1
|
Vargas J, Arbelaez N, Cardenas D, Murillo J, Ospina V, Robledo S, Soto J. In vitro antitumor capacity of extracts obtained from the plants Plukenetia volubilis (Sacha inchi) and Moringa oleifera in gastric cancer. F1000Res 2025; 14:194. [PMID: 40443907 PMCID: PMC12120421 DOI: 10.12688/f1000research.158563.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Background Gastric cancer is the fifth most common cancer and the third leading cause of cancer deaths worldwide. Perioperative or adjuvant chemotherapy improves survival in patients with stage 1B or higher cancers. Moringa oleifera and Plukenetia volubilis (Sacha inchi) have been reported to enhance various biological functions, including antitumor and antiproliferative activity. Methods In order to evaluate this potential present in crude extracts of the leaves of these plants, as well as the seed oil of P.volubilis, the antitumor activity was determined according to the effect of these derivatives on different biological parameters such as cytotoxicity, proliferation, cell cycle, apoptosis (among others), in AGS cells (CRL-1739). Results All extracts tested were cytotoxic at 90 and 160 μg/ml concentrations . P. volubilis seed oil showed 95% mortality at 1% concentration (CC 50 = 46.7%). Cell proliferation was inhibited, and all extracts affected the cell cycle, but the P. volubilis oil significantly induced an accumulation of AGS cells in the sub G1 phase, inducing DNA fragmentation as a mechanism of cell death. The ethanolic M. oleifera leaf extract also inhibited cell migration. Conclusion M. oleifera, P. volubilis leaf extracts and P. volubilis seed oil can potentially be antitumor products. Further validation in a murine model of gastric cancer is needed to investigate the antitumor potential of these extracts further and to continue the development of herbal products that can help in the management of this type of tumor.
Collapse
Affiliation(s)
- Jose Vargas
- BIOGEN-Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación Masira, Cúcuta, Colombia
| | - Natalia Arbelaez
- PECET-Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Denny Cardenas
- BIOGEN-Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación Masira, Cúcuta, Colombia
| | - Javier Murillo
- Grupo Estudios Preclínicos, Corporación CIDEPRO, Medellín, Colombia
| | - Victoria Ospina
- Grupo Estudios Preclínicos, Corporación CIDEPRO, Medellín, Colombia
| | - Sara Robledo
- PECET-Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Javier Soto
- BIOGEN-Universidad de Santander, Facultad de Ciencias Médicas y de la Salud, Instituto de Investigación Masira, Cúcuta, Colombia
| |
Collapse
|
2
|
Haffez H, Sanad HH, Ebrahim H, Hassan ZA. Synergistic effects of abietic acid combined with doxorubicin on apoptosis induction in a human colorectal cancer cell line. Sci Rep 2025; 15:16102. [PMID: 40341222 PMCID: PMC12062260 DOI: 10.1038/s41598-025-99616-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
Cancer is a significant global disease with high mortality and limited therapeutic options. Chemotherapy is a cancer treatment option; however, there are still issues, including severe side effects, inadequate response, and drug resistance. Abietic acid is a natural diterpene with diverse pharmacological properties and can be used for cancer treatment. Therefore, this study aimed to assess the anticancer efficacy of abietic acid in combination with doxorubicin, a highly clinically used chemotherapeutic agent. Biochemical investigations include initial viability assays, combination therapy using isobologram analysis, apoptosis and cell cycle assays, gene expression assay, ELISA analysis of protein expression, DNA fragmentation, and wound healing assays. The data showed that doxorubicin-abietic acid (DOX-AB) is an effective and safe anticancer combination for Caco-2 cells. DOX-AB had a high safety index with minimal cytotoxicity at the combination dose on normal WI-38 fibroblasts cells. DOX-AB significantly decreased the proliferation and viability of Caco-2 cells, with an increase in the apoptosis rate in the late stage and necrosis with cell cycle arrest at the G2/M phase. Significant changes in the expression of modulators related to apoptosis, inflammation, and epigenetics were observed in gene and protein levels. DOX-AB combination had more efficient anticancer activity than doxorubicin alone. This study suggested that the use of abietic acid in combination with doxorubicin is a promising treatment for colorectal cancer because it enhances doxorubicin activity at relatively low doses with minimal cytotoxicity and overcomes multidrug resistance in tumors; these findings merit further investigation.
Collapse
Affiliation(s)
- Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
- Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, Cairo, 11795, Egypt.
| | - Hend H Sanad
- Health Affairs Directorate, Mansoura Health Administration, Mansura city, , El Dakahlia, Egypt
| | - Hassan Ebrahim
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, P.O. Box 11795, Cairo, Egypt
| | - Zeineb A Hassan
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
3
|
Venkatesan KB, Alamelu S, Srinivasan MK, Pachaiappan P. Nerolidol loaded beta cyclodextrin nanoparticles: a promising strategy for inducing apoptosis in breast cancer cells (MCF-7). JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-31. [PMID: 40326384 DOI: 10.1080/09205063.2025.2491605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
This study investigates the synthesis, characterization and anticancer efficacy of nerolidol-loaded beta cyclodextrin polymeric nanoparticles (NER-βCD-NPs) against MCF-7 breast cancer cells. Nerolidol, a sesquiterpene with anti-inflammatory, antioxidant, antimicrobial and anticancer properties, faces challenges of poor solubility and bioavailability, limiting its therapeutic potential. Breast cancer, a leading cause of cancer-related deaths in women, necessitates alternative therapies with fewer side effects compared to conventional chemotherapy. NER-βCD-NPs were synthesized and characterized using UV-visible spectroscopy, fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), dynamic light scattering (DLS), zeta potential analysis and differential scanning calorimetry (DSC). Drug encapsulation efficiency and in vitro release were analyzed using HPLC, while molecular docking assessed NER-βCD interactions. Characterization confirmed successful nanoparticle synthesis. UV-visible spectra and FTIR indicated encapsulation-specific changes, SEM revealed surface morphology, and DLS, zeta potential and DSC analyses demonstrated increased size and stability. The encapsulation efficiency was 84.9%, with 86% NER release at pH 5.4 over 48 h. Docking studies supported strong binding between NER and βCD (binding energy: -3.55 kcal/mol). Cytotoxicity assays showed significant MCF-7 cell inhibition. Mechanistic studies revealed reactive oxygen species (ROS) generation, mitochondrial dysfunction, nuclear changes and cell cycle arrest in the G0-G1 phase. Molecular analysis demonstrated apoptosis through upregulation of Bax, Caspase 6, Caspase 9 and Cytochrome c, alongside Bcl-2 downregulation. These results highlight NER-βCD-NPs as a promising strategy for breast cancer therapy, offering targeted delivery and enhanced therapeutic efficacy while mitigating nerolidol limitations. Further studies are warranted to validate their potential in clinical applications.
Collapse
Affiliation(s)
- Kamalesh Balakumar Venkatesan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Saravanan Alamelu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Manoj Kumar Srinivasan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Pugalendhi Pachaiappan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| |
Collapse
|
4
|
Choudhury SD, Ghosh S, Kumar P, Bhardwaj A, Singh K, Singh A, Kumar A, Basu B, Giri R, Choudhury D. Attenuation of c-Myc expression in breast cancer by hesperidin-mediated stabilization of its promoter proximal G quadruplex region. Int J Biol Macromol 2025; 309:143000. [PMID: 40222510 DOI: 10.1016/j.ijbiomac.2025.143000] [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: 06/05/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Hesperidin, a citrus flavanone, demonstrates significant potential as an anticancer agent by targeting the c-Myc G-quadruplex (G4) silencer element (Pu-27), a key epigenetic regulator of c-Myc expression. Molecular docking analysis revealed a strong interaction with Pu-27 (binding energy: -48.344 kcal/mol), forming hydrogen bonds across five critical regions. This interaction stabilized the G4 structure, as confirmed by increased ellipticity, higher melting temperature, and enhanced nanostructure formation. In functional assays, Hesperidin selectively inhibited the viability of MDA-MB-231 breast cancer cells while sparing normal cells. It significantly reduced clonogenic potential, migration, and c-Myc expression, indicating its role in suppressing oncogenic pathways. Moreover, Hesperidin effectively reduced primer dimer formation in the PCR stop assay and decreased mTFP expression in the mTFP reporter assay, further supporting its specificity for G4 stabilization. Preclinical studies demonstrated that Hesperidin treatment led to a marked reduction in tumor volume with minimal systemic toxicity, highlighting its therapeutic potential. These findings establish Hesperidin as a promising small-molecule stabilizer of the c-Myc G4 silencer, offering a targeted strategy for breast cancer therapy. By directly modulating c-Myc expression, hesperidin holds promise for clinical translation as a selective and effective anticancer agent.
Collapse
Affiliation(s)
- Satabdi Datta Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Sandip Ghosh
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology (IIT) Mandi, Mandi, Himachal Pradesh 175005, India
| | - Aparna Bhardwaj
- School of Basic Sciences, Indian Institute of Technology (IIT) Mandi, Mandi, Himachal Pradesh 175005, India
| | - Krishna Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh 453552, India
| | - Aakriti Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh 453552, India
| | - Amit Kumar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh 453552, India
| | - Biswarup Basu
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Rajnish Giri
- School of Basic Sciences, Indian Institute of Technology (IIT) Mandi, Mandi, Himachal Pradesh 175005, India
| | - Diptiman Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India; Centre for Excellence in Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India.
| |
Collapse
|
5
|
Almatroodi SA, Rahmani AH. Unlocking the Pharmacological Potential of Myricetin Against Various Pathogenesis. Int J Mol Sci 2025; 26:4188. [PMID: 40362425 PMCID: PMC12071824 DOI: 10.3390/ijms26094188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Myricetin is a natural flavonoid with powerful antioxidant and anti-inflammatory potential commonly found in vegetables, fruits, nuts, and tea. The vital role of this flavonoid in the prevention and treatment of various diseases is evidenced by its ability to reduce inflammation and oxidative stress, maintain tissue architecture, and modulate cell signaling pathways. Thus, this review summarizes recent evidence on myricetin, focusing precisely on its mechanisms of action in various pathogenesis, including obesity, diabetes mellitus, arthritis, osteoporosis, liver, neuro, cardio, and reproductive system-associated pathogenesis. Moreover, it has been revealed that myricetin exhibits anti-microbial properties due to obstructive virulence factors, preventing biofilm formation and disrupting membrane integrity. Additionally, synergistic potential with other drugs and the role of myricetin-based nanoformulations in different diseases are properly discussed. This review seeks to increase the understanding of myricetin's pharmacological potential in various diseases, principally highlighting its effective mechanisms of action. Further wide-ranging research, as well as more randomized and controlled clinical trial studies, should be executed to reconnoiter this compound's therapeutic value, safety, and usefulness against various human pathogenesis.
Collapse
Affiliation(s)
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
6
|
Zikpi KE, Motto AE, Atchou K, Kougblenou KRA, Lawson-Evi P, Eklu-Gadegbeku K. Evaluation of the protective effects of Spirulina platensis against cyclophosphamide-induced genotoxicity. Lab Anim Res 2025; 41:10. [PMID: 40264193 PMCID: PMC12013084 DOI: 10.1186/s42826-025-00242-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/10/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Damage to normal cells is the most common limitation of cancer chemotherapy. Cyclophosphamide, one of the most widely used anticancer drugs due to its cytotoxicity, can bind to deoxyribonucleic acid (DNA), causing chromosomal breaks, micronuclei, and cell death. The use of natural sources helps to prevent this damage, and Spirulina platensis is highly appreciated for its numerous bioactive compounds. This study aimed to investigate the antigenotoxic effects of Spirulina platensis powder (PoSP) on mouse bone marrow cells in vivo via a micronucleus assay. RESULTS Compared to the positive control, the administration of powder significantly reduced the PCE/PCE + NCE (polychromatic erythrocytes, normochromatic erythrocytes) ratio in treated mice. A significant increase in the percentage of MnPCE (micronucleus in polychromatic erythrocytes) in cyclophosphamide-treated bone marrow cells was observed. Compared with the positive controls, the groups treated with different doses in combination with cyclophosphamide presented a significant (p<0.0001) decrease in MnPCE in a dose-dependent manner. Compared to the positive control, PoSP significantly decreased MDA (malondialdehyde) levels in the livers of treated animals. The same things were observed in the kidneys and spleen. The catalase activity was also significantly increased in tissues, compared to negative control. CONCLUSIONS These findings suggest that PoSP does not cause DNA damage and can prevent genotoxicity, probably through its antioxidant activities.
