Phytochemical profiling and in silico evaluation of Brassica oleracea stem bioactives as novel aldose reductase inhibitors for diabetic neuropathy treatment

Authors

  • Rajashree Dadasaheb Ghogare Pravara Rural College of Pharmacy, Loni, Tal - Rahata, Dist – Ahilyanagar, 413736, India https://orcid.org/0000-0002-7364-4929
  • Rahul Rajendra Kunkulol Pravara Institute of Medical Science, Loni, Tal - Rahata, Dist – Ahilyanagar, 413736, India
  • Deepak Babasaheb Nehe Padmashri Vikhe Patil College of Arts science & commerce, Pravaranagar, Tal - Rahata, Dist – Ahilyanagar, 413712, India

DOI:

https://doi.org/10.69857/joapr.v13i5.1450

Keywords:

Brassica oleracea, aldose reductase inhibitor, diabetic neuropathy, molecular docking, ADME analysis, drug-likeness

Abstract

Background: Diabetic neuropathy affects over 50% of diabetic patients, causing significant morbidity and economic burden exceeding $327 billion annually. Current treatments offer limited relief, accompanied by considerable side effects. Aldose reductase inhibition represents a promising therapeutic approach, yet synthetic inhibitors face clinical challenges, including hepatotoxicity and inadequate safety margins. This study investigates Brassica oleracea stem bioactives as novel aldose reductase inhibitors. Methodology: Sequential extraction of Brassica oleracea stems employed ethanol and acetone solvents. Phytochemical screening utilized standard chemical tests, while HR-LCMS enabled metabolite identification. Molecular docking against human aldose reductase (PDB: 1US0) was performed using GeinDock Suite. Drug-likeness and ADME properties were assessed using SwissADME, in accordance with Lipinski's Rule of Five. Comprehensive pharmacokinetic parameters were also evaluated. Results and Discussion: HR-LCMS identified 33 bioactive compounds with identification scores >90%. Four lead compounds demonstrated optimal aldose reductase inhibitory potential with superior drug-likeness: Indole-3-acetonitrile (-8.9 kcal/mol, Ki=0.30 μM) approached reference inhibitors epalrestat (-9.9 kcal/mol) and sorbinil (-9.4 kcal/mol) with zero Lipinski violations. 18-Oxooleate (-7.5 kcal/mol) and Salicylamide (-7.6 kcal/mol) exhibited exceptional bioavailability (0.85) with minimal CYP450 inhibition. Phytosphingosine (-6.7 kcal/mol) displayed advantageous peripheral selectivity. Conclusion: Four B. oleracea compounds demonstrate optimal convergence of aldose reductase inhibitory potential and pharmaceutical feasibility, offering promising orally bioavailable candidates for diabetic neuropathy management. Their natural origin and favorable ADME profiles warrant immediate progression to in vitro validation and in vivo studies.

Downloads

Download data is not yet available.

References

Pop-Busui R, Ang L, Boulton AJM, Feldman EL, Marcus RL, Mizokami-Stout K, et al. Diagnosis and Treatment of Painful Diabetic Peripheral Neuropathy. ADA Clinical Compendia, 2022(1), 1-32 (2022) https://doi.org/10.2337/db2022-01.

Quiroz-Aldave J, Durand-Vásquez M, Gamarra-Osorio E, Suarez-Rojas J, Jantine Roseboom P, Alcalá-Mendoza R, et al. Diabetic neuropathy: Past, present, and future. Caspian J Intern Med, 14(2), 153-169 (2023) https://doi.org/10.22088/cjim.14.2.153.

Elafros MA, Andersen H, Bennett DL, Savelieff MG, Viswanathan V, Callaghan BC, et al. Towards prevention of diabetic peripheral neuropathy: clinical presentation, pathogenesis, and new treatments. The Lancet Neurology, 21(10), 922-936 (2022) https://doi.org/10.1016/S1474-4422(22)00188-0.

Carmichael J, Fadavi H, Ishibashi F, Shore AC, Tavakoli M. Advances in Screening, Early Diagnosis and Accurate Staging of Diabetic Neuropathy. Front Endocrinol, 12, 671257 (2021) https://doi.org/10.3389/fendo.2021.671257.

Ismail CAN. Issues and challenges in diabetic neuropathy management: A narrative review. World J Diabetes, 14(6), 741-757 (2023) https://doi.org/10.4239/wjd.v14.i6.741.

