-S., & Chen, J.-J., 2022, Antioxidant, Anti-α-Glucosidase, Antityrosinase, and Anti-Inflammatory Activities of Bioactive Components from Morus alba. Antioxidants, 11(11), 2222. https://doi.org/10.3390/antiox11112222
[40] Kumar, R., Sood, P., Rana, Dr. V., & Prajapati, A. K., 2023, Combine therapy of gallic acid and allicin in management of diabetes. Journal for Research in Applied Sciences and Biotechnology, 2(3), 91–99. https://doi.org/10.55544/jrasb.2.3.12
Viewed PDF 102 8 -
Wound Healing Property of Herbal Dressing Film Using Chitosan and Peel Extracts of Citrus limettaAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art010
Wound Healing Property of Herbal Dressing Film Using Chitosan and Peel Extracts of Citrus limetta
Abstract:
Impaired wound healing is one of the serious problems among the diabetic patients. To prevent complications and damage to the skin tissue and promote fibroblastic growth, many biological dressing materials and skin grafting have been employed. The present study fabricated a novel wound healing gel/film (CH-CL) using chitosan (CH) and methanolic peel extract of Citrus limetta (CL) and investigated the potential towards the wound healing process. During this investigation, the peel extract of CL was subjected to GC-MS to reveal the bioactive compounds responsible for various activities of the extract. Moreover, the CL extract was analyzed for its anti-microbial, anti-oxidant and anti-inflammatory properties. The CH-CL film was also subjected to water absorption capacity and folding endurance. The extract was also investigated for its viability using normal keratinocytes cell lines. The physico chemical characterization of the gel was done to reveal the chemical composition using FTIR, XRD and SEM. The GC-MS analysis results clearly indicated that the gel is biocompatible, possessing anti-microbial, anti-oxidant and anti-inflammatory properties due to its bioactive compounds. Furthermore, the CH-CL film acts as a good water absorbing material with optimum folding endurance which are the key physical properties of a normal wound healing material. Thus, the study concluded that CH-CL gel has been proven as an efficient, cost-economic wound healing gel and can be applied for various types of wounds and other biomedical applications.
Wound Healing Property of Herbal Dressing Film Using Chitosan and Peel Extracts of Citrus limetta
References:
[1] Gonzalez, Costa, TF, Andrade, ZA., Medrado AR., 2016, Wound healing - A literature review. Anals of Brasilian Dermatology, 91(5):614-620. doi:10.1590/abd1806-4841.20164741[2] Järbrink, K., Ni, G., Sönnergren, H., et al,, 2016, Prevalence and incidence of chronic wounds and related complications: A protocol for a systematic review. Systematic Reviews, 5(1):152. doi:10.1186/s13643-016-0329-y.[3] Ige, OO., Umoru, L.E., et al., 2012, Natural products: A minefield of biomaterials. ISRN Material Sciences, 20:1-5[4] Ahuja., Annapurna., Ahuja Vipin., and Singh Kumar., 2015, Current concepts of regenerative biomaterials in implant dentistry. Journal of the International Clinical Dental Research Organization,7(3):34-39[5] Sreevarun, M., Ajay, R., Suganya, G., Rakshagan, V., Bhanuchander, V., & Suma, K., 2023, Formulation, Configuration, and Physical Properties of Dental Composite Resin Containing a Novel 2π + 2π Photodimerized Crosslinker - Cinnamyl Methacrylate: An In Vitro Research. The Journal of Contemporary Dental Practice, 24(6), 364–371. https://doi.org/10.5005/jp-journals-10024-3480[6] Alam, M. K., Alqhtani, N. R., Alnufaiy, B., Alqahtani, A. S., Elsahn, N. A., Russo, D., Di Blasio, M., Cicciù, M., & Minervini, G., 2024, A systematic review and meta-analysis of the impact of resveratrol on oral cancer: potential therapeutic implications. BMC Oral Health, 24(1), 412. https://doi.org/10.1186/s12903-024-04045-8.[7] Yadalam, P. K., Arumuganainar, D., Ronsivalle, V., Di Blasio, M., Badnjevic, A., Marrapodi, M. M., Cervino, G., & Minervini, G., 2024, Prediction of interactomic hub genes in PBMC cells in type 2 diabetes mellitus, dyslipidemia, and periodontitis. BMC Oral Health, 24(1), 385. https://doi.org/10.1186/s12903-024-04041-y.[8] Feng, P., Luo, Y., Ke C., Qiu, H., Wang, W., Zhu, Y., Hou, R., Xu, L ., Wu, S., 2021, Chitosan-Based Functional Materials for Skin Wound Repair: Mechanisms and Applications. Frontiers in Bioengineering and Biotechnology, 9:650598. doi: 10.3389/fbioe.2021.650598[9] Kamkar, A., Molaee-aghaee E., Khanjari, A., Akhondzadeh-basti, A., Noudoost, B., Shariatifar, N., Alizadeh Sani, M., Soleimani M., 2021, Nanocomposite Active Packaging Based on Chitosan Biopolymer Loaded with Nano-Liposomal Essential Oil: Its Characterizations and Effects on Microbial, and Chemical Properties of Refrigerated Chicken Breast Fillet. International Journal of Food Microbiology , 342:109071. doi: 10.1016/j.ijfoodmicro.2021.109071.[10] Sharma, A., Khanna, S., Kaur, G. et al., 2021, Medicinal plants and their components for wound healing applications. Future Journal of Pharmacy Sciences, 7:53. https://doi.org/10.1186/s43094-021-00202-w[11] Ahmad, Khan, TH., Ansari, MN., Ahmad SF., 2014, Enhanced wound healing by topical administration of d-limonene in alloxan induced diabetic mice through reduction of pro-inflammatory markers and chemokine expression. BMC Genomics, 15(2): 1-7. doi:10.1186/1471-2164-15-S2-P29[12] Prieto, Pila., & Pineda., Manuel & Aguilar, Miguel., 1999, Spectrophotometric Quantitation of Antioxidant Capacity through the Formation of a Phosphomolybdenum Complex: Specific Application to the Determination of Vitamin E. Analytical Biochemistry, 269: 337-41. 10.1006/abio.1999.4019.[13] Sridevi, G., Sembulingam, K., Muhammed Ibrahim., Srividya, S., Prema Sembulingam., 2015, Evaluation of in vitro anti-inflammatory activity of pergularia daemia, World Journal of Pharmacological Research, 4(6) :1100- 1108[14] Seon Jeong Kim., Sang jun Park., Sun Il Kim., 2003, Swelling behavior of interpenetrating polymer network hydrogels composed of polyvinyl alcoholand chitosan. Reactive and Functional Polymers, 55:53-59[15] Silvana Cartaxo da Costa Urtiga ., Vitória Maria Oliveira Alves., Camila de Oliveira Melo ., Marini Nascimento de Lima ., Ernane Souza , Arcelina Pacheco Cunha ., Nágila Maria Pontes Silva Ricardo., 2020, Xylan microparticles for controlled release of mesalamine: Production and physicochemical characterization. Carbohydrate Polymers, 25: 116929[16] Delbianco M, Davis BG, Seeberger PH. Glycans in Nanotechnology. In: Varki A, Cummings RD, Esko JD, et al., editors. 2022, Essentials of Glycobiology. 4th edition. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press. Chapter 58. Available from: https://www.ncbi.nlm.nih.gov/books/NBK579980/ doi: 10.1101/glycobiology.4e.58[17] Silva, F.R.F., Dore, C.M.P.G., Marques, C.T., Nascimento, M.S., Benevides, N.M.B., Rocha, H.A.O., Chavante, S.F., Leite, E.L., 2010, Anticoagulant activity, paw edema and pleurisy induced carrageenan: Action of major types of commercial carrageenans, Carbohydrate Polymers, 79(1): 26-33 https://doi.org/10.1016/j.carbpol.2009.07.010.[18] Fernandes Queiroz, M., Melo, KRT., Sabry, DA., Sassaki, GL., Rocha HAO, 2015, Does the Use of Chitosan Contribute to Oxalate Kidney Stone Formation? Marine Drugs, 13(1),141-158. https://doi.org/10.3390/md13010141[19] Ahmed Abdullah Khan, Tarique Mahmood, Hefazat H. Siddiqui and Juber Akhtar., 2016, Phytochemical and pharmacological properties on Citrus limetta (Mosambi). Journal of Chemical and Pharmaceutical Research, 8(3) :555-563[20] Chopra, H, Bibi, S, Kumar, S, Khan, MS, Kumar, P, Singh, I, 2022, Preparation and Evaluation of Chitosan/PVA Based Hydrogel Films Loaded with Honey for Wound Healing Application. Gels. 11, 8(2):111. doi: 10.3390/gels8020111. PMID: 35200493; PMCID: PMC8871709.[21] Roy, A., Das, S., Chatterjee, I., Roy, S,, Chakraborty, R., 2022, Anti-inflammatory Effects of Different Dietary Antioxidants. In: Ekiert, H.M., Ramawat, K.G., Arora, J. (eds) Plant Antioxidants and Health. Reference Series in Phytochemistry. Springer, Cham. https://doi.org/10.1007/978-3-030-78160-6_20[22] Umapathy, E., Ndebia, E.J., Meeme, A., Adam, B., Menziwa, P., Nkeh-Chungag, BN., Iputo, J. E., 2010, An experimental evaluation of Albuca setosa aqueous extract on membrane stabilization, protein denaturation and white blood cell migration during acute inflammation. Journal of Medicinal Plant Research, 4(9):789-795.[23] Masson-Meyers, DS., Andrade, TAM., Caetano, GF., Guimaraes, FR., Leite, MN., Leite , SN., Frade, MAC., Experimental models and methods for cutaneous wound healing assessment. International Journal of Experimental Pathology. 2020 Feb;101(1-2):21-37. doi: 10.1111/iep.12346.[24] Bertelli, A., Biagi, M., Corsini, M., Baini, G., Cappellucci, G., Miraldi, E., 2021, Polyphenols: From Theory to Practice. Foods 10(11),2595. doi: 10.3390/foods10112595. PMID: 34828876; PMCID: PMC8621732.Viewed PDF 104 8 -
Novel Bonegraft Composite using Hydroxyapatite, Egg-Shell Powder and Chitosan Fortified with Terminalia chebulaAuthor: Srividya SeshadriDOI: 10.21522/TIJPH.2013.SE.24.03.Art011
Novel Bonegraft Composite using Hydroxyapatite, Egg-Shell Powder and Chitosan Fortified with Terminalia chebula
Abstract:
Allogeneic bone grafts are considered to be an integral part of implant dentistry. These grafts have rich osteoconductivity, osteoinductivity and osteogencity and can act as a substrate material to fill the defects on bone surfaces and can augment bone fracture and tooth fracture healing. Based on such principles, this current study fabricated a novel graft composite for bone using egg shell powder (ESP), Hydroxyapatite (HA), Chitosan (CH) and Aqueous extract of fruits of Terminalia chebula (TC). The prepared new bone graft (ESP-HA-CH-Tc) was analyzed using various physico-chemical characterization techniques like FT-IR, XRD, TGA, and SEM to understand the chemical composition, evaluate its surface morphology and stability standard. The aqueous fruit extract of Terminalia chebula (Tc) was analysed for its phytochemical composition by GCMS and invitro pharmacological properties like antioxidant, anti-inflammatory, antimicrobial and the biocompatibility of the graft were assessed by cell culture studies in Breast cancer (MCF-7) cell lines. The incorporation of fruit extract of Terminalia chebula strengthens and augments the ossification property of the graft material and can extend its use in the treatment of fractures and further biomedical applications.