Collapse
Affiliation(s)
- Kouamé Ephrem Zikpi
- Laboratory of Physiology/ Pharmacology, Research Unit of Pathophysiology, Bioactive Substances and Safety (PSBI), Faculty of Sciences, University of Lomé, Lomé, 01 BP1515, Togo.
| | - Aku Enam Motto
- Laboratory of Physiology/ Pharmacology, Research Unit of Pathophysiology, Bioactive Substances and Safety (PSBI), Faculty of Sciences, University of Lomé, Lomé, 01 BP1515, Togo
| | - Kokou Atchou
- Laboratory of Physiology/ Pharmacology, Research Unit of Pathophysiology, Bioactive Substances and Safety (PSBI), Faculty of Sciences, University of Lomé, Lomé, 01 BP1515, Togo
| | - Kounouho R Adounkpe Kougblenou
- Laboratory of Physiology/ Pharmacology, Research Unit of Pathophysiology, Bioactive Substances and Safety (PSBI), Faculty of Sciences, University of Lomé, Lomé, 01 BP1515, Togo
- Regional Institute for Development and Health (IREDESA), Ex CREDESA (Centre Régional pour le Développement et la Santé), Pahou, Cotonou, 01 BP1822, Benin
| | - Povi Lawson-Evi
- Laboratory of Physiology/ Pharmacology, Research Unit of Pathophysiology, Bioactive Substances and Safety (PSBI), Faculty of Sciences, University of Lomé, Lomé, 01 BP1515, Togo
| | - Kwashie Eklu-Gadegbeku
- Laboratory of Physiology/ Pharmacology, Research Unit of Pathophysiology, Bioactive Substances and Safety (PSBI), Faculty of Sciences, University of Lomé, Lomé, 01 BP1515, Togo
| |
Collapse
|
7
|
Sulaiman NF, Zulkifli SZ, Saaidin AS, Lekkala R, Izzaty Hassan N, Pungot NH. Exploring β-carboline hybrids and their derivatives: A review on synthesis and anticancer efficiency. Eur J Med Chem 2025; 288:117412. [PMID: 39987835 DOI: 10.1016/j.ejmech.2025.117412] [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: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Abstract
β-Carboline is a crucial compound in medicinal chemistry known for its versatile pharmacological activities. Recent research has focused on hybrid molecules incorporating a β-carboline scaffold linked to other pharmacophore moieties. These hybrid compounds have demonstrated diverse therapeutic properties, including anticancer, antianxiety, antimalarial, antidepressant, anti-inflammatory, antileishmanial, and antioxidant effects. This review highlights studies conducted from 2014 to the present with a particular emphasis on the development of β-carboline hybrid compounds and their derivatives as potent anticancer agents. The structure-activity relationship (SAR) analysis reveals that these hybrids exhibit significant cytotoxicity against various cancer cell lines. This review aims to inspire further research into the novel synthesis and evolution of β-carboline hybrids and their derivatives, potentially leading to new therapeutic advancements.
Collapse
Affiliation(s)
- Nur Fatihah Sulaiman
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia
| | - Siti Zafirah Zulkifli
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia; Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Aimi Suhaily Saaidin
- Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia
| | - Ravindar Lekkala
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bandar Baru Bangi, Selangor, 43600, Malaysia
| | - Nurul Izzaty Hassan
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bandar Baru Bangi, Selangor, 43600, Malaysia
| | - Noor Hidayah Pungot
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, 40450, Malaysia; Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA, Cawangan Selangor, Bandar Puncak Alam, Selangor, 42300, Malaysia.
| |
Collapse
|
8
|
Jia W, Cheng X. In Silico Discovery of a Novel Natural Product Targeting PI3Kα for the Treatment of Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2025; 26:3565. [PMID: 40332095 PMCID: PMC12027195 DOI: 10.3390/ijms26083565] [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: 02/19/2025] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a major health burden, with abnormal activation of phosphatidylinositol 3-kinase alpha (PI3Kα) strongly implicated in its pathogenesis. Targeting PI3Kα represents a promising therapeutic strategy. In this study, we employed structure-based virtual screening to identify natural small-molecule inhibitors of PI3Kα. A total of 12,800 molecules were screened, and five compounds were selected for further evaluation based on binding affinity and interaction patterns. Pharmacokinetic properties were assessed using ADMET predictions, and molecular dynamics (MD) simulations were conducted to validate the binding stability. Among the candidates, Apigetrin demonstrated favorable ADMET properties, a high safety profile, and stable binding within the ATP-binding pocket of PI3Kα. These findings suggest that Apigetrin is a promising natural PI3Kα inhibitor with potential therapeutic relevance for HNSCC.
Collapse
Affiliation(s)
- Wenqing Jia
- College of Chemistry and Chemical Engineering, Qilu Normal University, Jinan 250200, China
| | - Xianchao Cheng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
9
|
Khine HEE, Suriya U, Rungrotmongkol T, Chamni S, Lu Y, Bénard A, Lan B, Mukhopadhyay D, Chang D, Biankin A, Schneider-Stock R, Grützmann R, Sungthong R, Pilarsky C, Chaotham C. Jorunnamycin A induces apoptosis in pancreatic ductal adenocarcinoma cells, spheroids, and patient-derived organoids by modulating KRAS-mediated survival pathways. Sci Rep 2025; 15:11376. [PMID: 40175514 PMCID: PMC11965318 DOI: 10.1038/s41598-025-95766-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a poor prognosis, frequently driven by oncogenic KRAS mutations. Among these, KRAS G12D is the most prevalent, contributing to chemoresistance and limiting the efficacy of current therapeutic strategies. This study investigates the therapeutic potential of jorunnamycin A (JA), a bioactive compound derived from the marine sponge Xestospongia, in PDAC. Molecular docking analyses were performed to assess JA's binding affinity for various KRAS protein variants. The synergistic effects of JA in combination with standard chemotherapeutic agents were evaluated using the Bliss independence model in pancreatic cancer cell lines and patient-derived PDAC organoids harboring distinct KRAS mutations. Furthermore, western blot analysis was performed to examine the impact the molecular mechanisms underlying JA's anticancer activity. JA demonstrated potent anticancer activity against PDAC cells, irrespective of their KRAS mutation status. In silico molecular docking and protein suppression studies indicated a strong binding affinity between JA and KRAS G12D. Synergistic interactions between JA and various PDAC chemotherapeutic agents, including oxaliplatin, SN-38, paclitaxel, 5-fluorouracil, and gemcitabine, were observed using the Bliss independence model. Notably, co-treatment with JA at a 10-fold lower concentration significantly enhanced the cytotoxicity of oxaliplatin, reducing its IC50 values around tenfold. This synergistic impact was further validated in both KRAS G12D spheroids and patient-derived PDAC organoids harboring KRAS G12D and other KRAS variants. Mechanistically, the JA-oxaliplatin combination enhanced caspase-3/7 activation, suppressed key KRAS-mediated survival pathways (STAT3, B/C-RAF, AKT, and ERK), and led to the downregulation of anti-apoptotic proteins (MCL-1 and BCL-2). These findings highlight JA as a promising therapeutic candidate for PDAC, particularly in the context of KRAS G12D-driven tumors. Further investigations into its pharmacokinetics and clinical feasibility are warranted to explore its full potential in PDAC treatment.
Collapse
Affiliation(s)
- Hnin Ei Ei Khine
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Utid Suriya
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Thanyada Rungrotmongkol
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yanxi Lu
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Alan Bénard
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Bin Lan
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410002, China
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, 32224, USA
| | - David Chang
- Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Andrew Biankin
- Experimental Tumor Pathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Regine Schneider-Stock
- Experimental Tumor Pathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Rungroch Sungthong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
10
|
Yasmeen N, Ahmad Chaudhary A, K Niraj RR, Lakhawat SS, Sharma PK, Kumar V. Screening of phytochemicals from Clerodendrum inerme (L.) Gaertn as potential anti-breast cancer compounds targeting EGFR: an in-silico approach. J Biomol Struct Dyn 2025; 43:2781-2823. [PMID: 38141177 DOI: 10.1080/07391102.2023.2294379] [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: 07/25/2023] [Accepted: 12/04/2023] [Indexed: 12/25/2023]
Abstract
Breast cancer (BC) is the most prevalent malignancy among women around the world. The epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor (RTK) of the ErbB/HER family. It is essential for triggering the cellular signaling cascades that control cell growth and survival. However, perturbations in EGFR signaling lead to cancer development and progression. Hence, EGFR is regarded as a prominent therapeutic target for breast cancer. Therefore, in the current investigation, EGFR was targeted with phytochemicals from Clerodendrum inerme (L.) Gaertn (C. inerme). A total of 121 phytochemicals identified by gas chromatography-mass spectrometry (GC-MS) analysis were screened against EGFR through molecular docking, ADMET analyses (Absorption, Distribution, Metabolism, Excretion, and Toxicity), PASS predictions, and molecular dynamics simulation, which revealed three potential hit compounds with CIDs 10586 [i.e. alpha-bisabolol (-6.4 kcal/mol)], 550281 [i.e. 2,(4,4-Trimethyl-3-hydroxymethyl-5a-(3-methyl-but-2-enyl)-cyclohexene) (-6.5 kcal/mol)], and 161271 [i.e. salvigenin (-7.4 kcal/mol)]. The FDA-approved drug gefitinib was used to compare the inhibitory effects of the phytochemicals. The top selected compounds exhibited good ADMET properties and obeyed Lipinski's rule of five (ROF). The molecular docking analysis showed that salvigenin was the best among the three compounds and formed bonds with the key residue Met 793. Furthermore, the molecular mechanics generalized born surface area (MMGBSA) calculations, molecular dynamics simulation, and normal mode analysis validated the binding affinity of the compounds and also revealed the strong stability and compactness of phytochemicals at the docked site. Additionally, DFT and DOS analyses were done to study the reactivity of the compounds and to further validate the selected phytochemicals. These results suggest that the identified phytochemicals possess high inhibitory potential against the target EGFR and can treat breast cancer. However, further in vitro and in vivo investigations are warranted towards the development of these constituents into novel anti-cancer drugs.
Collapse
Affiliation(s)
- Nusrath Yasmeen
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | | | | | | | - Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
- Amity Institute of Pharmacy, Amity University Rajasthan, Jaipur, India
| |
Collapse
|
11
|
Gerić M, Nanić L, Micek V, Novak Jovanović I, Gajski G, Rašić D, Orct T, Ljubojević M, Karaica D, Jurasović J, Vrhovac Madunić I, Peraica M, Sabolić I, de Andrade VM, Breljak D, Rubelj I. The Impact of Resveratrol and Melatonin on the Genome and Oxidative Status in Ageing Rats. Nutrients 2025; 17:1187. [PMID: 40218945 PMCID: PMC11990809 DOI: 10.3390/nu17071187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Given the growing challenges posed by an ageing population, particularly in Western countries, we aimed to investigate the potential geroprotective effects of resveratrol and melatonin in ageing rats. METHODS The animals were treated with these two compounds starting at 3 months of age and continuing until 1 year or 2 years of age. Using a multibiomarker approach, we assessed DNA damage, telomere length, and the oxidative status in their urine, liver, and kidneys. RESULTS Despite employing this experimental approach, our results did not provide conclusive evidence of geroprotective effects across the evaluated organs. However, we observed sex-dependent differences in response to treatment. CONCLUSIONS Given the high potency of these two compounds, further research is warranted to explore their incorporation into daily routines as a strategy to mitigate ageing-related effects.