Thakur S, Gupta SK, Ali V, Singh P, Verma M. Aldose Reductase: a cause and a potential target for the treatment of diabetic complications. Arch Pharm Res, 44(7), 655-667 (2021) https://doi.org/10.1007/s12272-021-01343-5.

Gupta JK. The Role of Aldose Reductase in Polyol Pathway: An Emerging Pharmacological Target in Diabetic Complications and Associated Morbidities. Current Pharmaceutical Biotechnology, 25(9), 1073-1081 (2024) https://doi.org/10.2174/1389201025666230830125147.

Singh M, Kapoor A, Bhatnagar A. Physiological and Pathological Roles of Aldose Reductase. Metabolites, 11(10), 655 (2021) https://doi.org/10.3390/metabo11100655.

Bernardoni BL, D'Agostino I, Scianò F, La Motta C. The challenging inhibition of Aldose Reductase for the treatment of diabetic complications: a 2019-2023 update of the patent literature. Expert Opinion on Therapeutic Patents, 34(11), 1085-1103 (2024) https://doi.org/10.1080/13543776.2024.2412573.

Pineschi C. Identification and Characterization of Human Aldose Reductase Differential Inhibitors. Doctoral Dissertation, University of Florence (2021) https://doi.org/10.25434/pineschi-carlotta_phd2021.

Najmi A, Javed SA, Al Bratty M, Alhazmi HA. Modern Approaches in the Discovery and Development of Plant-Based Natural Products and Their Analogues as Potential Therapeutic Agents. Molecules, 27(2), 349 (2022) https://doi.org/10.3390/molecules27020349.

Chaachouay N, Zidane L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. Drugs and Drug Candidates, 3(1), 184-207 (2024) https://doi.org/10.3390/ddc3010011.

Nasim N, Sandeep IS, Mohanty S. Plant-derived natural products for drug discovery: current approaches and prospects. Nucleus, 65(3), 399-411 (2022) https://doi.org/10.1007/s13237-022-00405-3.

Aware CB, Patil DN, Suryawanshi SS, Mali PR, Rane MR, Gurav RG, et al. Natural bioactive products as promising therapeutics: A review of natural product-based drug development. South African Journal of Botany, 151, 512-528 (2022) https://doi.org/10.1016/j.sajb.2022.05.028.

Thomas E, Stewart LE, Darley BA, Pham AM, Esteban I, Panda SS. Plant-Based Natural Products and Extracts: Potential Source to Develop New Antiviral Drug Candidates. Molecules, 26(20), 6197 (2021) https://doi.org/10.3390/molecules26206197.

Torres-Rodriguez A, Darvishzadeh R, Skidmore AK, Fränzel-Luiten E, Knaken B, Schuur B. High-throughput Soxhlet extraction method applied for analysis of leaf lignocellulose and non-structural substances. MethodsX, 12, 102644 (2024) https://doi.org/10.1016/j.mex.2024.102644.

Trolles-Cavalcante SY, Dutta A, Sofer Z, Borenstein A. The effectiveness of Soxhlet extraction as a simple method for GO rinsing as a precursor of high-quality graphene. Nanoscale Advances, 3(18), 5292-5300 (2021) https://doi.org/10.1039/D1NA00382H.

Farooq S, Shaheen G, Asif HM, Aslam MR, Zahid R, Rajpoot SR, et al. Preliminary Phytochemical Analysis: In-Vitro Comparative Evaluation of Anti-arthritic and Anti-inflammatory Potential of Some Traditionally Used Medicinal Plants. Dose-Response, 20(1), 15593258211069720 (2022) https://doi.org/10.1177/15593258211069720.

Kowalczyk T, Merecz-Sadowska A, Rijo P, Isca VMS, Picot L, Wielanek M, et al. Preliminary Phytochemical Analysis and Evaluation of the Biological Activity of Leonotis nepetifolia (L.) R. Br Transformed Roots Extracts Obtained through Rhizobium rhizogenes-Mediated Transformation. Cells, 10(5), 1242 (2021) https://doi.org/10.3390/cells10051242.

Vignesh A, Selvakumar S, Vasanth K. Comparative LC-MS analysis of bioactive compounds, antioxidants and antibacterial activity from leaf and callus extracts of Saraca asoca. Phytomedicine Plus, 2(1), 100167 (2022) https://doi.org/10.1016/j.phyplu.2021.100167.

Al-Dalahmeh Y, Al-Bataineh N, Al-Balawi SS, Lahham JN, Al-Momani IF, Al-Sheraideh MS, et al. LC-MS/MS Screening, Total Phenolic, Flavonoid and Antioxidant Contents of Crude Extracts from Three Asclepiadaceae Species Growing in Jordan. Molecules, 27(3), 859 (2022) https://doi.org/10.3390/molecules27030859.