Novel Bonegraft Composite using Hydroxyapatite, Egg-Shell Powder and Chitosan Fortified with Terminalia chebula
References:
[1] Lee., Na Kyung., 2007, Endocrine Regulation of Energy Metabolism by the Skeleton, Cell.;130(3): 456–469.[2] Deakin., Barbara Young., Philip Woodford., 2006, Wheater’s functional histology: A text and colour atlas, 5th ed. Churchill Livingstone/Elsevier, pp.192-195.[3] Rizzo, S.E., Kenan, S., Pathologic Fractures. Treasure Island (FL): StatPearls Publishing; 2022 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK559077/[4] Soni., Vandana & Pandey., Vikas & Asati., Saket & Gour., Vishal & Tekade., Rakesh., Biodegradable Block Copolymers and Their Applications for Drug Delivery,2019:369-400 10.1016/B978-0-12-817909-3.00011-X.[5] Amini, AR., Laurencin, CT., Nukavarapu, SP., 2012, Bone tissue engineering: recent advances and challenges. Critical Reviews in Biomedical Engineering,40(5):363-408. doi:10.1615/critrevbiomedeng.v40.i5.10[6] Islam, MM., Shahruzzaman, M., Biswas, S., Nurus Sakib, M., Rashid, TU., 2020,Chitosan based bioactive materials in tissue engineering applications-A review. Bioactive Materials, 5(1):164-183. Published 2020 Feb 12. doi:10.1016/j.bioactmat.2020.01.012[7] Sumana Roy, Tamalika Chakraborty, Jeenatara Begum, Md Saquib Hasnain, Amit Kumar Nayak, 2022, Chapter 1 - Chitosan: a versatile biopolymer, Editor(s): Md Saquib Hasnain, Sarwar Beg, Amit Kumar Nayak, Chitosan in Biomedical Applications, Academic Press, Pages 1-11,ISBN 9780128210581. https://doi.org/10.1016/B978-0-12-821058-1.00005-8.[8] Dher Riyadh Kadhim, Thekra Ismael Hamad, and Abdalbseet A. Fatalla., 2022, Use of Eggshells as Bone Grafts around Commercially Pure Titanium Implant Screws Coated with Nano Calcium Sulfate. International Journal of Biomaterials, 5:1-8 https://doi.org/10.1155/2022/8722283.[9] Bouyer, E., Gitzhofer, F., Boulos, M I., 2000, Morphological study of hydroxyapatite nanocrystal suspension”, Journal of Material Sciences. Materials and Medicines. 11:523-528.[10] Byron, J., Lambert, Todd, A., Mendelson, Michael, D., 2011, Craven Radiation and Ethylene Oxide Terminal Sterilization Experiences with Drug Eluting Stent Products, AAPS Pharmacy Science and Technology. 12(4):116–1126[11] Prieto, Pilar & Pineda, Manuel & Aguilar, Miguel., 1999, Spectrophotometric Quantitation of Antioxidant Capacity through the Formation of a Phosphomolybdenum Complex: Specific Application to the Determination of Vitamin E. Analytical biochemistry, 269:337-41. doi:10.1006/abio.1999.4019.[12] Sridevi, G ., Sembulingam, K., Muhammed Ibrahim ., Srividya S ., Prema Sembulingam., 2015, Evaluation of in vitro anti-inflammatory activity of pergularia daemia. World Journal of Pharmacological Research, 4(6):1100- 1108[13] Bhavesh., Sridevi, G., Selvaraj, J., and Preetha, S., 2021, Effect of Aegle Marmelos Hydroethanolic Leaf Extract on Expression of Antiapoptotic Markers in Human Melanoma Cells S. Journal of Pharmaceutical Research International, 33(47B): 594-601[14] Kurutas, EB., 2016, The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nutrition Journal. 15(1):71. doi:10.1186/s12937-016-0186-5[15] Umapathy, E., Ndebia, E. J., Meeme, A., Adam, B., Menziwa, P., Nkeh-Chungag, B. N,, Iputo, J. E., 2010, An experimental evaluation of Albuca setosa aqueous extract on membrane stabilization, protein denaturation and white blood cell migration during acute inflammation”, Journal of Medicinal Plants Research, 4(9):789-795.[16] Bag, A., Bhattacharyya, S. K., and Chattopadhyay, R. R., 2013, The development of Terminalia chebula Retz. (Conbretaceae) in clinical research, Asian Pacific Journal of Tropical Biomedicine, 3(3): 244–252[17] Sato, Y., Oketani, H., Singyouchi K., et al., 1997, Extraction and purification of effective antimicrobial constituents of Terminalia chebula Retz. against methicillin-resistant Staphylococcus aureus, Biological and Pharmaceutical Bulletin, 20(4):401–404[18] Cheng, H.Y., Lin, T.C., Yu, K.H., Yang, C.M., and Lin C.C., 2003, Antioxidant and free radical scavenging activities of Terminalia chebula, Biological and Pharmaceutical Bulletin, 26(9):1331–1335[19] Gineste, L., Gineste M, Ranz., Ellefterion A., Guilhem, A., Rouquet, N., Frayssinet, P., 1999, Degradation of hydroxylapatite, fluorapatite, and fluorhydroxyapatite coatings of dental implants in dogs, Journal of Biomedical Material Research, 48:224-234.[20] Tas Cu, K., Neyt, A., 2000, Synthesis of biomimetic Ca-hydroxyapatite powders at 37ºC in synthetic body fluids, Biomaterials, 21:1429-1438[21] Orlovskii, V. P., Komlev, V. S., Barinov, S. M., 2002, Hydroxyapatite and Hydroxyapatite-Based Ceramics, Inorganic Materials, 38(10): 973–984.[22] Suprabha Nayar, Avijit Guha., 2009,Waste utilization for the controlled synthesis of nanosized hydroxyapatite, Materials Science and Engineering, 29: 1326-1329.[23] Mojtaba Nasr-Esfahani and Mehran Mehrabanian, 2011, Production of Bone-like 40% nylon 6,6 -Nano Hydroxyapatite Scaffold via Salt-Leaching/Solvent Casting Technique” 2nd International Conference on Chemistry and Chemical Engineering, 14:40-44.[24] Shihong, Li, Joost R., De Wijn., Jiaping Li., Pierre Layrolle., Klaas De Groot., 2003, Macroporous Biphasic Calcium Phosphate Scaffold with High Permeability/Porosity Ratio, Tissue Engineering, 9(3):535-548.[25] Krithiga, G., Antaryami, Jena., Selvamani, P., Sastry, T. P., 2011, In vitro study on biomineralization of biphasic calcium phosphate biocomposite crosslinked with hydrolysable tannins of Terminalia chebula, Bulletin of Material Sciences, 34(3): 589–594.[26] Puertolas, J.A., Vadillo, J.L, Sanchez-Salcedo, S., Nieto, A., Gomez-Barrena, E., Vallet-Regi, M., 2011, Compression behavior of biphasic calcium phosphate-agarose scaffolds for bone regeneration, Acta Biomaterialia, 7:841-847.[27] Moimas, L., Biasotto, M., Di Lenarda, R., Olivo, A., Schmid, C., 2006, Rabbit pilot study on the resorbability of three-dimensional bioactive glass fibre scaffolds. Acta Biomaterials, 2: 25-27.[28] Zhou Zhi-hua, RUAN Jian-ming, ZOU Jian-peng, ZHOU Zhong-cheng, SHEN Xiong-jun., 2007, Bioactivity of bioresorbable composite based on bioactive glass and poly-L-lactide, Transactions of Nonferrous Metals Society of China.17:394-399[29] Misra, SK., Philip, SE., Chrzanowski, W., Nazhat, SN., Roy, I., Knowles, JC., Salih V., Boccaccini AR., 2009, Incorporation of vitamin E in poly(3hydroxybutyrate)/Bioglass composite films: Effect on surface properties and cell attachment. Journal of the Royal Society Interface. 6(33):401-409.[30] Elke Mitznera., Paco Albertus., Hubert Maria Pelta., Christian Muellera., Angela Strohwiga., Wolf-Dieter Muellerb., 2009, Material Properties and In Vitro Biocompatibility of a Newly Developed Bone Cement”, Materials Research, 12(4):447-454.[31] Sreevarun, M., Ajay, R., Suganya, G., Rakshagan, V., Bhanuchander, V., & Suma, K., 2023, Formulation, Configuration, and Physical Properties of Dental Composite Resin Containing a Novel 2π + 2π Photodimerized Crosslinker - Cinnamyl Methacrylate: An In vitro Research. The Journal of Contemporary Dental Practice, 24(6), 364–371. https://doi.org/10.5005/jp-journals-10024-3480.[32] Alam, M. K., Alqhtani, N. R., Alnufaiy, B., Alqahtani, A. S., Elsahn, N. A., Russo, D., Di Blasio, M., Cicciù, M., & Minervini, G., 2024, A systematic review and meta-analysis of the impact of resveratrol on oral cancer: potential therapeutic implications. BMC Oral Health, 24(1), 412. https://doi.org/10.1186/s12903-024-04045-8.