Collapse
Affiliation(s)
- Marko Gerić
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Lucia Nanić
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Vedran Micek
- Animal Breeding Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Ivana Novak Jovanović
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Goran Gajski
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Dubravka Rašić
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Tatjana Orct
- Division of Occupational and Environmental Health, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Marija Ljubojević
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Dean Karaica
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Jasna Jurasović
- Animal Breeding Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Ivana Vrhovac Madunić
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Maja Peraica
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Ivan Sabolić
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Vanessa Moraes de Andrade
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina–UNESC, Criciúma 88806-000, Brazil;
| | - Davorka Breljak
- Division of Toxicology, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Ivica Rubelj
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
12
|
Chittasupho C, Samee W, Mangmool S, Karuna N, Anuchapreeda S, Okonogi S, Athikomkulchai S. Phytochemical Characterization and Anticancer Activity of Clerodendrum chinense Leaf Extract Against Breast and Cervical Cancer Cells. Int J Mol Sci 2025; 26:2729. [PMID: 40141371 PMCID: PMC11943097 DOI: 10.3390/ijms26062729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/15/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer remains a significant global health challenge, necessitating novel therapeutic interventions. Clerodendrum chinense leaf extract (CCL) has gained interest for its potential anticancer properties due to its bioactive composition. This study aims to evaluate the cytotoxic effects of CCL against MCF-7 breast cancer and HeLa cervical cancer cells and elucidate its mechanisms of action. High-performance liquid chromatography identified verbascoside, isoverbascoside, and hispidulin as the major bioactive compounds. CCL exhibited time- and dose-dependent cytotoxicity, with MCF-7 cells showing greater sensitivity (IC50 = 126.8 µg/mL, 72 h) than HeLa cells (216.1 µg/mL, 72 h). Flow cytometry confirmed apoptotic induction, with late apoptosis increasing at moderate concentrations (16.03-23.55%) and necrosis prevailing at higher doses (50.80-63.68%). Reactive oxygen species generation was significantly elevated in MCF-7 (70.2%) and HeLa (60.4%) cells at 250 µg/mL. CCL effectively suppressed colony formation and cell migration in a dose-dependent manner. Molecular docking studies demonstrated that apoptosis induction of CCL bioactive compounds may mediate through the pro-apoptotic BCL2 associated X, apoptosis regulator (BAX) regulator. These findings highlight the potential of CCL as a natural anticancer agent with multiple mechanisms, including reactive oxygen species (ROS)-induced apoptosis, BAX activation, and inhibition of proliferation and metastasis.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (S.O.)
- Center of Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Weerasak Samee
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Srinakharinwirot University, Ongkharak, Nakhon Nayok 26120, Thailand;
| | - Supachoke Mangmool
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (N.K.)
| | - Narainrit Karuna
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.M.); (N.K.)
| | - Songyot Anuchapreeda
- Center of Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn Okonogi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (S.O.)
- Center of Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sirivan Athikomkulchai
- Department of Pharmacognosy, Faculty of Pharmacy, Srinakharinwirot University, Ongkharak, Nakhon Nayok 26120, Thailand
| |
Collapse
|
13
|
Bithi SA, Al Hasan MS, Bhuia MS, Mia E, Yana NT, Hasan AMW, Uddin MB, Sayeed MA, Emon Y, Hasan R, Chowdhury R, Islam MT. Botanical sources, biopharmaceutical profile, anticancer effects with mechanistic insight, toxicological and clinical evidence of prunetin: a literature review. Med Oncol 2025; 42:87. [PMID: 40029540 DOI: 10.1007/s12032-025-02646-z] [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: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
Prunetin (PRU), a naturally occurring flavonoid, has gained recognition for its wide-ranging therapeutic benefits, though its anticancer properties have yet to be extensively reviewed. This study explores the potential of PRU in targeting critical molecular pathways involved in tumor progression, including oxidative stress, apoptosis, cell cycle regulation, and metastasis. Data were compiled from reputable sources, including PubMed, Springer Link, Scopus, Wiley Online, Web of Science, ScienceDirect, and Google Scholar. The findings emphasize PRU's ability to mitigate oxidative stress, promote apoptosis, and regulate the cell cycle in cancer cells. Its anti-inflammatory and anti-angiogenic properties further enhance its effectiveness against cancer. Mechanistic studies reveal that PRU suppresses oncogenic pathways such as PI3K/Akt/mTOR (Phosphoinositide 3-kinase/Protein kinase B/Mammalian target of rapamycin) while activating tumor-suppressor mechanisms. Experimental models show that PRU effectively inhibits cancer cell proliferation and metastasis. Additionally, PRU exhibits favorable pharmacokinetics, demonstrating high intestinal absorption (95.5%), good Caco-2 permeability, and metabolism via CYP1A2, CYP2C19, CYP2C9, and CYP3A4, though it has poor blood-brain barrier (BBB) permeability and limited aqueous solubility, posing challenges for systemic bioavailability. Beyond its anticancer properties, PRU displays broad pharmacological relevance, including anti-inflammatory, cardioprotective, neuroprotective, anti-obesity, and osteoprotective effects, mediated through pathways, such as NF-κB, MAPK, and AMPK. Toxicological studies indicate a favorable safety profile, with low cytotoxicity in normal cells and no significant toxicity at high doses in preclinical models. While clinical evidence on PRU remains limited, studies on structurally related isoflavones suggest promising therapeutic potential, necessitating further clinical trials to establish its efficacy and safety in humans.
Collapse
Affiliation(s)
- Sumaya Akter Bithi
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh.
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh
| | - Emon Mia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh
| | - Noshin Tasnim Yana
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh
| | - Ali Mohamod Wasaf Hasan
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Santosh, Tangail, Bangladesh
| | | | - Md Abu Sayeed
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Santosh, Tangail, Bangladesh
| | - Yasin Emon
- Department of Pharmacy, Islamic University, Kushtia, 7003, Bangladesh
| | - Rubel Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh
| | - Muhammad Torequl Islam
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh.
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
14
|
Sipping M, Kumar TS, Kamdem N. Scientific investigation on antibacterial, antioxidant, cytotoxic effects and TLC bioautography of Terminalia schimperiania stem bark extracts. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025; 22:114-123. [PMID: 39789714 DOI: 10.1515/jcim-2024-0251] [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: 08/03/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES Terminalia schimperiana Hochst, belonging to the Combretaceae family, is known for its ethnomedicinal values, particularly in treating various diseases in Africa. This study aimed to investigate the antibacterial, antioxidant, and cytotoxic properties of T. schimperiana stem bark extracts, with a specific focus on assessing their bioactive potential and identifying active compounds via TLC bioautography. METHODS The in vitro antimicrobial activity was assessed using the agar well diffusion method against selected clinical strains. Antioxidant activity was evaluated using several methods including free radical scavenging, ferrous ion chelation assays and total phenolic content analysis. The cytotoxicity of the extracts was assessed using MTT assay towards Raw 264.7 and Vero cell lines. RESULTS All extracts demonstrated significant antibacterial activity against the bacteria tested, with inhibition zones (IZ) ranging from 6.50 ± 0.71 to 15.50 ± 0.71 mm and minimum inhibitory concentrations (MIC) ranging from 1.95 to 1,250 μg/mL. The hydroethanolic extract exhibited strongest antioxidant activities with EC50 values of 188.50; 245.30, and 281.50 μg/mL for DPPH; ABTS, ferrous ion chelation assays respectively, and a high content of phenolic compounds (101.67 ± 2.97 µgEFA/mg DW). Importantly, no cytotoxic effects were observed on Raw 264.7 and Vero cell lines. HPTLC analysis identified alkaloids and phenolic compounds in both aqueous and hydroethanolic extracts. CONCLUSIONS These findings indicate T. schimperiana provides a wealth of bio-compounds that can be utilised in the pharmaceutical industry as antibacterial and antioxidant agents to combat antibiotic resistance.
Collapse
Affiliation(s)
- Marius Sipping
- PKFokam Institute of Excellence, Yaoundé, Cameroon
- Laboratory of Phytoprotection and Valorization of Genetic Resources, Biotechnology Centre, Nkolbisson, 201336 University of Yaoundé 1 , Yaoundé, Cameroon
| | | | | |
Collapse
|
15
|
Gupta U, Kavya MP, Jayalakshmi K, Sinha N. Unlocking the Chemical Diversity of Plant Catharanthus roseus: Nuclear Magnetic Resonance Spectroscopy Approach. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2025; 63:205-215. [PMID: 39702899 DOI: 10.1002/mrc.5504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Catharanthus roseus, also known as Madagascar periwinkle, is a perennial plant renowned for its extensive pharmacological properties. It produces vital chemotherapeutic compounds, including vinblastine and vincristine, and exhibits anti-inflammatory, antidiabetic, and antioxidant activities. In this study, we utilized a range of two-dimensional (2D) nuclear magnetic resonance (NMR) techniques, such as 1H-1H correlation spectroscopy (COSY), 1H-1H J-resolved NMR, and 1H-13C heteronuclear single quantum coherence (HSQC) sensitivity-enhanced NMR spectroscopy, to identify key metabolites in C. roseus leaf extracts. Given the presence of numerous metabolites with closely spaced multiplet resonances, the 1H NMR spectra often exhibit significant signal overlap, making metabolite identification difficult or even impossible. However, the use of 2D NMR techniques effectively overcame this challenge, allowing for the precise identification of important alkaloids, such as vindoline, vinblastine, serpentine, and ajmalicine, along with essential metabolites like organic acids, amino acids, and carbohydrates. The extract contained a variety of bioactive compounds, including organic acids crucial for the tricarboxylic acid (TCA) cycle, branched-chain amino acids vital for metabolic functions, and alkaloids with substantial therapeutic potential. This comprehensive study underscores the continued significance of C. roseus in both traditional and modern medicine, emphasizing its intricate metabolic network and its potential in the development of novel therapeutics.
Collapse
Affiliation(s)
- Upasna Gupta
- Centre of Bio-Medical Research (CBMR), Sanjay Gandhi Post Institute of Medical Sciences Campus, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - M P Kavya
- Department of Chemistry, Karnatak University's Karnatak Science College, Dharwad, India
| | - K Jayalakshmi
- Department of Chemistry, Karnatak University's Karnatak Science College, Dharwad, India
| | - Neeraj Sinha
- Centre of Bio-Medical Research (CBMR), Sanjay Gandhi Post Institute of Medical Sciences Campus, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
16
|
Dave A, Park EJ, Piya S, Pezzuto JM. Long-Term Dietary Consumption of Grapes Alters Phenotypic Expression in Skeletal Muscle of Aged Male and Female Mice. Foods 2025; 14:695. [PMID: 40002138 PMCID: PMC11854663 DOI: 10.3390/foods14040695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background: Nutrigenomics investigates how diet influences gene expression and how genetic variation impacts dietary responses. Grapes, rich in phytochemicals, exhibit potential disease-preventive properties through nutrigenomic mechanisms rather than direct chemical interactions. This study aimed to explore the modulation of gene expression in muscle tissue resulting from long-term grape consumption. (2) Methods: A mouse model was employed to assess gene expression in the skeletal muscles of males and females fed a grape-enriched diet versus a bland diet over 2.5 years. Heatmaps and principal component analyses were performed to identify patterns, and pathway analyses using KEGG, GO, and Reactome were conducted. (3) Results: Significant sex-specific gene expression changes were observed, with female phenotypes showing greater alterations and converging toward male-like characteristics. Twenty-five differentially expressed genes associated with muscle health were identified. Up-regulated genes such as Ahsg, Alb, Apoa1, and Arg1, and down-regulated genes including Camp, Lcn2, and Irf4, suggest improved muscle function. (4) Conclusions: Long-term grape consumption appears to enhance female muscle traits toward a male-like phenotype, potentially indicating broader health benefits. Further studies and clinical trials are needed to confirm human applicability and the physiological implications of these findings. Nonetheless, this research underscores the role of nutrigenomics in understanding dietary influences on gene expression and sex-specific responses.
Collapse
Affiliation(s)
- Asim Dave
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA;
| | - Eun-Jung Park
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Sumi Piya
- Department of Pathology, UMass Chan Medical School-Baystate, Springfield, MA 01199, USA;
| | - John M. Pezzuto
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Medicine, UMass Chan Medical School-Baystate, Springfield, MA 01199, USA
| |
Collapse
|
17
|
Joshi H, Bhushan S, Dimri T, Sharma D, Sak K, Chauhan A, Chauhan R, Haque S, Ahmad F, Kumar M, Tuli HS, Kaur D. Anti-tumor potential of Harmine and its derivatives: recent trends and advancements. Discov Oncol 2025; 16:189. [PMID: 39954215 PMCID: PMC11829886 DOI: 10.1007/s12672-025-01893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
Harmine is a β-carboline alkaloid derived from Peganum harmala, showing a solid antitumor potential in different types of human cancer cells. Unfortunately, the clinical application of this natural alkaloid has been impeded till now by severe toxic side effects, especially neurotoxicity, besides its poor water solubility. Therefore, over the recent years, several semisynthetic derivatives of harmine have been prepared and studied concerning their abilities to inhibit tumor cell proliferation, survival, angiogenesis, migration, and invasion in diverse preclinical models. This review article summarizes the anticancer effects of harmine and its synthetic derivatives, demonstrating their high potential to be developed as novel anticancer drugs to supplement our current therapeutic arsenal in the fight against the globally increasing rate of malignant disorders.