Eisenberg SM, Knizner KT, Muddiman DC. Metabolite Annotation Confidence Score (MACS): A Novel MSI Identification Scoring Tool. J Am Soc Mass Spectrom, 34(10), 2222-2231 (2023) https://doi.org/10.1021/jasms.3c00178.

Silva RR da, Wang M, Nothias LF, Hooft JJJ van der, Caraballo-Rodríguez AM, Fox E, et al. Propagating annotations of molecular networks using in silico fragmentation. PLOS Computational Biology, 14(4), e1006089 (2018) https://doi.org/10.1371/journal.pcbi.1006089.

Umi Baroroh SS, Muscifa ZS, Destiarani W, Rohmatullah FG, Yusuf M. Molecular interaction analysis and visualization of protein-ligand docking using Biovia Discovery Studio Visualizer. Indonesian Journal of Computational Biology, 2(1), 22-30 (2023) https://doi.org/10.24198/ijcb.v2i1.46322.

Ayodele PF, Bamigbade A, Bamigbade OO, Adeniyi IA, Tachin ES, Seweje AJ, et al. Illustrated Procedure to Perform Molecular Docking Using PyRx and Biovia Discovery Studio Visualizer: A Case Study of 10kt With Atropine. Progress in Drug Discovery & Biomedical Science, 6, a0000424 (2023) https://doi.org/10.36877/pddbs.a0000424.

Sharma S, Sharma A, Gupta U. Molecular Docking studies on the Anti-fungal activity of Allium sativum (Garlic) against Mucormycosis (black fungus) by BIOVIA discovery studio visualizer 21.1.0.0. Annals of Antivirals and Antiretrovirals, 5(1), 028-032 (2021) https://doi.org/10.17352/aaa.000013.

Imran A, Shehzad MT, Shah SJA, Laws M, al-Adhami T, Rahman KM, et al. Development, Molecular Docking, and In Silico ADME Evaluation of Selective ALR2 Inhibitors for the Treatment of Diabetic Complications via Suppression of the Polyol Pathway. ACS Omega, 7(30), 26425-26436 (2022) https://doi.org/10.1021/acsomega.2c02326.

Jin R, Wang J, Li M, Tang T, Feng Y, Zhou S, et al. Discovery of a Novel Benzothiadiazine-Based Selective Aldose Reductase Inhibitor as Potential Therapy for Diabetic Peripheral Neuropathy. Diabetes, 73(4), 497-510 (2023) https://doi.org/10.2337/db23-0006.

Howard EI, Sanishvili R, Cachau RE, Mitschler A, Chevrier B, Barth P, et al. Ultrahigh resolution drug design I: Details of interactions in human aldose reductase-inhibitor complex at 0.66 Å. Proteins, 55(4), 792-804 (2004) https://doi.org/10.1002/prot.20015.

El-Kabbani O, Darmanin C, Schneider TR, Hazemann I, Ruiz F, Oka M, et al. Ultrahigh resolution drug design. II. Atomic resolution structures of human aldose reductase holoenzyme complexed with fidarestat and minalrestat: Implications for the binding of cyclic imide inhibitors. Proteins, 55(4), 805-813 (2004) https://doi.org/10.1002/prot.20001.

Urzhumtsev A, Tête-Favier F, Mitschler A, Barbanton J, Barth P, Urzhumtseva L, et al. A 'specificity' pocket inferred from the crystal structures of the complexes of aldose reductase with the pharmaceutically important inhibitors tolrestat and sorbinil. Structure, 5(5), 601-612 (1997) https://doi.org/10.1016/S0969-2126(97)00216-5.

Hao X, Wang X, Jiang Q, Zhu Y, Zhu Y, Chen M, et al. Novel Multifunctional Aldose Reductase Inhibitors Based on Quinoxalin-2(1H)-one Scaffold for Treatment of Diabetic Complications: Design, Synthesis, and Biological Evaluation. Chemistry & Biodiversity, 22(1), e202402358 (2025) https://doi.org/10.1002/cbdv.202402358.

Che X, Liu Q, Zhang L. An accurate and universal protein-small molecule batch docking solution using Autodock Vina. Results in Engineering, 19, 101335 (2023) https://doi.org/10.1016/j.rineng.2023.101335.