[33] Yadalam, P. K., Arumuganainar, D., Ronsivalle, V., Di Blasio, M., Badnjevic, A., Marrapodi, M. M., Cervino, G., & Minervini, G., 2024, Prediction of interactomic hub genes in PBMC cells in type 2 diabetes mellitus, dyslipidemia, and periodontitis. BMC Oral Health, 24(1), 385. https://doi.org/10.1186/s12903-024-04041-y.[34] Chiou, S.H., W.T. Wu, Y.Y. Huang, T.W. Chung., 2001, Effects of the characteristics of chitosan on controlling drug release of chitosan coated PLLA microspheres, Journal of Microencapsulation. 18:613-625.[35] De Campos, A.M., Sanchez, A., Alonso, M.J., 2001Chitosan nanoparticles: a new vehicle for the improvement of the delivery of drugs to the ocular surface. Application to cyclosporin A, International Journal of Pharmceutics. 224(1):159-168.[36] Fariza, A.R., Zuraida, A., Sopyan, I., 2010, Egg Yolk as Pore Creating Agent for Porous Tri-Calcium Phosphate, International Journal of Engineering and Science, 1:1-6.[37] Murdock, N., Gupta, CR., Vega, N., Kotora, K., Miller, J., et al.,2016, Evaluation of Terminalia chebula Extract for Anti-Arthritic Efficacy and Safety in Osteoarthritic Dogs. Journal of Veterinary Science and Technology, 7: 290. doi:10.4172/2157-7579.1000290.Viewed PDF 81 5 -
Molecular Mechanisms to Identify Anticancer Activity of Tomentin in A549 Lung Adeno Carcinoma Cells: Role of p53/Caspase-Mediated PathwaysAuthor: Vishnu Priya VeeraraghavanDOI: 10.21522/TIJPH.2013.SE.24.03.Art012
Molecular Mechanisms to Identify Anticancer Activity of Tomentin in A549 Lung Adeno Carcinoma Cells: Role of p53/Caspase-Mediated Pathways
Abstract:
Cancer, a multifaceted disease with increasing prevalence, remains a significant global health concern. Lung cancer, in particular, presents a formidable challenge due to its high mortality rates. Tomentin, a phytochemical extracted from Sphaeralcea angustifolia, has garnered interest for its potential anti-cancer properties. This study employs both in vitro and in silico methods to elucidate tomentin's efficacy against lung cancer, focusing on the A549 cell line, a model for lung adenocarcinoma. The study begins by exploring the cytotoxic effects of tomentin on A549 cells through viability assays, apoptosis induction, and molecular pathway modulation. Results indicate dose-dependent inhibition of cell proliferation and activation of apoptotic pathways by tomentin treatment. Further analysis reveals tomentin's ability to scavenge DPPH radicals and inhibit protein denaturation, suggesting potent antioxidant and anti-inflammatory properties. Moreover, mRNA expression analysis demonstrates tomentin's regulatory effects on key genes involved in inflammation and apoptosis. Molecular docking studies reveal strong binding affinity between tomentin and critical proteins implicated in cancer progression, including MCL1, p53, Bcl-2, and Caspases.
Molecular Mechanisms to Identify Anticancer Activity of Tomentin in A549 Lung Adeno Carcinoma Cells: Role of p53/Caspase-Mediated Pathways
References:
[1] Roy, P. S., & Saikia, B. J., 2016, Cancer and cure: A critical analysis. Indian journal of cancer, 53(3), 441–442. https://doi.org/10.4103/0019-509X.200658[2] Hanahan, D., & Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell, 144(5), 646–674. https://doi.org/10.1016/j.cell.2011.02.013[3] Jayaraman, S., Natarajan, S. R., Ponnusamy, B., Veeraraghavan, V. P., & Jasmine, S., 2023, Unlocking the potential of beta sitosterol: Augmenting the suppression of oral cancer cells through extrinsic and intrinsic signalling mechanisms. The Saudi Dental Journal, 35(8), 1007-1013.[4] Brody H., 2014, Lung cancer. Nature, 513(7517), S1. https://doi.org/10.1038/513S1.[5] Bilello, K. S., Murin, S., & Matthay, R. A., 2002, Epidemiology, etiology, and prevention of lung cancer. Clinics in chest medicine, 23(1), 1-25.[6] Hammerschmidt, S., & Wirtz, H., 2009, Lung cancer: current diagnosis and treatment. Deutsches Arzteblatt international, 106(49), 809–820. https://doi.org/10.3238/arztebl.2009.0809[7] Rivera, M. P., Detterbeck, F., & Mehta, A. C., 2003, Diagnosis of lung cancer: the guidelines. Chest, 123(1), 129S-136S.[8] Maghfoor, I., & Perry, M. C., 2005, Lung cancer. Annals of Saudi medicine, 25(1), 1–12. https://doi.org/10.5144/0256-4947.2005.1[9] Bertrams, W., Hönzke, K., Obermayer, B., Tönnies, M., Bauer, T. T., Schneider, P., Neudecker, J., Rückert, J. C., Stiewe, T., Nist, A., Eggeling, S., Suttorp, N., Wolff, T., Hippenstiel, S., Schmeck, B., & Hocke, A. C., 2022, Transcriptomic comparison of primary human lung cells with lung tissue samples and the human A549 lung cell line highlights cell type specific responses during infections with influenza A virus. Scientific reports, 12(1), 20608. https://doi.org/10.1038/s41598-022-24792-4[10] Jayaraman, S., Devarajan, N., Rajagopal, P., Babu, S., Ganesan, S. K., Veeraraghavan, V. P., Palanisamy, C. P., Cui, B., Periyasamy, V., & Chandrasekar, K., 2021, β-Sitosterol Circumvents Obesity Induced Inflammation and Insulin Resistance by down-Regulating IKKβ/NF-κB and JNK Signaling Pathway in Adipocytes of Type 2 Diabetic Rats. Molecules (Basel, Switzerland), 26(7), 2101. https://doi.org/10.3390/molecules26072101.[11] Yeung, Y. T., Aziz, F., Guerrero-Castilla, A., & Arguelles, S., 2018, Signaling Pathways in Inflammation and Anti-inflammatory Therapies. Current pharmaceutical design, 24(14), 1449–1484. https://doi.org/10.2174/1381612824666180327165604.[12] Huq, A. K. M. M., Roney, M., Imran, S., Khan, S. U., Uddin, M. N., Htar, T. T., Baig, A.[13] Barrera, K., González-Cortazar, M., Reyes-Pérez, R., Pérez-García, D., Herrera-Ruiz, M., Arellano-García, J., Cruz-Sosa, F., & Nicasio-Torres, P., 2023, Production of Two Isomers of Sphaeralcic Acid in Hairy Roots from Sphaeralcea angustifolia. Plants (Basel, Switzerland), 12(5), 1090. https://doi.org/10.3390/plants12051090.[14] Gatasheh, M. K., Natarajan, S. R., Krishnamoorthy, R., Alsulami, T. S., Rajagopal, P., Palanisamy, C. P., & Jayaraman, S., 2024, Molecular analysis to identify novel potential biomarkers as drug targets in colorectal cancer therapy: an integrated bioinformatics analysis. Molecular & Cellular Oncology, 11(1), 2326699.[15] Hatano, T., Edamatsu, R., Hiramatsu, M., MORI, A., Fujita, Y., Yasuhara, T., ... & OKUDA, T.,1989, Effects of the interaction of tannins with co-existing substances. VI.: effects of tannins and related polyphenols on superoxide anion radical, and on 1, 1-Diphenyl-2-picrylhydrazyl radical. Chemical and pharmaceutical bulletin, 37(8), 2016-2021.[16] Padmanabhan, P., & Jangle, S. N., 2012, Evaluation of in-vitro anti-inflammatory activity of herbal preparation, a combination of four medicinal plants. International journal of basic and applied medical sciences, 2(1), 109-116.[17] Elias, G., & Rao, M. N., 1988, Inhibition of albumin denaturation and antiinflammatory activity of dehydrozingerone and its analogs. Indian journal of experimental biology, 26(7), 540–542.[18] Perumal, S., Langeshwaran, K., Selvaraj, J., Ponnulakshmi, R., Shyamaladevi, B., & Balasubramanian, M. P., 2018, Effect of diosmin on apoptotic signaling molecules in N-nitrosodiethylamine-induced hepatocellular carcinoma in experimental rats. Molecular and Cellular Biochemistry, 449, 27-37.[19] Lavanya, M., Krishnamoorthy, R., Alshuniaber, M. A., Manoharadas, S., Palanisamy, C. P., Veeraraghavan, V. P., & Padmini, R., 2023, Formulation, characterization and evaluation of gelatin-syringic acid/zinc oxide nanocomposite for its effective anticancer, antioxidant and anti-inflammatory activities. Journal of King Saud University-Science, 35(8), 102909.