Collapse
Affiliation(s)
- Hemant Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sakshi Bhushan
- Department of Plant Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Tanisha Dimri
- Department of Biotechnology, All India Institute of Medical Science, New Delhi, 110029, India
| | - Deepak Sharma
- Department of Plant Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | | | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, Uttar Pradesh, 201313, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to Be University, Dehradun, Uttarakhand, 248002, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
- School of Medicine, Universidad Espiritu Santo, Samborondon, Ecuador
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, 632014, India.
| | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University Sadopur, Ambala, 134007, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Ambala, Mullana, 133207, India
| | - Damandeep Kaur
- University Center for Research and Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, India
| |
Collapse
|
18
|
Yu Q, Tian L, Zhang J. Chemoprotective Effect of Myrrhone against Diethylnitrosamine and Ferric Nitrile Induced Renal Cancer via Alteration of HO-1/Nrf2 and TRL4/NF-κB Signaling Pathway. DOKL BIOCHEM BIOPHYS 2025; 520:130-143. [PMID: 39847305 DOI: 10.1134/s160767292460091x] [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: 08/20/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is the most prevalent form of kidney cancer and is the primary malignancy affecting the genitourinary system. It represents the majority of kidney cancer cases and is distinguished by its aggressive nature and high mortality rate. The current study investigates the chemoprotective effect of myrrhone against Diethylnitrosamine (DEN) and ferric nitrile (Fe-NTA) induced RCC in rats and elucidates the underlying mechanism. METHODS Following a single dose of intraperitoneal DEN (200 mg/kg) and a twice-weekly administration of Fe-NTA, rats were administered either an oral dose of myrrhone (5, 10, or 15 mg/kg). The body weights and food intake of the rats were monitored at regular intervals, and the levels of renal cancer markers, antioxidants, inflammatory markers, and other parameters were assessed. Additionally, histopathological studies were conducted on the renal tissues, and the mRNA expression of Bax, Bcl-2, HO-1, SOD2, mtDNA, ATP8, PGC-1α, TRL4, and NF-κB was analyzed. RESULTS The dosage-dependent administration of myrrhone demonstrated a remarkable suppression of tumor incidence and an improvement in body weight and food intake. Myrrhone markedly decreased the level of ODC, Thymidine [3H] incorporation, and renal parameters such as creatinine, uric acid, BUN, Kim-1, Cysc-C, and LDH. Additionally, myrrhone significantly altered the levels of MDA, GSH, GPx, CAT, and SOD, as well as inflammatory cytokines such as TNF-α, INF-γ, IL-1β, IL-6, and IL-10, and inflammatory parameters such as COX-2, PGE2, TGF-β1, NF-κB, and iNOS. Furthermore, myrrhone significantly decreased the histopathological score and improved the condition of histopathology. Finally, myrrhone significantly altered the mRNA expression of Bax, Bcl-2, HO-1, SOD2, mtDNA, ATP8, PGC-1α, TRL4, and NF-κB. CONCLUSION The result clearly showed the chemoprotective effect of myrrhone against diethylnitrosamine and ferric nitrile induced Renal Cancer via alteration of HO-1/Nrf2 and TRL4/NF-κB Signaling pathway.
Collapse
Affiliation(s)
- Qian Yu
- Department of Geriatrics, Affiliated Hospital of Hebei University, 071000, Baoding City, Hebei Province, China
| | - Ling Tian
- Hebei Key Laboratory, Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, 071000, Baoding, Hebei, China
| | - Jiwei Zhang
- Affiliated Hospital of Hebei University, 071000, Baoding City, Hebei Province, China.
| |
Collapse
|
19
|
Sachdeva S, Sarethy IP. Diving into freshwater microbial metabolites: Pioneering research and future prospects. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025; 35:282-300. [PMID: 38887995 DOI: 10.1080/09603123.2024.2351153] [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: 10/16/2023] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
In practically every facet of life, especially nutrition, agriculture, and healthcare, microorganisms offer a prospective origin for abundant natural substances and products. Among these microorganisms, bacteria also possess the capability to rapidly acclimate to diverse environments, utilize varied resources, and effectively respond to environmental fluctuations, including those influenced by human activities like pollution and climate change. The ever-changing environment of freshwater bodies influences bacterial communities, offering opportunities for improving health and environmental conservation that remain unexplored. Herein, the study discusses the bacterial taxa along with specialised metabolites with antioxidant, antibacterial, and anticancer activity that have been identified from freshwater environments, thus achieving Sustainable Development Goals addressing health and wellbeing (SDG-3), economic growth (SDG-8) along with industrial development (SDG-9). The present review is intended as a compendium for research teams working in the fields of medicinal chemistry, organic chemistry, clinical research, and natural product chemistry.
Collapse
Affiliation(s)
- Saloni Sachdeva
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Indira P Sarethy
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
20
|
Sharma V, Kumar G, Kumar P, Khajuria R, Nemiwal M. Innovative anticancer molecule andrographolide: a concise review of its pharmacological targets. Nat Prod Res 2025:1-13. [PMID: 39798140 DOI: 10.1080/14786419.2025.2450783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Labdane diterpenoid lactone andrographolide has gained attention in medicinal research due to its potential anticancer properties in terms of suppression of the growth, propagation, and relocation of various types of cancerous cells. The current review provides deep insight into the pharmacological analysis of the anticancer secondary metabolite andrographolide. We have attempted to keep an overview on the interaction of promising drugs like ligand molecule andrographolide with various biological targets. The observation indicates that andrographolide significantly down-regulates the growth of cancer cells through various mechanisms via diminishing inducible nitric oxide synthase (iNOS) expression, attenuating Akt and JNK signalling cascade, inhibiting NF-κB activation, ROS generation in the neoplastic cells etc. This bio-molecule is a potent therapeutic agent that can be applied in treating and preventing inflammatory vascular diseases. This study may be beneficial in the area of drug development research, leading to better management of cancer and many other inflammatory diseases.
Collapse
Affiliation(s)
- Venu Sharma
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, India
| | - Gourav Kumar
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, India
| | - Parveen Kumar
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, India
| | - Rajni Khajuria
- Department of Chemistry, P.S.P.S. Government College for Women, Jammu and Kashmir, India
| | - Meena Nemiwal
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, India
| |
Collapse
|
21
|
Ahmad M, Tahir M, Hong Z, Zia MA, Rafeeq H, Ahmad MS, Rehman SU, Sun J. Plant and marine-derived natural products: sustainable pathways for future drug discovery and therapeutic development. Front Pharmacol 2025; 15:1497668. [PMID: 39834812 PMCID: PMC11743463 DOI: 10.3389/fphar.2024.1497668] [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: 09/17/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Plant- and marine-derived natural products are rich sources of bioactive compounds essential for drug discovery. These compounds contain complex mixtures of metabolites, which collectively contribute to their pharmacological properties. However, challenges arise in the isolation of individual bioactive compounds, owing to their intricate chemistry and low abundance in natural extracts. Despite these limitations, numerous plant and marine-derived compounds have achieved regulatory approval, particularly for treating cancer and infectious diseases. This review explores the therapeutic potential of plant and marine sources along with innovative extraction and isolation methods that support sustainable drug development. Future perspectives will highlight the role of responsible innovation, artificial intelligence, and machine learning in advancing drug discovery, underscoring the importance of continued research to meet global health needs.
Collapse
Affiliation(s)
- Muhammad Ahmad
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital-Ganzhou Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Institute of Physiology and Pharmacology, Faculty of Veterinary Science, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Maleha Tahir
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital-Ganzhou Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zibin Hong
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital-Ganzhou Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Muhammad Anjum Zia
- Enzyme Biotechnology Lab, Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Hamza Rafeeq
- Enzyme Biotechnology Lab, Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Muhammad Shaheez Ahmad
- Enzyme Biotechnology Lab, Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Saif ur Rehman
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital-Ganzhou Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junming Sun
- Laboratory Animal Center, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
22
|
Wu T, Chen Z, Liu X, Wu X, Wang Z, Guo W. Targeting RSK2 in Cancer Therapy: A Review of Natural Products. Anticancer Agents Med Chem 2025; 25:35-41. [PMID: 39248063 DOI: 10.2174/0118715206329546240830055233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
P90 ribosomal S6 kinase 2 (RSK2) is an important member of the RSK family, functioning as a kinase enzyme that targets serine and threonine residues and contributes to regulating cell growth. RSK2 comprises two major functional domains: the N-terminal kinase domain (NTKD) and the C-terminal kinase domain (CTKD). RSK2 is situated at the lower end of the Mitogen-activated protein kinases (MAPK) signaling pathway and is phosphorylated by the direct regulation of Extracellular signal-regulating kinase (ERK). RSK2 has been found to play a pivotal role in regulating cell proliferation, apoptosis, metastasis, and invasion in various cancer cells, including breast cancer and melanoma. Consequently, RSK2 has emerged as a potential target for the development of anti-cancer drugs. Presently, several inhibitors are undergoing clinical trials, such as SL0101. Current inhibitors of RSK2 mainly bind to its NTK or CTK domains and inhibit their activity. Natural products serve as an important resource for drug development and screening and with the potential to identify RSK2 inhibitors. This article discusses how RSK2 influences tumor cell proliferation, prevents apoptosis, arrests the cell cycle process, and promotes cancer metastasis through its regulation of downstream pathways or interaction with other biological molecules. Additionally, the paper also covers recent research progress on RSK2 inhibitors and the mechanisms of action of natural RSK2 inhibitors on tumors. This review emphasizes the significance of RSK2 as a potential therapeutic target in cancer and offers a theoretical basis for the clinical application of RSK2 inhibitors.
Collapse
Affiliation(s)
- Tianhui Wu
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, China
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Ziming Chen
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, China
| | - Xin Liu
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, China
| | - Xinyan Wu
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, China
| | - Zhaobo Wang
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, China
| | - Weiqiang Guo
- School of Chemistry and Life Science, Suzhou University of Science and Technology, Suzhou, China
| |
Collapse
|
23
|
Thilagam NBS, Karthik V, Gnanasambandan R, Sowmya C. A Comprehensive Review on Current Knowledge and Future Potential of Topical Therapies in Breast Cancer Treatment. Eur J Breast Health 2025; 21:9-15. [PMID: 39744878 PMCID: PMC11706127 DOI: 10.4274/ejbh.galenos.2024.2024-9-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Breast cancer remains one of the most prevalent malignancies among women globally. Despite advances in therapeutic options, the prognosis often remains challenging. Breast cancer typically originates in the epithelial lining of glandular tissue ducts (85%) or lobules (15%). Initially confined to these areas (in situ), it generally remains asymptomatic and poses little risk of metastasis. The primary treatments for breast cancer include surgery, radiation therapy, chemotherapy, hormone therapy, and targeted therapy. Although these interventions have advanced significantly and have improved patient survival rates, they are connected with numerous im mediate and long-term side effects. Effective breast cancer treatment aims to maximize efficacy while minimizing adverse effects. Given that many breast cancers are specific to the breast, developing safe and targeted therapeutic strategies will be of benefit. This review examined the current literature on the effectiveness of topical therapies for breast cancer. Studies and clinical trials were evaluated that have investigated these treatments, focusing on their safety, ease of application, and patient acceptance. Recently, topical drug delivery is transforming breast cancer therapy, offering precision and reduced systemic toxicity. Emu oil-enhanced tamoxifen showed superior transdermal effectiveness, while raloxifene gel achieved 2.77 times greater bioavailability than oral forms. Tamoxifen nanoemulgels and microneedle arrays with resveratrol further enhanced localized delivery, These therapies have gained patient acceptance due to their non-invasive nature, lack of gastrointestinal side effects, ease of application, and favourable safety and therapeutic profiles and setting a new benchmark for innovative and patient-friendly treatments. This review summarizes the findings from various studies, highlighting the benefits and limitations of topical therapies. Topical therapies offer a promising noninvasive option for breast cancer treatment with fewer side effects. These treatments have shown favorable therapeutic and safety profiles, making them an attractive option for patients.