Eberhardt J, Santos-Martins D, Tillack AF, Forli S. AutoDock Vina 1.2.0: New Docking Methods, Expanded Force Field, and Python Bindings. J Chem Inf Model, 61(8), 3891-3898 (2021) https://doi.org/10.1021/acs.jcim.1c00203.

Ding J, Tang S, Mei Z, Wang L, Huang Q, Hu H, et al. Vina-GPU 2.0: Further Accelerating AutoDock Vina and Its Derivatives with Graphics Processing Units. J Chem Inf Model, 63(7), 1982-1998 (2023) https://doi.org/10.1021/acs.jcim.2c01504.

Sarkar A, Concilio S, Sessa L, Marrafino F, Piotto S. Advancements and novel approaches in modified AutoDock Vina algorithms for enhanced molecular docking. Results in Chemistry, 7, 101319 (2024) https://doi.org/10.1016/j.rechem.2024.101319.

Tang S, Chen R, Lin M, Lin Q, Zhu Y, Ding J, et al. Accelerating AutoDock Vina with GPUs. Molecules, 27(9), 3041 (2022) https://doi.org/10.3390/molecules27093041.

Pham TNH, Nguyen TH, Tam NM, Vu TY, Pham NT, Huy NT, et al. Improving ligand-ranking of AutoDock Vina by changing the empirical parameters. Journal of Computational Chemistry, 43(3), 160-169 (2022) https://doi.org/10.1002/jcc.26779.

Ramírez D, Caballero J. Is It Reliable to Take the Molecular Docking Top Scoring Position as the Best Solution without Considering Available Structural Data? Molecules, 23(5), 1038 (2018) https://doi.org/10.3390/molecules23051038.

Pinzi L, Rastelli G. Molecular Docking: Shifting Paradigms in Drug Discovery. IJMS, 20(18), 4331 (2019) https://doi.org/10.3390/ijms20184331.

Ramirez MA, Borja NL. Epalrestat: An Aldose Reductase Inhibitor for the Treatment of Diabetic Neuropathy. Pharmacotherapy, 28(5), 646-655 (2008) https://doi.org/10.1592/phco.28.5.646.

Stefek M, Soltesova Prnova M, Majekova M, Rechlin C, Heine A, Klebe G. Identification of Novel Aldose Reductase Inhibitors Based on Carboxymethylated Mercaptotriazinoindole Scaffold. J Med Chem, 58(6), 2649-2657 (2015) https://doi.org/10.1021/jm5015814.

Sardar H. Drug like potential of Daidzein using SwissADME prediction: In silico Approaches. PHYTONutrients, 1, 02-08 (2023) https://doi.org/10.62368/pn.vi.18.

Mvondo JGM, Matondo A, Mawete DT, Bambi SMN, Mbala BM, Lohohola PO. In Silico ADME/T Properties of Quinine Derivatives using SwissADME and pkCSM Webservers. IJTDH, 42(11), 1-12 (2021) https://doi.org/10.9734/ijtdh/2021/v42i1130492.

Khare S, Chatterjee T, Gupta S, Ashish P. Bioavailability predictions, pharmacokinetics and drug-likeness of bioactive compounds from Andrographis paniculata using Swiss ADME. MGM Journal of Medical Sciences, 10(4), 651 (2023) https://doi.org/10.4103/mgmj.mgmj_245_23.

Sathasivam R, Park SU, Kim JK, Park YJ, Kim MC, Nguyen BV, et al. Metabolic Profiling of Primary and Secondary Metabolites in Kohlrabi (Brassica oleracea var. gongylodes) Sprouts Exposed to Different Light-Emitting Diodes. Plants, 12(6), 1296 (2023) https://doi.org/10.3390/plants12061296.

Gudiño I, Martín A, Casquete R, Prieto MH, Ayuso MC, Córdoba MG. Evaluation of broccoli (Brassica oleracea var. italica) crop by-products as sources of bioactive compounds. Scientia Horticulturae, 304, 111284 (2022) https://doi.org/10.1016/j.scienta.2022.111284.

Mattosinhos PDS, Sarandy MM, Novaes RD, Esposito D, Gonçalves RV. Anti-Inflammatory, Antioxidant, and Skin Regenerative Potential of Secondary Metabolites from Plants of the Brassicaceae Family: A Systematic Review of In Vitro and In Vivo Preclinical Evidence. Antioxidants, 11(7), 1346 (2022) https://doi.org/10.3390/antiox11071346.