[20] Jayaraman, S., & Veeraraghavan, V. P., 2023, Nucleostemin and p-STAT3 as early diagnostic potential markers in oral squamous cell carcinoma. Acta Marisiensis-Seria Medica, 69(4), 241-243.[21] Pei, J., Yan, Y., Jayaraman, S., Rajagopal, P., Natarajan, P. M., Umapathy, V. R., ... & Mironescu, M., 2024, A review on advancements in the application of starch-based nanomaterials in biomedicine: Precision drug delivery and cancer therapy. International Journal of Biological Macromolecules, 130746.[22] Nishida, J., & Kawabata, J., 2006, DPPH radical scavenging reaction of hydroxy- and methoxychalcones. Bioscience, Biotechnology, and Biochemistry, 70(1), 193–202. https://doi.org/10.1271/bbb.70.193[23] Ojiako, O. A., Chikezie, P. C., & Ogbuji, A. C., 2015, Radical scavenging potentials of single and combinatorial herbal formulations in vitro. Journal of Traditional and Complementary Medicine, 6(2), 153–159. https://doi.org/10.1016/j.jtcme.2014.11.037[24] Szerlauth, A., Muráth, S., Viski, S., & Szilagyi, I., 2019, Radical scavenging activity of plant extracts from improved processing. Heliyon, 5(11), e02763. https://doi.org/10.1016/j.heliyon.2019.e02763.[25] Zhang, Y., Kim, Y. H. B., Puolanne, E., & Ertbjerg, P., 2022, Role of freezing-induced myofibrillar protein denaturation in the generation of thaw loss: A review. Meat Science, 190, 108841. https://doi.org/10.1016/j.meatsci.2022.108841.[26] Ruzza, P., Honisch, C., Hussain, R., & Siligardi, G., 2021, Free Radicals and ROS Induce Protein Denaturation by UV Photostability Assay. International Journal of Molecular Sciences, 22(12), 6512. https://doi.org/10.3390/ijms22126512.[27] Johnson, F H., & Campbell, D. H., 1946, Pressure and protein denaturation. The Journal of Biological Chemistry, 163, 689–698.[28] Chaoul, N., & Albanesi, M., 2021, Tumor Infiltrating T Cell Cytotoxicity Assay. Methods in Molecular Biology (Clifton, N.J.), 2325, 41–54. https://doi.org/10.1007/978-1-0716-1507-2_3.[29] Olatunde, O. Z., Yong, J., & Lu, C., 2023, Chemical Constituents from the Roots of Jasminum sambac (L.) Ait. and their Cytotoxicity to the Cancer Cell Lines. Anti-cancer Agents in Medicinal Chemistry, 23(16), 1860–1865. https://doi.org/10.2174/1871520623666230504102455.[30] Lee, H., Jeon, Y., Moon, H., Lee, E. H., Lee, T. H., & Kim, H., 2023, Synthesis of 1,4-Dialkoxynaphthalene-Based Imidazolium Salts and Their Cytotoxicity in Cancer Cell Lines. International Journal of Molecular Sciences, 24(3), 2713. https://doi.org/10.3390/ijms24032713.[31] Sreevarun, M., Ajay, R., Suganya, G., Rakshagan, V., Bhanuchander, V., & Suma, K., 2023, Formulation, Configuration, and Physical Properties of Dental Composite Resin Containing a Novel 2π + 2π Photodimerized Crosslinker - Cinnamyl Methacrylate: An In Vitro Research. The Journal of Contemporary Dental Practice, 24(6), 364–371. https://doi.org/10.5005/jp-journals-10024-3480[32] Alam, M. K., Alqhtani, N. R., Alnufaiy, B., Alqahtani, A. S., Elsahn, N. A., Russo, D., Di Blasio, M., Cicciù, M., & Minervini, G., 2024, A systematic review and meta-analysis of the impact of resveratrol on oral cancer: potential therapeutic implications. BMC Oral Health, 24(1), 412. https://doi.org/10.1186/s12903-024-04045-8.[33] Yadalam, P. K., Arumuganainar, D., Ronsivalle, V., Di Blasio, M., Badnjevic, A., Marrapodi, M. M., Cervino, G., & Minervini, G., 2024, Prediction of interactomic hub genes in PBMC cells in type 2 diabetes mellitus, dyslipidemia, and periodontitis. BMC Oral Health, 24(1), 385. https://doi.org/10.1186/s12903-024-04041-y.[34] Jayaraman, S., Natarajan, S.R., Veeraraghavan, V.P., Jasmine, S., 2023, Unveiling the anti-cancer mechanisms of calotropin: Insights into cell growth inhibition, cell cycle arrest, and metabolic regulation in human oral squamous carcinoma cells (HSC-3). Journal of Oral Biology and Craniofacial Research, 13:704-713. 10.1016/j.jobcr.2023.09.002.[35] Zhang, W., Tian, W., Wang, Y., Jin, X., Guo, H., Wang, Y., Tang, Y., & Yao, X., 2022, Explore the mechanism and substance basis of Mahuang FuziXixin Decoction for the treatment of lung cancer based on network pharmacology and molecular docking. Computers in Biology and Medicine, 151(Pt A), 106293. https://doi.org/10.1016/j.compbiomed.2022.106293.[36] Arjmand, B., Hamidpour, S. K., Alavi-Moghadam, S., Yavari, H., Shahbazbadr, A., Tavirani, M. R., ... & Larijani, B., 2022, Molecular docking as a therapeutic approach for targeting cancer stem cell metabolic processes. Frontiers in Pharmacology, 13, 768556.[37] Sharma, K. K., Singh, B., Mujwar, S., & Bisen, P. S., 2020, Molecular docking based analysis to elucidate the DNA topoisomerase IIβ as the potential target for the ganoderic acid: A natural therapeutic agent in cancer therapy. Current Computer-Aided Drug Design, 16(2), 176-189.[38] Rajendran, P., Sekar, R., Zahra, H. A., Jayaraman, S., Rajagopal, P., Abdallah, B. M., Ali, E. M., Abdelsalam, S. A., & Veeraraghavan, V., 2023, Salivaomics to decode non-coding RNAs in oral cancer. A narrative review. Non-coding RNA Research, 8(3), 376–384. https://doi.org/10.1016/j.ncrna.2023.05.001.[39] Jayaraman, S., Natararaj, S., & Veeraraghavan, V. P., 2024, Hesperidin Inhibits Oral Cancer Cell Growth via Apoptosis and Inflammatory Signaling-Mediated Mechanisms: Evidence From In vitro and In Silico Analyses. Cureus, 16(2), e53458Viewed PDF 78 3 -
Studies on Anti-inflammatory and Anti-diabetic Potential of Andrographolide: Evidence from an In vitro, In silico and In vivo StudyAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art013
Studies on Anti-inflammatory and Anti-diabetic Potential of Andrographolide: Evidence from an In vitro, In silico and In vivo Study
Abstract:
Insulin function and sensitivity are compromised in type 2 diabetes (T2DM) due to various factors causing cellular stress and inflammation. With the increasing recognition of inflammation's role in both T1DM and T2DM, anti-inflammatory strategies are gaining importance in disease management. This study investigates the relationship between andrographolide and the enzymes α-glucosidase and α-amylase to elucidate its antidiabetic benefits. The study also evaluates andrographolide's ability to inhibit protein denaturation and examines its effects on the liver of T2DM rats through histological analysis. Methods included in vitro antidiabetic and anti-inflammatory activity assessments using α-glucosidase, α-amylase, and protein denaturation inhibition methods. Histopathological analysis of liver tissue from streptozotocin (STZ) and high-fat diet (HFD)-induced type-2 diabetic rats was conducted. In silico docking analysis was performed to confirm the binding affinity of andrographolide with pro-inflammatory signaling molecules. Data were analyzed using one-way ANOVA. Results indicated that molecular docking showed a good binding affinity with selected protein targets, attesting to andrographolide's powerful anti-inflammatory and antidiabetic effects. Histological analysis demonstrated that andrographolide could restore the hepatic architecture of diabetic livers. The in silico study further demonstrated high binding affinity against protein targets related to inflammatory and insulin signaling pathways. In conclusion, andrographolide may provide a promising basis for developing novel treatments and identifying critically needed pharmaceutical targets to address inflammation-related clinical problems in diabetes.
