1
|
T FX, R S, A K FR, S B, R K, M A, S V, S P, S A, K S, M T. Phytochemical composition, anti-microbial, anti-oxidant and anti-diabetic effects of Solanum elaeagnifolium Cav. leaves: in vitro and in silico assessments. J Biomol Struct Dyn 2025; 43:3688-3714. [PMID: 38180058 DOI: 10.1080/07391102.2023.2300124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
The aim of this study was to screen the chemical components of Solanum elaeagnifolium leaves and assess their therapeutic attributes with regard to their antioxidant, antibacterial, and antidiabetic activities. The antidiabetic effects were explored to determine the α-amylase and α-glucosidase inhibitory potential of the leaf extract. To identify the active antidiabetic drugs from the extracts, the GC-MS-screened molecules were docked with diabetes-related proteins using the glide module in the Schrodinger Tool. In addition, molecular dynamics (MD) simulations were performed for 100 ns to evaluate the binding stability of the docked complex using the Desmond module. The ethyl acetate had a significant total phenolic content (TPC), with a value of 79.04 ± 0.98 mg/g GAE. The ethanol extract was tested for its minimum inhibitory concentration (MIC) for its bacteriostatic properties. It suppressed the growth of B. subtilis, E. coli, P. vulgaris, R. equi and S. epidermis at a dosage of 118.75 µg/mL. Moreover, the IC50 values of the ethanol extract were determined to be 17.78 ± 2.38 in the α-amylase and and 27.90 ± 5.02 µg/mL in α-glucosidase. The in-silico investigation revealed that cyclolaudenol achieved docking scores of -7.94 kcal/mol for α-amylase. Likewise, the α-tocopherol achieved the docking scores of -7.41 kcal/mol for glycogen phosphorylase B and -7.21 kcal/mol for phosphorylase kinase. In the MD simulations, the cyclolaudenol and α-tocopherol complexes exhibited consistently stable affinities with diabetic proteins throughout the trajectory. Based on these findings, we conclude that this plant could be a good source for the development of novel antioxidant, antibacterial, and antidiabetic agents.
Collapse
Affiliation(s)
- Francis Xavier T
- Ethnopharmacological Research Unit, PG and Research Department of Botany, St. Joseph's College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Sabitha R
- Ethnopharmacological Research Unit, PG and Research Department of Botany, St. Joseph's College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Freeda Rose A K
- PG and Research Department of Botany, Holy Cross College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Balavivekananthan S
- Ethnopharmacological Research Unit, PG and Research Department of Botany, St. Joseph's College (Autonomous), Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Kariyat R
- Department of Biology, The University of Texas, Rio Grande Valley, W University Dr, Edinburg, TX, USA
| | - Ayyanar M
- PG and Research Department of Botany, A.V.V.M. Sri Pushpam College (Autonomous), Bharathidasan University, Poondi, Tamil Nadu, India
| | - Vijayakumar S
- PG and Research Department of Botany, A.V.V.M. Sri Pushpam College (Autonomous), Bharathidasan University, Poondi, Tamil Nadu, India
| | - Prabhu S
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences, Cochin, Kerala, India
| | - Amalraj S
- Division of Phytochemistry and Drug Design, Department of Biosciences, Rajagiri College of Social Sciences, Cochin, Kerala, India
| | - Shine K
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Thiruvengadam M
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Korea
| |
Collapse
|
2
|
Choudhury AA, V DR. Computational analysis of potential drug-like compounds from Solanum torvum - A promising phytotherapeutics approach for the treatment of diabetes. J Biomol Struct Dyn 2025; 43:2073-2091. [PMID: 38116744 DOI: 10.1080/07391102.2023.2293279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
Diabetes mellitus (DM) is a global pandemic that is characterized by high blood glucose levels. Conventional treatments have limitations, leading to the search for natural alternatives. This study focused on Solanum torvum (STV), a medicinal plant, to identify potential anti-diabetic compounds using molecular docking and molecular dynamics simulations. We focused on identifying natural inhibitors of two key enzymes involved in glucose metabolism: α-amylase (1HNY) and α-glucosidase (4J5T). In our preliminary docking study, rutin showed the highest binding affinity (-11.58 kcal/mol) to α-amylase, followed by chlorogenin (-7.58 kcal/mol) and myricetin (-5.82 kcal/mol). For α-glucosidase, rutin had the highest binding affinity (-11.78 kcal/mol), followed by chlorogenin (-7.11 kcal/mol) and fisetin (-6.44 kcal/mol). Hence, chlorogenin and rutin were selected for further analysis and compared with acarbose, an FDA-approved antidiabetic drug. Comparative docking revealed that chlorogenin had the highest binding affinity of (-9.9 kcal/mol) > rutin (-8.7 kcal/mol) and > acarbose (-7.7 kcal/mol) for α-amylase. While docking with α-glucosidase, chlorogenin again had the highest binding affinity of (-9.8 kcal/mol) > compared to rutin (-9.5 kcal/mol) and acarbose (-7.9 kcal/mol). Molecular dynamics (MD) simulations were conducted to assess their stability. We simulated 100 nanoseconds (ns) trajectories to analyze their stability on various parameters, including RMSD, RMSF, RG, SASA, H-bond analysis, PCA, FEL, and MM-PBSA on the six docked proteins. In conclusion, our study suggests that chlorogenin and rutin derived from STV may be effective natural therapeutic agents for diabetes management because of their strong binding affinities for the α-amylase and α-glucosidase enzymes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abbas Alam Choudhury