Da Silva RR, Wang M, Nothias LF, Van Der Hooft JJJ, Caraballo-Rodríguez AM, Fox E, et al. Propagating annotations of molecular networks using in silico fragmentation. PLoS Comput Biol, 14(4), e1006089 (2018) https://doi.org/10.1371/journal.pcbi.1006089.

Al-Dalahmeh Y, Al-Bataineh N, Al-Balawi SS, Lahham JN, Al-Momani IF, Al-Sheraideh MS, et al. LC-MS/MS Screening, Total Phenolic, Flavonoid and Antioxidant Contents of Crude Extracts from Three Asclepiadaceae Species Growing in Jordan. Molecules, 27(3), 859 (2022) https://doi.org/10.3390/molecules27030859.

Aware CB, Patil DN, Suryawanshi SS, Mali PR, Rane MR, Gurav RG, et al. Natural bioactive products as promising therapeutics: A review of natural product-based drug development. South African Journal of Botany, 151, 512-528 (2022) https://doi.org/10.1016/j.sajb.2022.05.028.

Najmi A, Javed SA, Al Bratty M, Alhazmi HA. Modern Approaches in the Discovery and Development of Plant-Based Natural Products and Their Analogues as Potential Therapeutic Agents. Molecules, 27(2), 349 (2022) https://doi.org/10.3390/molecules27020349.

Chaachouay N, Zidane L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. DDC, 3(1), 184-207 (2024) https://doi.org/10.3390/ddc3010011.

Nasim N, Sandeep IS, Mohanty S. Plant-derived natural products for drug discovery: current approaches and prospects. Nucleus, 65(3), 399-411 (2022) https://doi.org/10.1007/s13237-022-00405-3.

Thomas E, Stewart LE, Darley BA, Pham AM, Esteban I, Panda SS. Plant-Based Natural Products and Extracts: Potential Source to Develop New Antiviral Drug Candidates. Molecules, 26(20), 6197 (2021) https://doi.org/10.3390/molecules26206197.

Kumari P, Kohal R, Bhavana, Gupta GD, Verma SK. Selectivity challenges for aldose reductase inhibitors: A review on comparative SAR and interaction studies. Journal of Molecular Structure, 1318, 139207 (2024) https://doi.org/10.1016/j.molstruc.2024.139207.

Jin R, Wang J, Li M, Tang T, Feng Y, Zhou S, et al. Discovery of a Novel Benzothiadiazine-Based Selective Aldose Reductase Inhibitor as Potential Therapy for Diabetic Peripheral Neuropathy. Diabetes, 73(4), 497-510 (2024) https://doi.org/10.2337/db23-0006.

Kashyap K, Mahapatra PP, Ahmed S, Buyukbingol E, Siddiqi MI. Identification of Potential Aldose Reductase Inhibitors Using Convolutional Neural Network-Based in Silico Screening. J Chem Inf Model, 63(19), 6261-6282 (2023) https://doi.org/10.1021/acs.jcim.3c00547.

Sonowal H, Ramana KV. Development of Aldose Reductase Inhibitors for the Treatment of Inflammatory Disorders and Cancer: Current Drug Design Strategies and Future Directions. CMC, 28(19), 3683-3712 (2021) https://doi.org/10.2174/0929867327666201027152737.

Bakal RL, Jawarkar RD, Manwar JV, Jaiswal MS, Ghosh A, Gandhi A, et al. Identification of potent aldose reductase inhibitors as antidiabetic (Anti-hyperglycemic) agents using QSAR based virtual Screening, molecular Docking, MD simulation and MMGBSA approaches. Saudi Pharmaceutical Journal, 30(5), 693-710 (2022) https://doi.org/10.1016/j.jsps.2022.04.003.

Hao X, Wang X, Jiang Q, Zhu Y, Zhu Y, Chen M, et al. Novel Multifunctional Aldose Reductase Inhibitors Based on Quinoxalin-2(1H)-one Scaffold for Treatment of Diabetic Complications: Design, Synthesis, and Biological Evaluation. Chemistry & Biodiversity, 22(1), e202402358 (2025) https://doi.org/10.1002/cbdv.202402358.

Published

2025-10-31

How to Cite

Ghogare, R. D., Kunkulol, R. R., & Nehe, D. B. (2025). Phytochemical profiling and in silico evaluation of Brassica oleracea stem bioactives as novel aldose reductase inhibitors for diabetic neuropathy treatment. Journal of Applied Pharmaceutical Research, 13(5), 254-270. https://doi.org/10.69857/joapr.v13i5.1450

Issue

Section

Articles