Studies on Anti-inflammatory and Anti-diabetic Potential of Andrographolide: Evidence from an In vitro, In silico and In vivo Study
References:
[1] Ma, Q., Li, Y., Li, P., Wang, M., Wang, J., Tang, Z., Wang, T., Luo, L., Wang, C., Wang, T., & Zhao, B., 2019, Research progress in the relationship between type 2 diabetes mellitus and intestinal flora. Biomedicine & pharmacotherapy, 117, 109138. https://doi.org/10.1016/j.biopha.2019.109138.[2] Roep, B. O., Thomaidou, S., van Tienhoven, R., & Zaldumbide, A., 2021, Type 1 diabetes mellitus as a disease of the β-cell (do not blame the immune system?). Nature reviews. Endocrinology, 17(3), 150–161. https://doi.org/10.1038/s41574-020-00443-4.[3] Rebecca Roy, J., Janaki, C. S., Jayaraman, S., Periyasamy, V., Balaji, T., Vijayamalathi, M., Veeraraghavan, V. P., Krishnamoorthy, K., & Prasad, M., 2023, Carica Papaya Reduces High Fat Diet and Streptozotocin-Induced Development of Inflammation in Adipocyte via IL-1β/IL-6/TNF-α Mediated Signaling Mechanisms in Type-2 Diabetic Rats. Current Issues in Molecular Biology, 45(2), 852–884. https://doi.org/10.3390/cimb45020056.[4] Kandanur, S. G. S., Tamang, N., Golakoti, N. R., & Nanduri, S., 2019, Andrographolide: A natural product template for the generation of structurally and biologically diverse diterpenes. European Journal of Medicinal Chemistry, 176, 513–533. https://doi.org/10.1016/j.ejmech.2019.05.022.[5] Jayakumar, T., Hsieh, C. Y., Lee, J. J., & Sheu, J. R., 2013, Experimental and Clinical Pharmacology of Andrographis paniculata and Its Major Bioactive Phytoconstituent Andrographolide. Evidence-Based Complementary and Alternative Medicine : eCAM, 2013, 846740. https://doi.org/10.1155/2013/846740.[6] Premanath, R., Nanjaiah, L., 2015, Antidiabetic and antioxidant potential of Andrographis paniculata Nees. leaf ethanol extract in streptozotocin induced diabetic rats. Journal of Applied Pharmaceutical Science, 5:069-076. 10.7324/JAPS.2015.50113.[7] Tan, W. S. D., Liao, W., Zhou, S., & Wong, W. S. F., 2017, Is there a future for andrographolide to be an anti-inflammatory drug? Deciphering its major mechanisms of action. Biochemical Pharmacology, 139, 71–81. https://doi.org/10.1016/j.bcp.2017.03.024.[8] Vishaka, S., Sridevi, G., & Selvaraj, J., 2022, An in vitro analysis on the antioxidant and anti-diabetic properties of Kaempferia galanga rhizome using different solvent systems. Journal of Advanced Pharmaceutical Technology & Research, 13(2), S505–S509. https://doi.org/10.4103/japtr.japtr_189_22.[9] Prakasam, N.V., Devi, R.G., Selvaraj, J., 2022, In-vitro Antidiabetic on leaf extracts of Mimosa pudica and Euphorbia hirta-A Comparative Study. Research Journal of Pharmacy and Technology, 15:5459-5463. 10.52711/0974-360X.2022.00920.[10] Kaddour, S.M., Arrar, L., Baghiani, A., 2020, Anti-Inflammatory Potential Evaluation (In-Vitro and In-Vivo) of Arthrophytum scoparium Aerial Part. JDDT, 10: 213-218. 10.22270/jddt.v10i5.4409.[11] Neralla, M., M, H., Preethi, A., Selvakumar, S. C., & Sekar, D., 2024, Expression levels of microRNA-7110 in oral squamous cell carcinoma. Minerva dental and oral science, 73(3), 155–160. https://doi.org/10.23736/S2724-6329.23.04801-5.[12] Nugroho, A. E., Andrie, M., Warditiani, N. K., Siswanto, E., Pramono, S., & Lukitaningsih, E., 2012, Antidiabetic and antihiperlipidemic effect of Andrographis paniculata (Burm. f.) Nees and andrographolide in high-fructose-fat-fed rats. Indian Journal of Pharmacology, 44(3), 377–381. https://doi.org/10.4103/0253-7613.96343.[13] Roy, J. R., Janaki, C. S., Jayaraman, S., Periyasamy, V., Balaji, T., Vijayamalathi, M., & Veeraraghavan, V. P., 2022, Carica papaya Reduces Muscle Insulin Resistance via IR/GLUT4 Mediated Signaling Mechanisms in High Fat Diet and Streptozotocin-Induced Type-2 Diabetic Rats. Antioxidants (Basel, Switzerland), 11(10), 2081. https://doi.org/10.3390/antiox11102081.[14] Roy, J. R., Janaki, C. S., Jayaraman, S., Veeraraghavan, V. P., Periyasamy, V., Balaji, T., Vijayamalathi, M., Bhuvaneswari, P., & Swetha, P., 2023,Hypoglycemic Potential of Carica papaya in Liver Is Mediated through IRS-2/PI3K/SREBP-1c/GLUT2 Signaling in High-Fat-Diet-Induced Type-2 Diabetic Male Rats. Toxics, 11(3), 240. https://doi.org/10.3390/toxics11030240.[15] Salehi, B., Ata, A., V Anil Kumar, N., Sharopov, F., Ramírez-Alarcón, K., Ruiz-Ortega, A., Abdulmajid Ayatollahi, S., Tsouh Fokou, P. V., Kobarfard, F., Amiruddin Zakaria, Z., Iriti, M., Taheri, Y., Martorell, M., Sureda, A., Setzer, W. N., Durazzo, A., Lucarini, M., Santini, A., Capasso, R., Ostrander, E. A., Sharifi-Rad, J., 2019, Antidiabetic Potential of Medicinal Plants and Their Active Components. Biomolecules, 9(10), 551. https://doi.org/10.3390/biom9100551.[16] Mwakalukwa, R., Amen, Y., Nagata, M., & Shimizu, K., 2020, Postprandial Hyperglycemia Lowering Effect of the Isolated Compounds from Olive Mill Wastes - An Inhibitory Activity and Kinetics Studies on α-Glucosidase and α-Amylase Enzymes. ACS omega, 5(32), 20070–20079. https://doi.org/10.1021/acsomega.0c01622.[17] Indu, S., Vijayalakshmi, P., Selvaraj, J., & Rajalakshmi, M., 2021, Novel Triterpenoids from Cassia fistula Stem Bark Depreciates STZ-Induced Detrimental Changes in IRS-1/Akt-Mediated Insulin Signaling Mechanisms in Type-1 Diabetic Rats. Molecules (Basel, Switzerland), 26(22), 6812. https://doi.org/10.3390/molecules26226812.[18] Ramalingam, K., Yadalam, P. K., Ramani, P., Krishna, M., Hafedh, S., Badnjević, A., Cervino, G., & Minervini, G., 2024, Light gradient boosting-based prediction of quality of life among oral cancer-treated patients. BMC Oral Health, 24(1), 349. https://doi.org/10.1186/s12903-024-04050-x.[19] Dirir, A. M., Daou, M., Yousef, A. F., & Yousef, L. F., 2022, A review of alpha-glucosidase inhibitors from plants as potential candidates for the treatment of type-2 diabetes. Phytochemistry Peviews : Proceedings of the Phytochemical Society of Europe, 21(4), 1049–1079. https://doi.org/10.1007/s11101-021-09773-1.[20] Kashtoh, H., & Baek, K. H., 2022, Recent Updates on Phytoconstituent Alpha-Glucosidase Inhibitors: An Approach towards the Treatment of Type Two Diabetes. Plants (Basel, Switzerland), 11(20), 2722. https://doi.org/10.3390/plants11202722.[21] American Diabetes Association., 2020, Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2020. Diabetes Care, 43(Suppl 1), S98–S110. https://doi.org/10.2337/dc20-S009.[22] Somtimuang, C., Olatunji, O. J., & Ovatlarnporn, C., 2018, Evaluation of In Vitro α-Amylase and α-Glucosidase Inhibitory Potentials of 14 Medicinal Plants Constituted in Thai Folk Antidiabetic Formularies. Chemistry & Biodiversity, 15(4), e1800025. https://doi.org/10.1002/cbdv.201800025.[23] Kurutas E. B., 2016, The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nutrition Journal, 15(1), 71. https://doi.org/10.1186/s12937-016-0186-5.[24] Salve, P., Vinchurkar, A., Raut, R., Chondekar, R., Lakkakula, J., Roy, A., Hossain, M. J., Alghamdi, S., Almehmadi, M., Abdulaziz, O., Allahyani, M., Dablool, A. S., Sarker, M. M. R., & Nur Azlina, M. F., 2022, An Evaluation of Antimicrobial, Anticancer, Anti-Inflammatory and Antioxidant Activities of Silver Nanoparticles Synthesized from Leaf Extract of Madhuca longifolia Utilizing Quantitative and Qualitative Methods. Molecules (Basel, Switzerland), 27(19), 6404. https://doi.org/10.3390/molecules27196404.[25] Dharmadeva, S., Galgamuwa, L. S., Prasadinie, C., & Kumarasinghe, N., 2018, In vitro anti-inflammatory activity of Ficus racemosa L. bark using albumin denaturation method. Ayu, 39(4), 239–242. https://doi.org/10.4103/ayu.AYU_27_18.[26] Dsouza, M.R., Athoibi, S., Prabha, S., 2020, Pharmacognostical Investigation of Andrographis paniculata (Green Chiretta) and Crystallization of the Bioactive component Andrographolide. Int. J. Pharmtech Res. 13: 40-50. 10.20902/ijptr.2019.130207.[27] Harsha, L., & Subramanian, A. K., 02022, Comparative Assessment of pH and Degree of Surface Roughness of Enamel When Etched with Five Commercially Available Etchants: An In Vitro Study. The Journal of Contemporary Dental Practice, 23(2), 181–185.[28] Roy, J. R., Janaki, C. S., Jayaraman, S., Veeraraghavan, V. P., Periyasamy, V., Balaji, T., Vijayamalathi, M., Bhuvaneswari, P., & Swetha, P., 2023, Hypoglycemic Potential of Carica papaya in Liver Is Mediated through IRS-2/PI3K/SREBP-1c/GLUT2 Signaling in High-Fat-Diet-Induced Type-2 Diabetic Male Rats. Toxics, 11(3), 240. https://doi.org/10.3390/toxics11030240.[29] Motshakeri, M., Ebrahimi, M., Goh, Y. M., Othman, H. H., Hair-Bejo, M., & Mohamed, S. (2014). Effects of Brown Seaweed (Sargassum polycystum) Extracts on Kidney, Liver, and Pancreas of Type 2 Diabetic Rat Model. Evidence-Based Complementary and alternative medicine : eCAM, 2014, 379407. https://doi.org/10.1155/2014/379407.Viewed PDF 80 9 -
Fabrication and Characterization of a Novel Dental Filling Herbal Composite Using Biphasic Calcium Phosphate and Leaf Extracts of Cassia occidentalisAuthor: Sridevi GopathyDOI: 10.21522/TIJPH.2013.SE.24.03.Art014
Fabrication and Characterization of a Novel Dental Filling Herbal Composite Using Biphasic Calcium Phosphate and Leaf Extracts of Cassia occidentalis
Abstract:
In the field of Biomedicine, allogeneic grafts that possess excellent biocompatibility and immuno-compatibility play a major role in the treatment of untreated dental bone defects. Filling these defects with bone substitute material prevents resorption of bone, preserves the alveolar ridge, and provides sufficient bone for immediate or subsequent implant placement. In this regard, the present study focused on the fabrication of a novel herbal bone graft material rich in biopolymers and phytochemicals was used as bone graft. The bone graft material was synthesized using biphasic calcium phosphate, casein, chitosan and ethanolic leaf extracts of Cassia Occidentalis. The prepared bone graft was subjected to various characterizations like FTIR, X-ray diffraction, thermos-gravimetric analysis, scanning electron microscopy to show its chemical composition, surface morphology, stability, mechanical strength to show its chemical composition, stability and porosity and GCMS analysis, anti-microbial, anti-oxidant, anti-inflammatory activity to reveal its bioactive components. Results revealed that the prepared bone graft of Cassia Occidentalis showed excellent osteogenic and can be well suggested for various biomedical applications like orthopedics, dental fillings, bone tissue engineering and in the treatment of rheumatoid arthritis. It also replaces the use of autogenous graft with high biocompatibility and osteogenesis.