Collapse
Affiliation(s)
- N. Bharathi Sai Thilagam
- Department of Pharmaceutics, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - V.P. Karthik
- Department of Pharmacology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - R. Gnanasambandan
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT University, Vellore, India
| | - C. Sowmya
- Department of Pharmaceutics, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
24
|
Haghighat ZA, Safekordi A, Ardjmand M, Akbarzadeh A. Exploring the Antitumor Efficacy of PEGylated Liposomes Loaded with Licorice Extract for Cancer Therapy. Curr Cancer Drug Targets 2025; 25:357-369. [PMID: 38685810 DOI: 10.2174/0115680096292153240416115744] [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: 11/12/2023] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Glycyrrhizic Acid (GA), a compound derived from licorice, has exhibited promising anticancer properties against several cancer types, including Prostate Cancer (PCa) and Gastric Cancer (GCa). OBJECTIVE This study has introduced a novel approach involving the encapsulation of GA and Licorice extract (Lic) into Polyethylene Glycol Liposomes (PEG-Lip) and assessed their efficacy against AGS (human gastric cancer) and PC-3 (human prostate cancer) cells, marking the first report of this endeavor. METHODS We synthesized GA-loaded PEG-Lip (GA PEG-Lip) and Lic-loaded PEG-Lip (Lic PEG-Lip) through the reverse-phase evaporation method. RESULTS Characterization of these liposomal formulations revealed their size, drug encapsulation, and loading efficiencies to be 110 ± 2.05 nm, 117 ± 1.24 nm; 61 ± 0.81%, 34 ± 0.47%; and 8 ± 0.41% and 4.6 ± 0.21%, respectively. Importantly, the process has retained the chemical structure of both GA and Lic. Furthermore, GA and Lic have been released from the PEG-Lip formulations in a controlled manner. In our experiments, both nanoformulations exhibited enhanced cytotoxic effects against AGS and PC-3 cells. Notably, GA PEG-Lip outperformed Lic PEG-Lip, reducing the viability of PC-3 and AGS cells by 12.5% and 15.9%, respectively. CONCLUSION These results have been corroborated by apoptosis assays, which have demonstrated GA PEG-Lip and Lic PEG-Lip to induce stronger apoptotic effects compared to free GA and Lic on both PC-3 and AGS cells. This study has underscored the potential of encapsulating GA and Lic in PEG-Lip as a promising strategy to augment their anticancer efficacy against prostate and gastric cancers.
Collapse
Affiliation(s)
- Zeinab Azizi Haghighat
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Aliakbar Safekordi
- Chemical Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Mehdi Ardjmand
- Department of Chemical Engineering, South Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Azim Akbarzadeh
- Department of Pilot Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
25
|
Meng Y, Wang Y, Li S, Cai Z, Zhuang G, Yang Y. Design, Synthesis, and Antitumor Activity of Novel Eupatilin Derivatives Based on the Mannich Reaction. Chem Pharm Bull (Tokyo) 2025; 73:112-120. [PMID: 40010724 DOI: 10.1248/cpb.c24-00599] [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] [Indexed: 02/28/2025]
Abstract
Eupatilin, a natural bioactive flavone, is the active ingredient in traditional Chinese medicine Artemisia argyi Levl. et Vant. To enhance the antitumor effect of eupatilin, we designed a series of novel eupatilin-Mannich derivatives and investigated antitumor activity against several human cancer cell lines, including gastric cancer cells (AGS), esophageal cancer cells (Eca-109), and breast cancer cells (MDA-MB-231). Among all derivatives, the majority demonstrated superior antitumor activity compared to eupatilin, with compound 3d exhibiting the most effective antitumor activity against AGS cells. Furthermore, compound 3d effectively inhibited colony formation and migration of AGS cells. Network pharmacology combined with molecular docking studies indicated that compound 3d exerts antitumor activity by targeting the Hsp90AA1 and multiple signaling pathways. In addition, the Western blot experiment results showed that compound 3d reduced the expression of Hsp90AA1 in AGS cells, indicating that Hsp90AA1 may be the potential target of compound 3d. In summary, several novel eupatilin derivatives were prepared via the Mannich reaction, representing the first structure modification study of eupatilin. The mechanism of action of compound 3d was estimated through cell experiments, network pharmacology, molecular docking, and Western blot experiments, to provide lead compounds for the discovery of natural product-based antitumor candidates.
Collapse
Affiliation(s)
- Yaqi Meng
- Center of Scientific Research (Institute of Nanyang Geoherbs), Nanyang Medical College, Nanyang 473061, China
- Key Laboratory of Research on Effective Substances and Quality Control of Traditional Chinese Medicine, Nanyang Medical College, Nanyang 473061, China
| | - Yuqing Wang
- Center of Scientific Research (Institute of Nanyang Geoherbs), Nanyang Medical College, Nanyang 473061, China
- Key Laboratory of Research on Effective Substances and Quality Control of Traditional Chinese Medicine, Nanyang Medical College, Nanyang 473061, China
| | - Shujiao Li
- Center of Scientific Research (Institute of Nanyang Geoherbs), Nanyang Medical College, Nanyang 473061, China
- Key Laboratory of Research on Effective Substances and Quality Control of Traditional Chinese Medicine, Nanyang Medical College, Nanyang 473061, China
| | - Zhiyan Cai
- Center of Scientific Research (Institute of Nanyang Geoherbs), Nanyang Medical College, Nanyang 473061, China
- Key Laboratory of Research on Effective Substances and Quality Control of Traditional Chinese Medicine, Nanyang Medical College, Nanyang 473061, China
| | - Guo Zhuang
- Center of Scientific Research (Institute of Nanyang Geoherbs), Nanyang Medical College, Nanyang 473061, China
- Key Laboratory of Research on Effective Substances and Quality Control of Traditional Chinese Medicine, Nanyang Medical College, Nanyang 473061, China
| | - Yanli Yang
- Center of Scientific Research (Institute of Nanyang Geoherbs), Nanyang Medical College, Nanyang 473061, China
- Key Laboratory of Research on Effective Substances and Quality Control of Traditional Chinese Medicine, Nanyang Medical College, Nanyang 473061, China
- School of Pharmacy, Henan University, Kaifeng 475001, China
| |
Collapse
|
26
|
Elbadawi M, Efferth T. In Vivo and Clinical Studies of Natural Products Targeting the Hallmarks of Cancer. Handb Exp Pharmacol 2025; 287:95-121. [PMID: 38797749 DOI: 10.1007/164_2024_716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Despite more than 200 approved anticancer agents, cancer remains a leading cause of death worldwide due to disease complexity, tumour heterogeneity, drug toxicity, and the emergence of drug resistance. Accordingly, the development of chemotherapeutic agents with higher efficacy, a better safety profile, and the capability of bypassing drug resistance would be a cornerstone in cancer therapy. Natural products have played a pivotal role in the field of drug discovery, especially for the pharmacotherapy of cancer, infectious, and chronic diseases. Owing to their distinctive structures and multiple mechanistic activities, natural products and their derivatives have been utilized for decades in cancer treatment protocols. In this review, we delve into the potential of natural products as anticancer agents by targeting cancer's hallmarks, including sustained proliferative signalling, evading growth suppression, resisting apoptosis and cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. We highlight the molecular mechanisms of some natural products, in vivo studies, and promising clinical trials. This review emphasizes the significance of natural products in fighting cancer and the need for further studies to uncover their fully therapeutic potential.
Collapse
Affiliation(s)
- Mohamed Elbadawi
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
27
|
Georgiev B, Sidjimova B, Berkov S. Phytochemical and Cytotoxic Aspects of Amaryllidaceae Alkaloids in Galanthus Species: A Review. PLANTS (BASEL, SWITZERLAND) 2024; 13:3577. [PMID: 39771275 PMCID: PMC11678157 DOI: 10.3390/plants13243577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
The genus Galanthus (Amaryllidaceae) currently contains 25 plant species naturally occurring in Europe and the Middle East region. These perennial bulbous plants possess well-known medicinal and ornamental qualities. Alkaloid diversity is their most distinctive phytochemical feature. A total of 127 compounds (≈20% of all known Amaryllidaceae alkaloids) grouped in 16 structural types have been previously found in Galanthus extracts. Some structural types like galanthindole, graciline and plicamine were first discovered in Galanthus plants. Nine Galanthus species, however, remain unstudied regarding their alkaloid patterns. Intraspecific variability has only been studied in G. nivalis and G. elwesii. Amaryllidaceae alkaloids are molecules with anticholinesterase, antibacterial, antifungal, antiviral and anticancer properties. Galanthamine, isolated for the first time from Galanthus woronowii Losinsk., stands out as an acetylcholinesterase inhibitor approved for medical use by the FDA for the treatment of symptoms of Alzheimer's disease. Lycorine, narciclasine and pancratistatin are noteworthy cytotoxic and antitumor alkaloids. Structural types like galanthamine, homolycorine and haemanthamine are fairly well studied in anticancer research, but little to no information is available on galanthindole, graciline and other types. This review aims to present an update on the alkaloid diversity of Galanthus spp. and highlight the need for further research on the antitumor potential of these molecules.
Collapse
Affiliation(s)
- Borislav Georgiev
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | | | - Strahil Berkov
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| |
Collapse
|
28
|
Hossain MM, Soha K, Rahman A, Auwal A, Pronoy TUH, Rashel KM, Nurujjaman M, Rahman H, Roy TG, Khanam JA, Islam F. Rhodium complex [RhLI 2]I: a novel anticancer agent inducing tumor inhibition and apoptosis. Discov Oncol 2024; 15:782. [PMID: 39692939 DOI: 10.1007/s12672-024-01632-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024] Open
Abstract
Numerous chemotherapeutic agents are currently employed in cancer treatment, but many are associated with significant side effects. This study aims to identify a novel anticancer drug that minimizes or eliminates these adverse effects. The anticancer activity of the Rhodium (III) complex cis-[RhLI2]I was evaluated through both in vivo and in vitro functional assays. Apoptosis in cancer cells post-treatment was assessed using microscopy and gene expression analysis. In cytotoxicity screening via the brine shrimp lethality bioassay, the compound exhibited an LC50 value of 25.90 µg/mL (P < 0.001). It also achieved an 88.96% inhibition of cell growth (P < 0.001), an 82.39% increase in lifespan (P < 0.001), and a significant reduction in tumor weight at a dosage of 200 µg/kg in Ehrlich ascites carcinoma (EAC)-bearing Swiss albino mice. Restoration of hematological parameters, such as RBC, WBC, and hemoglobin levels, was observed in treated tumor-bearing mice compared to untreated EAC-bearing mice. The compound inhibited the growth and proliferation of breast cancer (MCF7) cells in a dose-dependent manner, achieving a maximum inhibition of 88.9% at 200 µg/mL. Apoptotic induction in MCF7 cells occurred through the upregulation of p53, Bax, caspase-3, -8, and -9, alongside the downregulation of the anti-apoptotic protein Bcl-2. No long-term adverse effects on hematological or biochemical parameters or tissue levels were observed in the mice. Given these findings, this compound demonstrates significant cytotoxic effects and has the potential to serve as a promising chemotherapeutic agent, warranting further investigation at more advanced stages.
Collapse
Affiliation(s)
- M Matakabbir Hossain
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Kazi Soha
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Arifur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Abdul Auwal
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Tasfik Ul Haque Pronoy
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - K M Rashel
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - M Nurujjaman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Habibur Rahman
- Department of Chemistry, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Tapashi G Roy
- Department of Chemistry, Faculty of Science, University of Chittagong, Chattogram, 4331, Bangladesh
| | - Jahan Ara Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Queenslan, 4222, Australia.
| |
Collapse
|
29
|
Suha H, Tasnim SA, Rahman S, Alodhayb A, Albrithen H, Poirier RA, Uddin KM. Evaluating the Anticancer Properties of Novel Piscidinol A Derivatives: Insights from DFT, Molecular Docking, and Molecular Dynamics Studies. ACS OMEGA 2024; 9:49639-49661. [PMID: 39713673 PMCID: PMC11656217 DOI: 10.1021/acsomega.4c07808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024]
Abstract
Cancer is characterized by uncontrolled cell growth and spreading throughout the body. This study employed computational approaches to investigate 18 naturally derived anticancer piscidinol A derivatives (1-18) as potential therapeutics. By examining their interactions with 15 essential target proteins (HIF-1α, RanGAP, FOXM1, PARP2, HER2, ERα, NGF, FAS, GRP78, PRDX2, SCF complex, EGFR, Bcl-xL, ERG, and HSP70) and comparing them with established drugs such as camptothecin, docetaxel, etoposide, irinotecan, paclitaxel, and teniposide, compound 10 emerged as noteworthy. In molecular dynamics simulations, the protein with the strongest binding to the crucial 1A52 protein exceeded druglikeness criteria and displayed extraordinary stability within the enzyme's pocket over varied temperatures (300-320 K). Additionally, density functional theory was used to calculate dipole moments and molecular orbital characteristics, as well as analyze the thermodynamic stability of the putative anticancer derivatives. This finding reveals a well-defined, potentially therapeutic relationship supported by theoretical analysis, which is in good agreement with subsequent assessments of their potential in vitro cytotoxic effects of piscidinol A derivatives (6-18) against various cancer cell lines. Future in vivo and clinical studies are required to validate these findings further. Compound 10 thus emerges as an intriguing contender in the fight against cancer.