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Devi Rajeswari V
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
3
|
Das B, Somkuwar BG, Chaudhary SK, Kharlyngdoh E, Pakyntein CL, Basor K, Shukla JK, Bhardwaj PK, Mukherjee PK. Therapeutics of bitter plants from Northeast region of India and their pharmacological and phytochemical perspectives. Pharmacol Res 2025; 212:107626. [PMID: 39875018 DOI: 10.1016/j.phrs.2025.107626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Natural resources have been used for food and medicine since the beginning of human civilization, and they have always been a low-cost, easily accessible source for individuals. Northeast region of India (NER) represents a significant portion of India's flora and fauna. Marginality, fragility, inaccessibility, ethnicity, and cultural diversity thrived in the region, resulting in the richest reservoir of genetic variation of bioresources. Several bitter plants are used by the locals as both food and medicine to treat a variety of diseases. These medicinal plants are an excellent source of chemically diverse biologically active phytometabolites. There have been few efforts to raise awareness about health benefits of bitter plant resources abound in this region that may provides opportunities for their sustainable utilization. Understanding the structural features of plant derived bitterants in relationship with specific bitter receptor will provide research prospects to identify biomolecules with health benefits. In this context the present review is intended to deliver phyto-pharmacological aspects of bitter plant resources of NER together with detailed understanding of possible association between plant derived phytometabolites as bitter agonists with extraoral bitter receptors.
Collapse
Affiliation(s)
- Bhaskar Das
- BRIC-Institute of Bioresources and Sustainable Development (BRIC-IBSD), Department of Biotechnology, Government of India, Imphal, Manipur795001, India.
| | | | | | | | | | - Kishor Basor
- BRIC-IBSD, Meghalaya Center, Shillong, Meghalaya 793009, India.
| | | | | | - Pulok Kumar Mukherjee
- BRIC-Institute of Bioresources and Sustainable Development (BRIC-IBSD), Department of Biotechnology, Government of India, Imphal, Manipur795001, India; BRIC-IBSD, Mizoram Center, Aizawl, Mizoram 796005, India; BRIC-IBSD, Meghalaya Center, Shillong, Meghalaya 793009, India.
| |
Collapse
|
4
|
Lai HC, Weng JC, Huang HC, Ho JX, Kuo CL, Cheng JC, Huang ST. Solanum torvum induces ferroptosis to suppress hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118670. [PMID: 39117020 DOI: 10.1016/j.jep.2024.118670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Solanum torvum Sw. (ST) is used to clear heat toxins, promote blood circulation, and alleviate blood stasis. Therefore, this plant has traditionally been used as an ethnomedicine for common cold, chronic gastritis, and tumors. AIM OF THE STUDY This study aimed to elucidate the mechanism by which ST induces ferroptosis in hepatocellular carcinoma (HCC), the combination effect with lenvatinib, and the impact on lenvatinib-resistant cells. MATERIALS AND METHODS Cell viability assays were performed using different hepatoma cell lines treated with ST. Lipid peroxidation and iron assays were performed using flow cytometry. Molecules involved in the ferroptosis pathway were detected by Western blotting. Finally, a lenvatinib-resistant cell line was established to evaluate the antiproliferative effects of ST. RESULTS ST ethanol extract inhibited the growth of various hepatoma cell lines. A significant reduction in glutathione peroxidase 4 (GPX4) expression was observed following ST treatment, which was accompanied by increased lipid peroxidation and Fe2+ accumulation. ST induced ferroptosis mainly through heme oxygenase-1 (HO-1) expression. HO-1 knockdown reduced ST-induced lipid peroxidation and reversed GPX4 suppression. Acyl-CoA synthetase long-chain family member 4 (ACSL4) also participated in ST-induced ferroptosis. ST and lenvatinib combination showed an additive effect, and ST retained its potential anti-HCC efficacy in a lenvatinib-resistant cell line. CONCLUSION This study demonstrated that the ethanol extract of ST inhibits hepatoma cell growth by inducing ferroptosis. ST displayed an additive effect with lenvatinib in Hep 3B cells and showed remarkable anti-HCC activity in lenvatinib-resistant Hep 3B cells. Collectively, the study shows that ST might have the potential to reduce lenvatinib use in clinical practice and salvage cases of lenvatinib resistance.