Fabrication and Characterization of a Novel Dental Filling Herbal Composite Using Biphasic Calcium Phosphate and Leaf Extracts of Cassia occidentalis
References:
[1] Christel, P, Meunier, A, Dorlot, J, M, Crolet, J, M, Witvoet, J, Sedel, L, Boutin, P, 1988, Biomechanical compatibility and design of ceramic implants for orthopedic surgery. Annals of the New York Academy of Sciences, ,523, 234–256. doi: 10.1111/j.1749-6632.1988.tb38516.x.[2] Kumar, S, Desai, N, Joshi, S, Hirani, T, Gajjar, S, Patel, C, Bhakkand, S, R, Girdhar, G, A, Govindool, S, R, Wan Ahmad Fakuradzi WFS, Haque, M, 2022, Biphasic Calcium Phosphate Versus Demineralized Freeze-Dried Bone Allograft in the Treatment of Periodontal Disease: A Clinical and Radiographical Evaluation, Cureus, 14(9), 291-31, doi: 10.7759/cureus.29131. PMID: 36133502; PMCID: PMC9470539.[3] Patlolla, A, Arinzeh, T. L., 2014, Evaluating apatite formation and osteogenic activity of electrospun composites for bone tissue engineering. Biotechnology and Bioengineering, 111(5), 1000-17, doi: 10.1002/bit.25146. Epub 2013 Nov 22. PMID: 24264603.[4] Baskar, K, Saravana Karthikeyan, B, Gurucharan, I, et al., 2022, Eggshell derived nano-hydroxyapatite incorporated carboxymethyl chitosan scaffold for dentine regeneration: A laboratory investigation. International Endonditics Journal, 55(1), 89-102, doi:10.1111/iej.13644[5] Kunam, D, Sampath, V, Manimaran, S, Sekar, M, 2019, Effect of Indigenously Developed Nano-Hydroxyapatite Crystals from Chicken Egg Shell on the Surface Hardness of Bleached Human Enamel: An In Vitro Study. Contemporary Clinical Dentistry, 10(3), 489-493, doi:10.4103/ccd.ccd_810_18=[6] Selvakumar Kritika, B, Jothimani, Sampath Vidhya, Kavitha Sanjeev, Sekar Mahalaxmi, Venkatachalapathy, B, Sureshkumar, S, 2021, Incorporation of hydrophobic nanochitosan improves wear resistance of dental sealants, International Journal of Polymeric Materials and Polymeric Biomaterials, 70 (5), 309-17, DOI: 10.1080/00914037.2020.1713782[7] Mohammed, M.; Aboki, M, A, Saidu, H,.M, Victor, O, Tawakalitu, A, Maikano, S,A, 2012, Phytochemical and Some Antimicrobial Activity of Cassia Occidentalis L. (Caesalpiniaceae), International Journal of Science and Technology, 2, 4.[8] Bouyer, E, Gitzhofer, F, Boulos, M, I, 2000, Morphological study of hydroxyapatite nanocrystal suspension, Journal of Materials Science: Materials in Medicine, 11(8), 523-531.[9] Krithiga, G, Antaryami Jena, Selvamani, P, Sastry, T, P, 2011, In vitro study on biomineralization of biphasic calcium phosphate biocomposite crosslinked with hydrolysable tannins of Terminalia chebula, Bulletin of Material Sciences, 34(3), 589–594.[10] Lim, G, K, Wang, J, Ng, Sc, Chew, C, H, Gan, L, M, 1999, Formation of nanocrystalline hydroxyapatite in nonionic surfactant emulsions, Langmuir, 15(22), 7472-7477.[11] Lambert, B, J, Mendelson, T, A, Craven, M, D, 2011, Radiation and ethylene oxide terminal sterilization experiences with drug eluting stent products, AAPS Pharmacy Science and Technology, 12(4), 1116-1126, doi:10.1208/s12249-011-9644-8.[12] Prieto, P, Pineda, M, Aguilar, M, 1999, Spectrophotometric quantitation of antioxidant capacity through the formation of a phosphomolybdenum complex: specific application to the determination of vitamin E, Annals of Biochemistry, 269(2), 337-41.[13] Williams, L, A, O'Connar, A, Latore, L, Dennis, O, Ringer, S, Whittaker, J, A, Conrad, J, Vogler, B, Rosner, H, Kraus, W, 2008, The in vitro anti-denaturation effects induced by natural products and non-steroidal compounds in heat treated (immunogenic) bovine serum albumin is proposed as a screening assay for the detection of anti-inflammatory compounds, without the use of animals, in the early stages of the drug discovery process, West Indian Medical Journal, 57(4), 327-31, PMID: 19566010[14] Sridevi, G, Sembulingam, K, Muhammed Ibrahim, Srividya, S, Prema Sembulingam, 2015, Evaluation of in vitro anti-inflammatory activity of pergularia daemia, World Journal of Pharmaceutical Research, 4(6), 1100-1108.[15] Wu X, H, Cai, J, J, Ruan, J, L, Lou, J, S, Duan, H, Q, et al., 2011, Acetylated anthraquinone glycosides from Cassia obtusifolia, Journal of Asian Natural Product Research, 13(6), 486-491.[16] J, E, Kelmanson, A, K, Jager, J, Van-Staden Zulu, 2000, Medicinal plants with antibacterial activity, Journal of Ethnopharmacology, 69, 241-246.[17] Zakaria, M., Tahereh, E, 2012, Anti-fungal activity of cold and hot water extracts of spices against fungal pathogens of roselle (Hibiscus sabdariffa) in vitro, Microbiology and Pathology, 52, 125–129, doi: 10.1016/j.micpath.2011.11.001.[18] Bajpai, S, Farhan, Ferooz, Shah, Manjula, Bajpai, Mamta, Jadaun, Pooja, Jyotishi, & Shah, Farhan, 2017, Dynamic release of Amoxicillin from Orally Dissolving Film (ODF) composed of Casein and Sodium alginate. Journal of Drug Research and Development, 3, 134.[19] Santhosh Kumar, B, Thangavelu Muthukumar, Deepachitra, R, Charumathy, R, K, Hemalatha, T, Sastry, T, P, 2014, In-vitro evaluation of biphasic calcium phosphate/casein incorporated with Myristica fragrans for bone tissue engineering, Ceramics International, 41 (1), 1725–1734.[20] Mediaswanti, K, Wen, C, Ivanova, E, P, Berndt, C, C, Malherbe, F, et al., 2013, .A Review on Bioactive Porous Metallic Biomaterials, Journal of Biomimetic and Biomaterial Tissue Engineering, 18, 104, doi:10.4172/1662-100X.1000104[21] Ramalingam, K., Yadalam, P. K., Ramani, P., Krishna, M., Hafedh, S., Badnjević, A., Cervino, G., & Minervini, G., 2024, Light gradient boosting-based prediction of quality of life among oral cancer-treated patients. BMC Oral Health, 24(1), 349. https://doi.org/10.1186/s12903-024-04050-x.[22] Nakkeeran, K, P, Saravanan, K, Babu, P, John, R, R, 2019, Evaluation of bone regeneration in periapical osseous defects with and without platelet rich plasma, combined calcium sulfate and autologous bone graft - A comparative study, Journal of Stomatology Oral Maxillofacial Surgery, 120(3), 196-202, doi:10.1016/j.jormas.2018.11.008[23] Harsha, L., & Subramanian, A. K., 2022, Comparative Assessment of pH and Degree of Surface Roughness of Enamel When Etched with Five Commercially Available Etchants: An In Vitro Study. The journal of contemporary dental practice, 23(2), 181–185.[24] Siva, Suvetha, Sivakumar, Arvind Kumar, Aravind, Dinesh, Saravana. 2018, Cytotoxicity of SK surgical implants used for anchorage in orthodontics: In vitro study. Drug Invention Today,10, 2927-2929.[25] Neralla, M., M, H., Preethi, A., Selvakumar, S. C., & Sekar, D., 2024, Expression levels of microRNA-7110 in oral squamous cell carcinoma. Minerva dental and oral science, 73(3), 155–160. https://doi.org/10.23736/S2724-6329.23.04801-5.[26] Mohan, S, Srinivasan, D, Arumugam, E, Devasahayam, D, Kannan, R, 2022, Effect of placement angle, diameter, length and bone density on the pull-out strength of orthodontic mini-implants: An in vitro study, Journal of Orthodontics, 49(2), 143-150, doi:10.1177/14653125211053338.[27] Gopathy Sridevi, Seshadri Srividya, 2022, Novel Dental Implants with Herbal Composites: A Review. Current Concepts in Dental Implantology - From Science to Clinical Research. Intechopen publishers, 1-15, DOI: http://dx.doi.org/10.5772/intechopen.101489[28] Ramsundar, K., Jain, R. K., Balakrishnan, N., & Vikramsimha, B., 2023, Comparative evaluation of bracket bond failure rates of a novel non-primer adhesive with a conventional primer-based orthodontic adhesive - a pilot study. Journal of dental research, dental clinics, dental prospects, 17(1), 35–39. https://doi.org/10.34172/joddd.2023.36953.[29] Krithiga, G, Jena, A, Selvamani, P, Sastry T,P, 2011, In vitro study on biomineralization of biphasic calcium phosphate biocomposite crosslinked with hydrolysable tannins of Terminalia chebula, Bulletin of Materials Science, 34(3), 589-594.[30] Santhosh Kumar, B, Hemalatha, Thiagarajan, Ramani, Deepachitra, Raghavan, Narasimha, Prabu, P, Sastry Thotapalli, 2015, Biphasic calcium phosphate-casein bone graft fortified with Cassia occidentalis for bone tissue engineering and regeneration. Bulletin of Material Science. 38 (1), 259. DOI:10.1007/s12034-014-0799-2[31] Reyes, VC, Opot, SO, Mahendra, S, 2015, Planktonic and biofilm-grown nitrogen-cycling bacteria exhibit different susceptibilities to copper nanoparticles, Environ Toxicol Chem, 34(4), 887–897.[32] Niemirowicz, K, Swiecicka, I, Wilczewska, AZ, et al., 2014, Gold-functionalized magnetic nanoparticles restrict growth of Pseudomonas aeruginosa, Int J Nanomedicine, 9, 2217–2224.Viewed PDF 93 8 -
Antidiabetic and Antioxidant Potential of Ethyl Iso-allocholate is Mediated Through Insulin Receptor/IRS-1/Akt/GLUT 4 Mediated Pathways: In vitro and In Silico MechanismsAuthor: Vishnu Priya VeeraraghavanDOI: 10.21522/TIJPH.2013.SE.24.03.Art015
Antidiabetic and Antioxidant Potential of Ethyl Iso-allocholate is Mediated Through Insulin Receptor/IRS-1/Akt/GLUT 4 Mediated Pathways: In vitro and In Silico Mechanisms
Abstract:
Ethyl iso-allocholate (EIA) has emerged as a compound of interest due to its potential antioxidant and antidiabetic properties. This study aimed to evaluate the antidiabetic and antioxidant potential of EIA through a combination of in vitro assays and in silico analysis. The antioxidant activity of EIA was assessed using the DPPH radical scavenging assay. EIA demonstrated significant antioxidant activity with inhibition percentages of 29% at 100μg, increasing to 88% at 500μg, compared to Vitamin C, the standard antioxidant, which showed 41% and 95% inhibition, respectively. In terms of antidiabetic potential, EIA’s efficacy was evaluated through alpha-amylase and alpha-glucosidase inhibition assays. EIA exhibited dose-dependent inhibition of alpha-amylase, with a maximum inhibition of 71.3% at 50μg, compared to 96% by acarbose, a standard antidiabetic agent. Similarly, in the alpha-glucosidase assay, EIA showed up to 70.25% inhibition at 50μg, while acarbose achieved 95.7%. The cytotoxicity of EIA was assessed in 3T3-L1 cells over 48 hours, indicating a favorable safety profile. Additionally, Real-time PCR analysis revealed that EIA positively modulated the expression of key insulin signaling components (IR, IRS1, Akt, PI3K, and GLUT4) in 3T3-L1 cells. In silico molecular docking studies further supported these findings, showing strong binding affinities of EIA with insulin receptor (IR), IRS1, Akt, GLUT4, and PI3K, with the highest binding affinity observed with GLUT4 (-8.5 kcal/mol) and PI3K (-8.8 kcal/mol). These results suggest that EIA could be a promising candidate for further research into its therapeutic potential for diabetes and oxidative stress management.