Collapse
Affiliation(s)
- Humaera
Noor Suha
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Syed Ahmed Tasnim
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Shofiur Rahman
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah Alodhayb
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Department
of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hamad Albrithen
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Department
of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Raymond A. Poirier
- Department
of Chemistry, Memorial University, St. John’s, Newfoundland
and Labrador A1C 5S7, Canada
| | - Kabir M. Uddin
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| |
Collapse
|
30
|
Famurewa AC, George MY, Ukwubile CA, Kumar S, Kamal MV, Belle VS, Othman EM, Pai SRK. Trace elements and metal nanoparticles: mechanistic approaches to mitigating chemotherapy-induced toxicity-a review of literature evidence. Biometals 2024; 37:1325-1378. [PMID: 39347848 DOI: 10.1007/s10534-024-00637-7] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Anticancer chemotherapy (ACT) remains a cornerstone in cancer treatment, despite significant advances in pharmacology over recent decades. However, its associated side effect toxicity continues to pose a major concern for both oncology clinicians and patients, significantly impacting treatment protocols and patient quality of life. Current clinical strategies to mitigate ACT-induced toxicity have proven largely unsatisfactory, leaving a critical unmet need to block toxicity mechanisms without diminishing ACT's therapeutic efficacy. This review aims to document the molecular mechanisms underlying ACT toxicity and highlight research efforts exploring the protective effects of trace elements (TEs) and their nanoparticles (NPs) against these mechanisms. Our literature review reveals that the primary driver of ACT toxicity is redox imbalance, which triggers oxidative inflammation, apoptosis, endoplasmic reticulum stress, mitochondrial dysfunction, autophagy, and dysregulation of signaling pathways such as PI3K/mTOR/Akt. Studies suggest that TEs, including zinc, selenium, boron, manganese, and molybdenum, and their NPs, can potentially counteract ACT-induced toxicity by inhibiting oxidative stress-mediated pathways, including NF-κB/TLR4/MAPK/NLRP3, STAT-3/NLRP3, Bcl-2/Bid/p53/caspases, and LC3/Beclin-1/CHOP/ATG6, while also upregulating protective signaling pathways like Sirt1/PPAR-γ/PGC-1α/FOXO-3 and Nrf2/HO-1/ARE. However, evidence regarding the roles of lncRNA and the Wnt/β-catenin pathway in ACT toxicity remains inconsistent, and the impact of TEs and NPs on ACT efficacy is not fully understood. Further research is needed to confirm the protective effects of TEs and their NPs against ACT toxicity in cancer patients. In summary, TEs and their NPs present a promising avenue as adjuvant agents for preventing non-target organ toxicity induced by ACT.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki, Ebonyi, Nigeria.
- Centre for Natural Products Discovery, School of P harmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cletus A Ukwubile
- Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Bama Road, Maiduguri, Borno, Nigeria
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mehta V Kamal
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vijetha S Belle
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Eman M Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Cancer Therapy Research Center, Department of Biochemistry-I, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074, BiocenterWürzburg, Germany
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
31
|
Hai P, Jia H, Luo Z, Fan H, He Y, Li X, Lin P, Zhang Q, Gao Y, Yang J. Meroterpenoids with anti-triple negative breast cancer and antimicrobial activities from Arnebia euchroma. Fitoterapia 2024; 179:106234. [PMID: 39332506 DOI: 10.1016/j.fitote.2024.106234] [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: 07/13/2024] [Revised: 09/14/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
Two new meroterpenoids, arneuchrols A and B (1 and 2), together with twelve known analogs (3-14) were isolated from the root of Arnebia euchroma. The structures of 1 and 2 including their absolute configurations were elucidated by NMR, HRESIMS, and DFT calculation of their NMR and ECD data. The structure of pseudoshikonin I, firstly isolated from Lithospermi radix was revised as shikonofuran E (4). Anti-triple negative breast cancer (anti-TNBC) and antimicrobial activities of the isolated compounds were tested. Compounds 3, 4, 6, 7, 9, 10, and 13 exhibited potent inhibitory activity against TNBC (MDA-MB-231 cells) with IC50 values in the range of 0.18-4.58 μM. Compound 10 displayed antifungal activity against five plant pathogenic fungi with MIC values in the range of 6.25-25 μg/mL. Compound 9 exhibited antibacterial activity against Micrococcus lysodeikticus with MIC value of 12.5 μg/mL.
Collapse
Affiliation(s)
- Ping Hai
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China
| | - Haiyan Jia
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China; Evaluation and Research Center of Daodi Herbs of Jiangxi Province, Ganjiang New District 330000, China
| | - Zhiqiang Luo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Huixia Fan
- Evaluation and Research Center of Daodi Herbs of Jiangxi Province, Ganjiang New District 330000, China
| | - Yunqing He
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China; Key Lab of Process Analysis and Control of Sichuan Universities, Yibin 644000, Sichuan, China
| | - Xianyan Li
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China
| | - Peng Lin
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China
| | - Qin Zhang
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China.
| | - Yuan Gao
- Faculty of Materials and Chemical Engineering, Yibin University, Yibin 644000, China.
| | - Jian Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Evaluation and Research Center of Daodi Herbs of Jiangxi Province, Ganjiang New District 330000, China.
| |
Collapse
|
32
|
R P, Ramasubramanian A, Ramani P, Doble M. Identification of Novel Potential Herbal Drug Targets against Beta-Catenin in the Treatment of Oral Squamous Cell Carcinoma. Asian Pac J Cancer Prev 2024; 25:4181-4188. [PMID: 39733408 PMCID: PMC12008329 DOI: 10.31557/apjcp.2024.25.12.4181] [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: 05/07/2024] [Indexed: 12/31/2024] Open
Abstract
OBJECTIVE The study aims to identify potential pharmacophore models for targeting beta-catenin, a crucial protein involved in the development of oral squamous cell carcinoma (OSCC), using a combination of herbal compounds and computational approaches. METHODS Five natural compounds namely Quercetin, Lycopene, Ovatodiolide, Karsil, and Delphinidin were selected based on their reported activity against beta-catenin. Ligand characteristics were analyzed using SwissADME to evaluate drug-likeness, lipophilicity (logP), and bioavailability. The three-dimensional structure of beta-catenin was retrieved from the Protein Data Bank (PDB). Pharmacophore modeling was performed using Pharmagist software, followed by molecular docking using Swissdock to assess binding interactions and energies. RESULTS Out of thousands of pharmacophore hits generated, 23 were selected based on drug-likeness properties. Molecular docking revealed that ZINC94512303, derived from the combination of the selected herbal compounds, exhibited the highest binding energy of -8.91 kcal/mol with beta-catenin, outperforming individual herbal compounds. This compound adhered to all drug-likeness rules and demonstrated optimal pharmacokinetic properties. CONCLUSION The identified pharmacophore, ZINC94512303, shows promise as a therapeutic agent targeting beta-catenin in OSCC. The combination of computational drug design with herbal compounds offers a novel approach to enhance the efficacy of cancer treatment. Further pharmacokinetic and pharmacodynamic studies, along with in vitro and clinical evaluations, are recommended to validate the therapeutic potential of this compound.
Collapse
Affiliation(s)
- Priyadharshini R
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Abilasha Ramasubramanian
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Pratibha Ramani
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Mukesh Doble
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
33
|
Peng X, Cai X, Tang J, Ge J, Chen G. Rescuing and utilizing anticancer Nothapodytes species: Integrated studies from plant resources to natural medicines. Clin Transl Med 2024; 14:e70110. [PMID: 39587903 PMCID: PMC11589384 DOI: 10.1002/ctm2.70110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Affiliation(s)
- Xingrong Peng
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of BotanyChinese Academy of ScienceKunmingChina
| | - Xianghai Cai
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of BotanyChinese Academy of ScienceKunmingChina
| | - Jia Tang
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
| | - Jia Ge
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- Yunnan Key Laboratory for Integrative Conservation of Plant Species with Extremely Small PopulationsKunmingChina
| | - Gao Chen
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of BotanyChinese Academy of SciencesKunmingChina
- Yunnan Key Laboratory for Integrative Conservation of Plant Species with Extremely Small PopulationsKunmingChina
| |
Collapse
|
34
|
Jojić AA, Liga S, Uţu D, Ruse G, Suciu L, Motoc A, Şoica CM, Tchiakpe-Antal DS. Beyond Essential Oils: Diterpenes, Lignans, and Biflavonoids from Juniperus communis L. as a Source of Multi-Target Lead Compounds. PLANTS (BASEL, SWITZERLAND) 2024; 13:3233. [PMID: 39599442 PMCID: PMC11598787 DOI: 10.3390/plants13223233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Common Juniper (Juniperus communis L.) is a gymnosperm that stands out through its fleshy, spherical female cones, often termed simply "berries". The cone berries and various vegetative parts (leaves, twigs and even roots) are used in traditional phytotherapy, based on the beneficial effects exerted by a variety of secondary metabolites. While the volatile compounds of Juniperus communis are known for their aromatic properties and have been well-researched for their antimicrobial effects, this review shifts focus to non-volatile secondary metabolites-specifically diterpenes, lignans, and biflavonoids. These compounds are of significant biomedical interest due to their notable pharmacological activities, including antioxidant, anti-inflammatory, antimicrobial, and anticancer effects. The aim of this review is to offer an up-to-date account of chemical composition of Juniperus communis and related species, with a primary emphasis on the bioactivities of diterpenes, lignans, and biflavonoids. By examining recent preclinical and clinical data, this work assesses the therapeutic potential of these metabolites and their mechanisms of action, underscoring their value in developing new therapeutic options. Additionally, this review addresses the pharmacological efficacy and possible therapeutic applications of Juniperus communis in treating various human diseases, thus supporting its potential role in evidence-based phytotherapy.
Collapse
Affiliation(s)
- Alina Arabela Jojić
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.A.J.); (S.L.); (L.S.); (C.M.Ş.)
- Research Center for Pharmacotoxicologic Evaluations (FARMTOX), “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Sergio Liga
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.A.J.); (S.L.); (L.S.); (C.M.Ş.)
- Department of Applied Chemistry and Engineering of Organic and Natural Compounds, Faculty of Chemical Engineering, Biotechnologies and Environmental Protection, Politehnica University Timisoara, 6 Vasile Parvan, 300223 Timisoara, Romania
| | - Diana Uţu
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.A.J.); (S.L.); (L.S.); (C.M.Ş.)
| | - Graţiana Ruse
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Liana Suciu
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.A.J.); (S.L.); (L.S.); (C.M.Ş.)
| | - Andrei Motoc
- Department of Anatomy-Embryology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Codruța Marinela Şoica
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (A.A.J.); (S.L.); (L.S.); (C.M.Ş.)
- Research Center for Pharmacotoxicologic Evaluations (FARMTOX), “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Diana-Simona Tchiakpe-Antal
- Research Center for Pharmacotoxicologic Evaluations (FARMTOX), “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| |
Collapse
|
35
|
Li X, Xu J, Yan L, Tang S, Zhang Y, Shi M, Liu P. Targeting Disulfidptosis with Potentially Bioactive Natural Products in Metabolic Cancer Therapy. Metabolites 2024; 14:604. [PMID: 39590840 PMCID: PMC11596291 DOI: 10.3390/metabo14110604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Metabolic cancers are defined by metabolic reprogramming. Although this reprograming drives rapid tumour growth and invasion, it also reveals specific metabolic vulnerabilities that can be therapeutically exploited in cancer therapy. A novel form of programmed cell death, known as disulfidptosis, was identified last year; tumour cells with high SLC7A11 expression undergo disulfidptosis when deprived of glucose. Natural products have attracted increasing attention and have shown potential to treat metabolic cancers through diverse mechanisms. METHODS We systematically searched electronic databases involving PubMed, Web of Science, Gooale Scholar. To ensue comprehensive exploration, keywords including metabolic reprogramming, metabolic cancer, disulfidptosis, natural products and some other words were employed. RESULTS In this review, we focus on the shared characteristics and metabolic vulnerabilities of metabolic cancers. Additionally, we discuss the molecular mechanisms underlying disulfidptosis and highlight key regulatory genes. Furthermore, we predict bioactive natural products that target disulfidptosis-related genes, offering new perspectives for anticancer strategies through the modulation of disulfidptosis. CONCLUSIONS By summarizing current research progress, this review mainly analyzed the potential mechanisms of natural products in the treatment of metabolic cancer.
Collapse
Affiliation(s)
- Xinyan Li
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
| | - Mengjiao Shi
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China; (J.X.); (L.Y.); (S.T.); (Y.Z.)