Collapse
Affiliation(s)
- Hsiang-Chun Lai
- Graduate Institute of Chinese Medicine, School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jui-Chun Weng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Hui-Chi Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jin-Xuan Ho
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| | - Sheng-Teng Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan; Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
5
|
Mukundh ST, Natarajan SR, Veeraraghavan VP, Jayaraman S. Study on Antidiabetic Potential of Sessuvium Portulacastrum Aqueous Extract: An In-Silico and In-Vitro Analysis. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1291-S1294. [PMID: 38882866 PMCID: PMC11174331 DOI: 10.4103/jpbs.jpbs_589_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/10/2023] [Accepted: 11/17/2023] [Indexed: 06/18/2024] Open
Abstract
Diabetes mellitus is a persistent metabolic condition marked by elevated blood glucose levels due to compromised insulin secretion or functionality. The search for natural antidiabetic agents has gained attention due to their potential effectiveness and safety profiles. Sessuvium portulacastrum, a coastal plant, has been traditionally used for various medicinal purposes. This study investigates the antidiabetic potential of Sessuvium portulacastrum aqueous extract by analyzing its inhibitory effects on key enzymes involved in carbohydrate metabolism and exploring its molecular interactions with critical target proteins. The aqueous extract of Sessuvium portulacastrum was prepared and used for in vitro analysis. The reduced activity of the extract against α-amylase and α-glucosidase enzymes, crucial in glucose absorption and postprandial hyperglycemia, was assessed. Molecular docking techniques were employed to explore the potential interactions between active compounds in the extract and diabetes-related proteins, including BAX, GSK3β, and CADH. The study revealed significant inhibition of both alpha-amylase and alpha-glucosidase enzymes by Sessuvium portulacastrum aqueous extract, indicating its potential to reduce glucose absorption and postprandial hyperglycemia. Moreover, the molecular docking analysis demonstrated strong binding interactions between active compounds in the extract and key proteins involved in diabetes-related pathways, namely apoptotic pathways, glycogen synthesis, and cell adhesion. The findings of this study highlight the promising antidiabetic potential of Sessuvium portulacastrum aqueous extract. Upcoming research should get an attention on isolating and characterizing the active compounds responsible for these effects on antidiabetic therapies from natural sources.
Collapse
Affiliation(s)
- S Tarun Mukundh
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Sathan R Natarajan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
6
|
Nithin Krishna K, Krishnamoorthy K, Veeraraghavan VP, Jayaraman S. Development of Anti-Inflammatory Drug from Crataeva Nurvala: In Silico and In Vitro Approach. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1308-S1311. [PMID: 38882780 PMCID: PMC11174216 DOI: 10.4103/jpbs.jpbs_594_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 06/18/2024] Open
Abstract
Background Crataeva nurvala, a medicinal plant with potential therapeutic properties, offers a promising avenue for the development of novel anti-inflammatory drugs. This study adopted a combined in silico and in vitro approach to investigate the anti-inflammatory potential of compounds derived from Crataeva nurvala. Materials and Methods In the in silico phase, virtual screening and molecular docking analyses were conducted to identify bioactive compounds from Crataeva nurvala that could interact with key inflammatory targets. Subsequently, selected compounds were synthesized and subjected to in vitro experimentation. Cellular models were employed to assess the anti-inflammatory effects of Crataeva nurvala-derived compounds, focusing on the modulation of pro-inflammatory cytokine levels and the underlying signaling pathways. Results Virtual screening and molecular docking led to the identification of several bioactive compounds with favorable interactions with inflammatory targets. In the in vitro experiments, treatment with Crataeva nurvala-derived compounds resulted in a significant reduction in pro-inflammatory cytokine production. Moreover, the compounds exhibited the ability to modulate inflammatory signaling pathways, further substantiating their anti-inflammatory potential. Conclusions This study not only contributes to the development of effective anti-inflammatory drugs but also underscores the value of harnessing natural sources such as Crataeva nurvala for therapeutic interventions in inflammatory disorders. The dual-phase strategy presented here provides a robust framework for anti-inflammatory drug discovery and validation.
Collapse
Affiliation(s)
- K Nithin Krishna
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Kalaiselvi Krishnamoorthy
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|