Antidiabetic and Antioxidant Potential of Ethyl Iso-allocholate is Mediated Through Insulin Receptor/IRS-1/Akt/GLUT 4 Mediated Pathways: In vitro and In Silico Mechanisms
References:
[1] American Diabetes Association., 2022, Classification and diagnosis of diabetes: Standards of Medical Care in Diabetes—2022. Diabetes Care, 45(Supplement 1), S17-S38. doi:10.2337/dc22-S002.[2] Kregel, K. C., & Zhang, H. J., 2007, An integrated view of oxidative stress in aging: Basic mechanisms, functional effects, and pathological considerations. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 292(1), R18-R36. doi:10.1152/ajpregu.00327.2006.[3] Chiang J. Y., 2017, Recent advances in understanding bile acid homeostasis. F1000Research, 6, 2029. doi.org/10.12688/f1000research.12449.1.[4] White M. F., 2003, Insulin signaling in health and disease. Science (New York, N.Y.), 302(5651), 1710–1711. https://doi.org/10.1126/science.1092952.[5] Lee, S. H., Park, S. Y., & Choi, C. S., 2022, Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes & metabolism journal, 46(1), 15–37. https://doi.org/10.4093/dmj.2021.0280.[6] Manning, B. D., & Cantley, L. C., 2007, AKT/PKB signaling: Navigating downstream. Cell, 129(7), 1261-1274. doi:10.1016/j.cell.2007.06.009.[7] Van Gerwen, J., Shun-Shion, A. S., & Fazakerley, D. J., 2023, Insulin signalling and GLUT4 trafficking in insulin resistance. Biochemical Society transactions, 51(3), 1057–1069. https://doi.org/10.1042/BST20221066.[8] Kim, K. H., Lee, J. H., & Kim, Y. J., 2015, The effect of Ethyl iso-allocholate on glucose uptake and insulin signaling pathways in insulin-resistant cell models. Journal of Diabetes Research, 2015, 472514. doi:10.1155/2015/472514.[9] Al-Shorman, H. M., Abu-Naba'a, L. A., Sghaireen, M. G., & Alam, M. K., 2024, The Effect of Various Preparation and Cementation Techniques of Dental Veneers on Periodontal Status: A Systematic Review and Meta-Analysis. European Journal of Dentistry, 18(2), 458–467. https://doi.org/10.1055/s-0043-1776120.[10] Alam, M. K., Alqhtani, N. R., Alnufaiy, B., Alqahtani, A. S., Elsahn, N. A., Russo, D., Di Blasio, M., Cicciù, M., & Minervini, G., 2024, A systematic review and meta-analysis of the impact of resveratrol on oral cancer: potential therapeutic implications. BMC Oral Health, 24(1), 412. https://doi.org/10.1186/s12903-024-04045-8.[11] Sreevarun, M., Ajay, R., Suganya, G., Rakshagan, V., Bhanuchander, V., & Suma, K., 2023, Formulation, Configuration, and Physical Properties of Dental Composite Resin Containing a Novel 2π + 2π Photodimerized Crosslinker - Cinnamyl Methacrylate: An In Vitro Research. The Journal of Contemporary Dental Practice, 24(6), 364–371. https://doi.org/10.5005/jp-journals-10024-3480.[12] McDougall, G. J., Karonen, M., & Ellis, T., 2005, Alpha-amylase and alpha-glucosidase inhibition by plant phenolics. Journal of Agricultural and Food Chemistry, 53(4), 1334-1340. doi:10.1021/jf048574r.[13] Khan, M. N., Banu, S., & Kaur, H., 2012, Evaluation of alpha-amylase and alpha-glucosidase inhibitory potential of herbal extracts. Journal of Ethnopharmacology, 140(2), 261-265. doi:10.1016/j.jep.2011.12.030.[14] Yin, J., Zhang, H., & Ye, J., 2013, Alpha-glucosidase inhibitors as a treatment for type 2 diabetes: Mechanism and clinical efficacy. European Journal of Pharmacology, 715(1-3), 103-110. doi:10.1016/j.ejphar.2013.04.028.[15] Ning, Y., Zhang, X., & Zhao, Y., 2013, Inhibition of alpha-glucosidase and alpha-amylase by polysaccharides from different species of mushrooms. Journal of Food Science, 78(10), H1586-H1591. doi:10.1111/1750-3841.12215.[16] Kumar, P., Kumar, V., & Vyas, A., 2017, Non-toxic nature of herbal compounds and their potential role in diabetes management. Phytotherapy Research, 31(10), 1531-1545. doi:10.1002/ptr.5882.[17] Sakaue, H., Fukunaga, K., & Nakamura, H., 2010, Insulin signaling pathways and insulin resistance in type 2 diabetes. Journal of Biochemistry, 147(3), 293-303. doi:10.1093/jb/mvp188.[18] Liu, X., Wu, Y., & Zhao, X., 2016, Natural compounds as potential modulators of insulin signaling pathways. Pharmaceutical Research, 33(5), 1224-1232. doi:10.1007/s11095-015-1825-1.[19] Deenadayalan, A., Subramanian, V., Paramasivan, V., Veeraraghavan, V. P., Rengasamy, G., Coiambatore Sadagopan, J., & Jayaraman, S., 2021, Stevioside attenuates insulin resistance in skeletal muscle by facilitating IR/IRS-1/Akt/GLUT 4 signaling pathways: An in vivo and in silico approach. Molecules, 26(24), 7689. doi:10.3390/molecules26247689.[20] Indu, S., Vijayalakshmi, P., Selvaraj, J., & Rajalakshmi, M., 2021, Novel triterpenoids from Cassia fistula stem bark depreciates STZ-induced detrimental changes in IRS-1/Akt-mediated insulin signaling mechanisms in type-1 diabetic rats. Molecules, 26(22), 6812. doi:10.3390/molecules26226812.[21] Roy, J. R., Janaki, C. S., Jayaraman, S., Periyasamy, V., Balaji, T., Vijayamalathi, M., & Veeraraghavan, V. P., 2022, Carica papaya reduces muscle insulin resistance via IR/GLUT4 mediated signaling mechanisms in high fat diet and streptozotocin-induced type-2 diabetic rats. Antioxidants, 11(10), 2081. doi:10.3390/antiox11102081.[22] Selvaraj, J., Sathish, S., Mayilvanan, C., & Balasubramanian, K., 2013, Excess aldosterone-induced changes in insulin signaling molecules and glucose oxidation in gastrocnemius muscle of adult male rat. Molecular and Cellular Biochemistry, 372, 113-126. doi:10.1007/s11010-012-1377-4.[23] Jayaraman, S., Natarajan, S. R., Ponnusamy, B., Veeraraghavan, V. P., & Jasmine, S. (2023). Unlocking the potential of beta sitosterol: Augmenting the suppression of oral cancer cells through extrinsic and intrinsic signalling mechanisms. The Saudi Dental Journal, 35(8), 1007-1013. doi:10.1016/j.sdentj.2023.07.004.Viewed PDF 78 7 -
Molecular and Epigenetic Studies on the Effect of Hesperidin on IRS-1/Akt/GLUT4 Signaling Molecules in the Gastrocnemius Muscle of Streptozotocin-induced Type-2 Diabetic RatsAuthor: Selvaraj JayaramanDOI: 10.21522/TIJPH.2013.SE.24.03.Art016
Molecular and Epigenetic Studies on the Effect of Hesperidin on IRS-1/Akt/GLUT4 Signaling Molecules in the Gastrocnemius Muscle of Streptozotocin-induced Type-2 Diabetic Rats
Abstract:
Diabetes mellitus is a significant global health issue, affecting 425 million people worldwide, with projections estimating an increase to 629 million by 2045. The need for potent pharmacological agents is urgent, as current oral hypoglycemic drugs have adverse side effects. Hesperidin, a bioflavonoid with anti-hyperglycemic and anti-hyperlipidemic properties, offers promise as a natural therapeutic option. This study aimed to evaluate hesperidin's molecular and epigenetic effects on insulin signal transduction in the gastrocnemius muscle of STZ-induced type 2 diabetic rats. Methods involved dividing fully-grown male Wistar rats into five groups: Healthy control, STZ-induced diabetic, Diabetes+Hesperidin (100mg/kg), Diabetes+Metformin (50mg/kg), and Control+Hesperidin. At the experiment's conclusion, blood samples and gastrocnemius muscle tissues were collected to measure fasting blood glucose, serum insulin, antioxidant enzymes, oxidative stress markers, and histopathological and mRNA expression of insulin signalling molecules. Data were analyzed using one-way ANOVA, with significance set at p<0.05. Results indicated that STZ-induced diabetic rats exhibited significant increases in hyperglycemia, hyperinsulinemia, dyslipidemia, and oxidative stress markers, along with reduced superoxide dismutase (SOD) activity. Histopathological analysis revealed reduced muscle fibres and disrupted skeletal fibres. Additionally, mRNA expression of IRS-1, Akt, and GLUT 4 was significantly reduced. Remarkably, hesperidin treatment normalized these altered parameters. In conclusion, hesperidin effectively regulates insulin signalling in skeletal muscle, reducing diabetic risk. Thus, hesperidin shows potential as a therapeutic candidate for treating type 2 diabetes and its associated complications.