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education of China, Xi’an 710061, China
| |
Collapse
|
36
|
Chipón C, Riffo P, Ojeda L, Salas M, Burgos RA, Ehrenfeld P, López-Muñoz R, Zambrano A. Impact of Nordihydroguaiaretic Acid on Proliferation, Energy Metabolism, and Chemosensitization in Non-Small-Cell Lung Cancer (NSCLC) Cell Lines. Int J Mol Sci 2024; 25:11601. [PMID: 39519155 PMCID: PMC11546251 DOI: 10.3390/ijms252111601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Lung cancer (LC) is the leading cause of cancer death worldwide. LC can be classified into small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC), with the last subtype accounting for approximately 85% of all diagnosed lung cancer cases. Despite the existence of different types of treatment for this disease, the development of resistance to therapies and tumor recurrence in patients have maintained the need to find new therapeutic options to combat this pathology, where natural products stand out as an attractive source for this search. Nordihydroguaiaretic acid (NDGA) is the main metabolite extracted from the Larrea tridentata plant and has been shown to have different biological activities, including anticancer activity. In this study, H1975, H1299, and A549 cell lines were treated with NDGA, and its effect on cell viability, proliferation, and metabolism was evaluated using a resazurin reduction assay, incorporation of BrdU, and ki-67 gene expression and glucose uptake measurement, respectively. In addition, the combination of NDGA with clinical chemotherapeutics was investigated using an MTT assay and Combenefit software (version 2.02). The results showed that NDGA decreases the viability and proliferation of NSCLC cells and differentially modulates the expression of genes associated with different metabolic pathways. For example, the LDH gene expression decreased in all cell lines analyzed. However, GLUT3 gene expression increased after 24 h of treatment. The expression of the HIF-1 gene decreased early in the H1299 and A549 cell lines. In addition, the combination of NDGA with three chemotherapeutics (carboplatin, gemcitabine, and taxol) shows a synergic pattern in the decrease of cell viability on the H1299 cell line. In summary, this research provides new evidence about the role of NDGA in lung cancer. Interestingly, using NDGA to enhance the anticancer activity of antitumoral drugs could be an improved therapeutic resource against lung cancer.
Collapse
Affiliation(s)
- Carina Chipón
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.C.); (P.R.); (L.O.); (M.S.)
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia 5090000, Chile;
| | - Paula Riffo
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.C.); (P.R.); (L.O.); (M.S.)
| | - Loreto Ojeda
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.C.); (P.R.); (L.O.); (M.S.)
| | - Mónica Salas
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.C.); (P.R.); (L.O.); (M.S.)
| | - Rafael A. Burgos
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia 5090000, Chile;
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile;
| | - Pamela Ehrenfeld
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile;
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Rodrigo López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia 5090000, Chile;
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile;
| | - Angara Zambrano
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.C.); (P.R.); (L.O.); (M.S.)
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile;
| |
Collapse
|
37
|
Yue Z, Xu Y, Cai M, Fan X, Pan H, Zhang D, Zhang Q. Floral Elegance Meets Medicinal Marvels: Traditional Uses, Phytochemistry, and Pharmacology of the Genus Lagerstroemia L. PLANTS (BASEL, SWITZERLAND) 2024; 13:3016. [PMID: 39519935 PMCID: PMC11548200 DOI: 10.3390/plants13213016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
The genus Lagerstroemia L. (Lythraceae), known for its exquisite flowers and prolonged flowering period, is commonly employed in traditional medicinal systems across Asian countries, where it has always been consumed as tea or employed to address ailments such as diabetes, urinary disorders, coughs, fevers, inflammation, pain, and anesthesia. Its diverse uses may be attributed to its rich active ingredients. Currently, at least 364 biological compounds have been identified from Lagerstroemia extracts, encompassing various types such as terpenes, flavonoids, phenolic acids, alkaloids, and phenylpropanoids. Extensive in vitro and in vivo experiments have examined the pharmacological activities of different extracts, revealing their potential in various domains, including but not limited to antidiabetic, anti-obesity, antitumor, antimicrobial, antioxidant, anti-inflammatory, analgesic, and hepatoprotective effects. Additionally, 20 core components have been proven to be associated with antidiabetic and hypoglycemic effects of Lagerstroemia. Overall, Lagerstroemia exhibit substantial medicinal potential, and the alignment between its traditional applications and contemporary pharmacological findings present promising opportunities for further investigation, particularly in food and health products, drug development, herbal teas, and cosmetics. However, evidence-based pharmacological research has largely been confined to in vitro screening and animal model, lacking clinical trials and bioactive compound isolations. Consequently, future endeavors should adopt a more holistic approach.
Collapse
Affiliation(s)
- Ziwei Yue
- Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, National Engineering Research Center for Floriculture, School of Landscape Architecture, Beijing Forestry University, Beijing 100083, China; (Z.Y.); (Y.X.); (H.P.); (Q.Z.)
| | - Yan Xu
- Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, National Engineering Research Center for Floriculture, School of Landscape Architecture, Beijing Forestry University, Beijing 100083, China; (Z.Y.); (Y.X.); (H.P.); (Q.Z.)
| | - Ming Cai
- Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, National Engineering Research Center for Floriculture, School of Landscape Architecture, Beijing Forestry University, Beijing 100083, China; (Z.Y.); (Y.X.); (H.P.); (Q.Z.)
| | - Xiaohui Fan
- Luoyang Landscape and Greening Center, Luoyang 471000, China;
| | - Huitang Pan
- Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, National Engineering Research Center for Floriculture, School of Landscape Architecture, Beijing Forestry University, Beijing 100083, China; (Z.Y.); (Y.X.); (H.P.); (Q.Z.)
| | - Donglin Zhang
- Department of Horticulture, University of Georgia, Athens, GA 30602, USA;
| | - Qixiang Zhang
- Beijing Key Laboratory of Ornamental Plants Germplasm Innovation & Molecular Breeding, National Engineering Research Center for Floriculture, School of Landscape Architecture, Beijing Forestry University, Beijing 100083, China; (Z.Y.); (Y.X.); (H.P.); (Q.Z.)
| |
Collapse
|
38
|
Chaturvedi S, Sarethy IP. GC-MS-based metabolite fingerprinting reveals the presence of novel anticancer compounds in the microcolonial fungus Aureobasidium sp. TD-062 from the under-explored Thar Desert. Nat Prod Res 2024:1-7. [PMID: 39440598 DOI: 10.1080/14786419.2024.2418450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Aureobasidium sp., strain TD-062, a micro colonial black yeast, was obtained as part of a screening program from the Thar Desert of India, which has been inadequately explored for novel microorganisms/bioactive metabolites. The anticancer activity of aqueous and organic solvent extracts of culture supernatant of TD-062 was investigated against A-375 myeloma and MCF-7 breast cancer cell lines. Following column chromatography, bioactive fractions were subjected to GC-MS analysis in two cycles. The GC-MS metabolite fingerprinting revealed 20 compounds belonging to various chemical groups. Further, based on observed and calculated mass ion spectra, retention time, and retention indices, many compounds could be considered novel. The second purification cycle resulted in three compounds- ATD-1, ATD-2 and ATD-3 with different calculated retention indices from that of the nearest matching compounds, squalene and tocopherol. In silico prediction study of their ADMET profiles, suggests that these new compounds have a suitable contour for use as safe anticancer drugs, without the toxicity normally associated with anticancer compounds.
Collapse
Affiliation(s)
- Swapnil Chaturvedi
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Indira P Sarethy
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
39
|
Li Y, Shen Q, Feng L, Zhang C, Jiang X, Liu F, Pang B. A nanoscale natural drug delivery system for targeted drug delivery against ovarian cancer: action mechanism, application enlightenment and future potential. Front Immunol 2024; 15:1427573. [PMID: 39464892 PMCID: PMC11502327 DOI: 10.3389/fimmu.2024.1427573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/22/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological malignancies in the world and is the leading cause of cancer-related death in women. The complexity and difficult-to-treat nature of OC pose a huge challenge to the treatment of the disease, Therefore, it is critical to find green and sustainable drug treatment options. Natural drugs have wide sources, many targets, and high safety, and are currently recognized as ideal drugs for tumor treatment, has previously been found to have a good effect on controlling tumor progression and reducing the burden of metastasis. However, its clinical transformation is often hindered by structural stability, bioavailability, and bioactivity. Emerging technologies for the treatment of OC, such as photodynamic therapy, immunotherapy, targeted therapy, gene therapy, molecular therapy, and nanotherapy, are developing rapidly, particularly, nanotechnology can play a bridging role between different therapies, synergistically drive the complementary role of differentiated treatment schemes, and has a wide range of clinical application prospects. In this review, nanoscale natural drug delivery systems (NNDDS) for targeted drug delivery against OC were extensively explored. We reviewed the mechanism of action of natural drugs against OC, reviewed the morphological composition and delivery potential of drug nanocarriers based on the application of nanotechnology in the treatment of OC, and discussed the limitations of current NNDDS research. After elucidating these problems, it will provide a theoretical basis for future exploration of novel NNDDS for anti-OC therapy.
Collapse
Affiliation(s)
- Yi Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Shen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Feng
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanlong Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Pang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
40
|
Hou J, Cao R, Wang S, Ma J, Xu J, Guo Y. Bucidarasin A suppresses the proliferation and metastasis of HCC by targeting the FAK and STAT3 pathways. Chem Biol Interact 2024; 402:111191. [PMID: 39121898 DOI: 10.1016/j.cbi.2024.111191] [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: 04/20/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Hepatocellular carcinoma (HCC) is a significant global health concern, with high rates of morbidity and mortality. Bucidarasin A, a natural diterpenoid, has been shown to exert notable cytotoxic effects across a range of tumor cell lines. However, the underlying mechanisms responsible for this cytotoxicity remain unclear. In this study, we sought to elucidate the antitumor mechanisms of bucidarasin A, a natural diterpenoid derived from Casearia graveolens, with a particular focus on its effects on HCC. Furthermore, we employed surface plasmon resonance (SPR), molecular docking, and cellular thermal shift assay (CETSA) to gain further insight into the target protein of bucidarasin A. Our findings revealed that bucidarasin A exhibited pronounced cytotoxicity towards HepG2 cells. In vitro analysis indicated that bucidarasin A interrupted the cell cycle at the S phase and inhibited the proliferation and metastasis of HepG2 cells by modulating the FAK and STAT3 signaling pathways. Moreover, in vivo studies demonstrated that bucidarasin A not only exhibited antitumor effects but also impeded neovascularization, a finding that was corroborated by SPR interactions between vascular endothelial growth factor (VEGF) and bucidarasin A. This research substantiated that bucidarasin A, a clerodane diterpenoid, held promise as a therapeutic candidate against HCC, showcasing substantial antitumor efficacy both in vitro and in vivo through direct targeting of the STAT3 and FAK signaling pathways.
Collapse
Affiliation(s)
- Jiantong Hou
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, PR China
| | - Ruyu Cao
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, PR China
| | - Sibei Wang
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, PR China
| | - Jun Ma
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, PR China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, PR China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China.
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemistry Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, PR China.
| |
Collapse
|
41
|
Wang XY, Wang YJ, Hou ZL, Guo BW, Wang RQ, Liu Q, Yao GD, Song SJ. Ingenane-type diterpenoids inhibit non-small cell lung cancer cells by regulating SRC/PI3K/Akt pathway. Nat Prod Res 2024; 38:3460-3465. [PMID: 37615118 DOI: 10.1080/14786419.2023.2247536] [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: 04/15/2023] [Revised: 07/22/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023]
Abstract
Ingenane-type diterpenoids (ITDs) are distinct components of plants belonging to the genus Euphorbia. These compounds have significant cytotoxic effects on non-small cell lung cancer (NSCLC) cells. However, the underlying molecular mechanism has yet to be reported. To explore the mechanism of the anticancer effect of ITDs, we carried out a network pharmacology prediction study. PPI network suggested that SRC and PI3K had high levels of interaction. In addition, KEGG analysis revealed that these common targets were significantly enriched in the PI3K/Akt signalling pathway. 13-oxyingenol-dodecanoate (13OD) was used for validation after the biological evaluation of some ITDs against NSCLC cells. It demonstrated that 13OD could significantly inhibit the growth of NSCLC cells by inducing apoptosis. The results from molecular docking and Western blotting showed that 13OD interacted with SRC and PI3K and down-regulated the SRC/PI3K/Akt signalling pathway in NSCLC cells. This study provided the underlying mechanism of ITDs against NSCLC.