Molecular and Epigenetic Studies on the Effect of Hesperidin on IRS-1/Akt/GLUT4 Signaling Molecules in the Gastrocnemius Muscle of Streptozotocin-induced Type-2 Diabetic Rats
References:
[1] Manna, P., & Jain, S. K., 2015, Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metabolic Syndrome and Related Disorders, 13(10), 423–444. https://doi.org/10.1089/met.2015.0095.
[2] Bindu Jacob, & Narendhirakannan, R.T., 2019, Role of medicinal plants in the management of diabetes mellitus: a review. 3 Biotech, 9(1), 4. https://doi.org/10.1007/s13205-018-1528-0.
[3] Kim, J., Wie, M. B., Ahn, M., Tanaka, A., Matsuda, H., & Shin, T., 2019, Benefits of hesperidin in central nervous system disorders: a review. Anatomy & cell biology, 52(4), 369–377. https://doi.org/10.5115/acb.19.119.
[4] Giannini, I., Amato, A., Basso, L., Tricomi, N., Marranci, M., Pecorella, G., Tafuri, S., Pennisi, D., & Altomare, D. F., 2015, Flavonoids mixture (diosmin, troxerutin, hesperidin) in the treatment of acute hemorrhoidal disease: a prospective, randomized, triple-blind, controlled trial. Techniques in Coloproctology, 19(6), 339–345. https://doi.org/10.1007/s10151-015-1302-9.
[5] Prasad, M., Rajagopal, P., Devarajan, N., Veeraraghavan, V. P., Palanisamy, C. P., Cui, B., Patil, S., & Jayaraman, S., 2022, A comprehensive review on high -fat diet-induced diabetes mellitus: an epigenetic view. The Journal of Nutritional Biochemistry, 107, 109037. https://doi.org/10.1016/j.jnutbio.2022.109037.
[6] Georgel, P. T., & Georgel, P. (2021). Where Epigenetics Meets Food Intake: Their Interaction in the Development/Severity of Gout and Therapeutic Perspectives. Frontiers in Immunology, 12, 752359. https://doi.org/10.3389/fimmu.2021.752359.
[7] Drabkin, D.L., Austin, J.H., 1932, Spectrophotometric Studies: I. Spectrophotometric Constants for Common Hemoglobin Derivatives in Human, Dog, and Rabbit Blood. Journal of Biological Chemistry, 1932, 98, 719-733. 10.4236/oalib.1100648.
[8] Bannon P., 1982, Effect of pH on the elimination of the labile fraction of glycosylated hemoglobin. Clinical Chemistry, 28(10), 2183.
[9] Slaoui, M., & Fiette, L., 2011, Histopathology procedures: from tissue sampling to histopathological evaluation. Methods in molecular biology (Clifton, N.J.), 691, 69–82. https://doi.org/10.1007/978-1-60761-849-2_4.
[10] Gabe, M., 1968, Techniques Histologiques, 6th ed.; Massie e Cie: Paris, France, 1113p.
[11] Slaoui, M., & Fiette, L., 2011, Histopathology procedures: from tissue sampling to histopathological evaluation. Methods in Molecular Biology (Clifton, N.J.), 691, 69–82. https://doi.org/10.1007/978-1-60761-849-2_4.
[12] Gancedo, J. M., & Gancedo, C., 1971, Fructose-1,6-diphosphatase, phosphofructokinase and glucose-6-phosphate dehydrogenase from fermenting and non fermenting yeasts. Archiv fur Mikrobiologie, 76(2), 132–138. https://doi.org/10.1007/BF00411787.
[13] Galicia-Garcia, U., Benito-Vicente, A., Jebari, S., Larrea-Sebal, A., Siddiqi, H., Uribe, K. B., Ostolaza, H., & Martín, C., 2020, Pathophysiology of Type 2 Diabetes Mellitus. International Journal of Molecular Sciences, 21(17), 6275. https://doi.org/10.3390/ijms21176275.
[14] Merz, K. E., & Thurmond, D. C., 2020, Role of Skeletal Muscle in Insulin Resistance and Glucose Uptake. Comprehensive Physiology, 10(3), 785–809. https://doi.org/10.1002/cphy.c190029.
[15] Thomas, D. D., Corkey, B. E., Istfan, N. W., & Apovian, C. M., 2019, Hyperinsulinemia: An Early Indicator of Metabolic Dysfunction. Journal of the Endocrine Society, 3(9), 1727–1747. https://doi.org/10.1210/js.2019-00065.
[16] Wilcox G., 2005, Insulin and insulin resistance. The Clinical Biochemist. Reviews, 26(2), 19–39.
[17] Zhao, C., Yang, C., Wai, S. T. C., Zhang, Y., P Portillo, M., Paoli, P., Wu, Y., San Cheang, W., Liu, B., Carpéné, C., Xiao, J., & Cao, H., 2019, Regulation of glucose metabolism by bioactive phytochemicals for the management of type 2 diabetes mellitus. Critical Reviews in Food Science and Nutrition, 59(6), 830–847. https://doi.org/10.1080/10408398.2018.1501658.
[18] Stanley Mainzen Prince, P., & Kamalakkannan, N., 2006, Rutin improves glucose homeostasis in streptozotocin diabetic tissues by altering glycolytic and gluconeogenic enzymes. Journal of Biochemical and Molecular Toxicology, 20(2), 96–102. https://doi.org/10.1002/jbt.20117.
[19] Latha, M., & Pari, L., 2003, Antihyperglycaemic effect of Cassia auriculata in experimental diabetes and its effects on key metabolic enzymes involved in carbohydrate metabolism. Clinical and Experimental Pharmacology & Physiology, 30(1-2), 38–43. https://doi.org/10.1046/j.1440-1681.2003.03785.x.
[20] Peng, P., Jin, J., Zou, G., Sui, Y., Han, Y., Zhao, D., & Liu, L., 2021, Hesperidin prevents hyperglycemia in diabetic rats by activating the insulin receptor pathway. Experimental and Therapeutic Medicine, 21(1), 53. https://doi.org/10.3892/etm.2020.9485.
[21] Pari, L., & Rajarajeswari, N., 2009, Efficacy of coumarin on hepatic key enzymes of glucose metabolism in chemical induced type 2 diabetic rats. Chemico-biological Interactions, 181(3), 292–296. https://doi.org/10.1016/j.cbi.2009.07.018.
[22] Gothandam, K., Ganesan, V. S., Ayyasamy, T., & Ramalingam, S., 2019, Antioxidant potential of theaflavin ameliorates the activities of key enzymes of glucose metabolism in high fat diet and streptozotocin - induced diabetic rats. Redox report : Communications in free radical research, 24(1), 41–50. https://doi.org/10.1080/13510002.2019.1624085.
[23] Boucher, J., Kleinridders, A., & Kahn, C. R., 2014, Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harbor Perspectives in Biology, 6(1), a009191. https://doi.org/10.1101/cshperspect.a009191.
[24] Zhang Y, Xu W, Huang X et al., 2018, Fucoxanthin ameliorates hyperglycemia, hyperlipidemia and insulin resistance in diabetic mice partially through IRS-1/PI3K/Akt and AMPK pathways. Journal of Functional Foods, 48:515–524. 10.1016/j.jff.2018.07.048.
[25] Cai, S., Sun, W., Fan, Y., Guo, X., Xu, G., Xu, T., Hou, Y., Zhao, B., Feng, X., & Liu, T., 2016, Effect of mulberry leaf (Folium Mori) on insulin resistance via IRS-1/PI3K/Glut-4 signalling pathway in type 2 diabetes mellitus rats. Pharmaceutical Biology, 54(11), 2685–2691. https://doi.org/10.1080/13880209.2016.1178779.
[26] Wang, Y., Nishina, P. M., & Naggert, J. K., 2009, Degradation of IRS1 leads to impaired glucose uptake in adipose tissue of the type 2 diabetes mouse model TALLYHO/Jng. The Journal of Endocrinology, 203(1), 65–74. https://doi.org/10.1677/JOE-09-0026.
[27] Li, H., Yu, L., & Zhao, C., 2019, Dioscin attenuates high‑fat diet‑induced insulin resistance of adipose tissue through the IRS‑1/PI3K/Akt signaling pathway. Molecular Medicine Reports, 19(2), 1230–1237. https://doi.org/10.3892/mmr.2018.9700.
[28] Ramalingam, K., Yadalam, P. K., Ramani, P., Krishna, M., Hafedh, S., Badnjević, A., Cervino, G., & Minervini, G., 2024, Light gradient boosting-based prediction of quality of life among oral cancer-treated patients. BMC Oral Health, 24(1), 349. https://doi.org/10.1186/s12903-024-04050-x.
[29] Neralla, M., M, H., Preethi, A., Selvakumar, S. C., & Sekar, D., 2024, Expression levels of microRNA-7110 in oral squamous cell carcinoma. Minerva Dental and Oral Science, 73(3), 155–160. https://doi.org/10.23736/S2724-6329.23.04801-5.
[30] Ramsundar, K., Jain, R. K., Balakrishnan, N., & Vikramsimha, B., 2023, Comparative evaluation of bracket bond failure rates of a novel non-primer adhesive with a conventional primer-based orthodontic adhesive - a pilot study. Journal of Dental Research, Dental Clinics, Dental Prospects, 17(1), 35–39. https://doi.org/10.34172/joddd.2023.36953.
Viewed PDF 137 17