Collapse
Affiliation(s)
- Xin-Ye Wang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Yu-Jue Wang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zi-Lin Hou
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Bo-Wen Guo
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Ru-Qi Wang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
42
|
Rahman AU, Panichayupakaranant P. Exploring the diverse biological activities of Garcinia cowa: Implications for future cancer chemotherapy and beyond. FOOD BIOSCI 2024; 61:104525. [DOI: 10.1016/j.fbio.2024.104525] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
43
|
Hasan‐Abad A, Atapour A, Sobhani‐Nasab A, Motedayyen H, ArefNezhad R. Plant-Based Anticancer Compounds With a Focus on Breast Cancer. Cancer Rep (Hoboken) 2024; 7:e70012. [PMID: 39453820 PMCID: PMC11506041 DOI: 10.1002/cnr2.70012] [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: 10/18/2023] [Revised: 07/11/2024] [Accepted: 08/11/2024] [Indexed: 10/27/2024] Open
Abstract
Breast cancer is a common form of cancer among women characterized by the growth of malignant cells in the breast tissue. The most common treatments for this condition include chemotherapy, surgical intervention, radiation therapy, hormone therapy, and biological therapy. The primary issues associated with chemotherapy and radiation therapy are their adverse events and significant financial burden among patients in underdeveloped countries. This highlights the need to explore and develop superior therapeutic options that are less detrimental and more economically efficient. Plants provide an abundant supply of innovative compounds and present a promising new avenue for investigating cancer. Plants and their derivations are undergoing a revolution due to their reduced toxicity, expediency, cost-effectiveness, safety, and simplicity in comparison to conventional treatment methods. Natural products are considered promising candidates for the development of anticancer drugs, due perhaps to the diverse pleiotropic effects on target events. The effects of plant-derived products are limited to cancer cells while leaving healthy cells unaffected. Identification of compounds with strong anticancer properties and development of plant-based medications for cancer treatment might be crucial steps in breast cancer therapy. Although bioactive compounds have potent anticancer properties, they also have drawbacks that need to be resolved before their application in clinical trials and improved for the approved drugs. This study aims to give comprehensive information on known anticancer compounds, including their sources and molecular mechanisms of actions, along with opportunities and challenges in plant-based anticancer therapies.
Collapse
Affiliation(s)
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Ali Sobhani‐Nasab
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
| | - Hossein Motedayyen
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
| | - Reza ArefNezhad
- Department of Anatomy, School of MedicineShiraz University of Medical SciencesShirazIran
| |
Collapse
|
44
|
Shahpouri P, Mehralitabar H, Kheirabadi M, Kazemi Noureini S. Potential suppression of multidrug-resistance-associated protein 1 by coumarin derivatives: an insight from molecular docking and MD simulation studies. J Biomol Struct Dyn 2024; 42:9184-9200. [PMID: 37667877 DOI: 10.1080/07391102.2023.2250456] [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: 04/24/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023]
Abstract
Human MRP1 protein plays a vital role in cancer multidrug resistance. Coumarins show promising pharmacological properties. Virtual screening, ADMET, molecular docking and molecular dynamics (MD) simulations were utilized as pharmacoinformatic tools to identify potential MRP1 inhibitors among coumarin derivatives. Using in silico ADMET, 50 hits were further investigated for their selectivity toward the nucleotide-binding domains (NBDs) of MRP1 using molecular docking. Accordingly, coumarin, its symmetrical ketone derivative Lig. No. 4, and Reversan were candidates for focused docking study with the NBDs domains compared with ATP. The result indicates that Lig. No. 4, with the best binding score, interacts with NBDs via hydrogen bonds with residues: GLN713, LYS684, GLY683, CYS682 in NBD1, and GLY1432, GLY771, SER769 and GLN1374 in NBD2, which mostly overlap with ATP binding residues. Moreover, doxorubicin (Doxo) was docked to the transmembrane domains (TMDs) active site of MRP1. Doxo interaction with TMDs was subjected to MD simulation in the NBDs free and occupied with Lig. No. 4 states. The results showed that Doxo interacts more strongly with TMD residues in inward facing feature of TMDs helices. However, when Lig. No. 4 exists in NBDs, Doxo interactions are different, and TMD helices show more outward-facing conformation. This result may suggest a partial competitive inhibition mechanism for the Lig. No. 4 on MRP1 compared with ATP. So, it may inhibit active complex formation by interfering with ATP entrance to NBDs and locking MRP1 conformation in outward-facing mode. This study suggests a valuable coumarin derivative that can be further investigated for potent MRP1 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Parisa Shahpouri
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Havva Mehralitabar
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mitra Kheirabadi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | | |
Collapse
|
45
|
Sharma V, Sinha ES, Singh J. Investigation of In Vitro Anti-cancer and Apoptotic Potential of Onion-Derived Nanovesicles Against Prostate and Cervical Cancer Cell Lines. Appl Biochem Biotechnol 2024; 196:6957-6973. [PMID: 38441782 DOI: 10.1007/s12010-024-04872-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 11/21/2024]
Abstract
Plant-derived compounds have recently garnered significant interest in the field of medicine due to their rich repertoire of phytochemicals, which holds promise for exploring novel therapies to treat cancer. This study embarks on the first-time investigation of the anti-cancerous effect of onion-derived nanovesicles (ODNVs). ODNVs were isolated employing differential centrifugation followed by ultracentrifugation and subsequent characterization using dynamic light scattering (DLS), field emission scanning electron microscopy (FESEM), and Fourier transform infrared spectroscopy (FTIR). Furthermore, we delineated the anti-cancerous effect of ODNVs on two cancer cell line models HeLa (cervical cancer) and PC-3 (prostate cancer) using MTT assay, DAPI-based DNA damage using immunofluorescence microscopy, colony formation assay, migration assay, cell cycle analysis, and evaluation of apoptosis using flow cytometry and western blotting. The findings revealed dose- and time-dependent anti-proliferative effects of ODNVs on both HeLa and PC3 cell lines, accompanied by selective cytotoxicity against cancer cells. Additional results highlighted that ODNVs prevented colony growth and induced S-phase cell cycle arrest. Apoptosis induction was evaluated through alterations in nuclear morphology and the number of apoptotic cells, which increased significantly after ODNV treatment in both cancer cell lines. Furthermore, annexin V/PI staining evaluation of apoptotic cells by flow cytometry demonstrated that ODNV treatment significantly increased the number of apoptotic cells in both PC-3 and HeLa cells. Finally, Western blot analysis indicated changes in apoptosis-related proteins including bcl-2, bax, and caspase-3, emphasizing that the anti-cancerous effect of ODNVs is attributed to the induction of apoptosis and suggests the unexplored anti-cancerous potential of ODNVs.
Collapse
Affiliation(s)
- Vinayak Sharma
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Eshu Singhal Sinha
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Jagtar Singh
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
46
|
Yousef MM, Abdelhafez OH, Alsenani F, Al Mouslem AK, Hisham M, El Zawily A, P Glaeser S, Kämpfer P, Abdelmohsen UR, El-Katatnye MH. Anti-infective potential of the endophytic fungus Aspergillus sp. associated with Aptenia cordifolia root supported by metabolomics analysis and docking studies. Nat Prod Res 2024; 38:3421-3426. [PMID: 37590004 DOI: 10.1080/14786419.2023.2245114] [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: 12/27/2022] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Endophytic fungi are known to be a rich source for anti-infective drugs. In this study, Aptenia cordifolia associated endophytic fungi were explored for the first time. Seven isolates were identified morphologically followed by screening of these fungi by plug diffusion assay which revealed their potential activity against Staphylococcus aureus (ATCC 9144), Bacillus cereus (ATCC 14579), Serratia marcescens (ATCC 14756), Fusarium oxysporum (ATCC 48112), and Aspergillus flavus (ATCC 22546). Additionally, the crude ethyl acetate extract of the most potent three isolates in plug diffusion assay showed that Aspergillus sp. ACEFR2 was the most potent as anti-infective in disc diffusion assay; Accordingly, Aspergillus sp. ACEFR2 was investigated using phylogenetic analysis and LC-HR-ESI-MS. The phylogenetic analysis placed the strain into the Aspergillus section Niger close related to few species including A. niger. Whereas the metabolomic profiling revealed the presence of diverse pool of metabolites. Furthermore, in silico molecular docking study was carried out to predict which compounds most likely responsible for the anti-infective activity.
Collapse
Affiliation(s)
- Miram Magdy Yousef
- Department of Botany and Microbiology, Faculty of Science, Minia University, Minia, Egypt
| | - Omnia Hesham Abdelhafez
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia City, Minia, Egypt
| | - Faisl Alsenani
- Department of Pharmacognosy, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulaziz K Al Mouslem
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Mohamed Hisham
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New-Minia, Egypt
| | - Amr El Zawily
- Department of Biology, University of IA, Iowa City, IA, USA
- Department of Plant and Microbiology, Faculty of Science, Damanhour University, Egypt
| | - Stefanie P Glaeser
- Institute of Applied Microbiology, Justus-Liebig University Gießen, Gießen, Germany
| | - Peter Kämpfer
- Institute of Applied Microbiology, Justus-Liebig University Gießen, Gießen, Germany
| | | | | |
Collapse
|
47
|
Adu-Amankwaah F, Februarie C, Nyambo K, Maarman G, Tshililo N, Mabasa L, Mavumengwana V, Baatjies L. Cytotoxic properties, glycolytic effects and high-resolution respirometry mitochondrial activities of Eriocephalus racemosus against MDA-MB 231 triple-negative breast cancer. BMC Complement Med Ther 2024; 24:332. [PMID: 39256791 PMCID: PMC11389270 DOI: 10.1186/s12906-024-04615-x] [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: 05/17/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) represents a significant global health crisis due to its resistance to conventional therapies and lack of specific molecular targets. This study explored the potential of Eriocephalus racemosus (E. racemosus) as an alternative treatment for TNBC. The cytotoxic properties and high-resolution respirometry mitochondrial activities of E. racemosus against the MDA-MB 231 TNBC cell line were evaluated. METHODS Hexane solvent and bioactive fraction extractions of E. racemosus were performed, while mass spectrometry-based metabolite profiling was used to identify the phytochemical constituents of the extracts. The extracts were further tested against MDA-MB 231 TNBC cells to determine their cytotoxicity. The mode of cell death was determined using flow cytometry. The activities of caspases 3, 8, and 9 were assessed using a multiplex activity assay kit. Glycolytic activity and High-resolution respirometry measurements of mitochondrial function in the MDA-MB 231 cell line were conducted using the Seahorse XFp and Oroboros O2K. RESULTS Metabolite profiling of E. racemosus plant crude extracts identified the presence of coumarins, flavonoids, sesquiterpenoids, triterpenoids, and unknown compounds. The extracts demonstrated promising cytotoxic activities, with a half maximal inhibitory concentration (IC50) of 12.84 µg/mL for the crude hexane extract and 15.49 µg/mL for the bioactive fraction. Further, the crude hexane and bioactive fraction extracts induced apoptosis in the MDA-MB-231 TNBC cells, like the reference drug cisplatin (17.44%, 17.26% and 20.25%, respectively) compared to untreated cells. Caspase 3 activities confirmed the induction of apoptosis in both cisplatin and the plant crude extracts, while caspase 8 and 9 activities confirmed the activation of both the intrinsic and extrinsic apoptosis pathways. Increased levels of glycolytic activity were observed in the hexane crude extract. High-resolution respiratory measurements showed elevated mitochondrial activities in all mitochondrial states except for complex-IV activity. CONCLUSION These findings support further exploration of E. racemosus as a potential therapeutic agent for TNBC, offering a promising avenue for the development of targeted treatments with minimal adverse effects.
Collapse
Affiliation(s)
- Francis Adu-Amankwaah
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Candice Februarie
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Science, CARMA: Centre for Cardio-Metabolic Research in Africa, Stellenbosch University, Cape Town, 8000, South Africa
| | - Kudakwashe Nyambo
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerald Maarman
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Science, CARMA: Centre for Cardio-Metabolic Research in Africa, Stellenbosch University, Cape Town, 8000, South Africa
| | - Ndivhuwo Tshililo
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Vuyo Mavumengwana
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Lucinda Baatjies
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa.
| |
Collapse
|
48
|
Zhang X, Lou X, Qiao H, Jiang Z, Sun H, Shi X, He Z, Sun J, Sun M. Supramolecular self-sensitized dual-drug nanoassemblies potentiating chemo-photodynamic therapy for effective cancer treatment. Int J Pharm 2024; 662:124496. [PMID: 39033943 DOI: 10.1016/j.ijpharm.2024.124496] [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: 02/29/2024] [Revised: 06/30/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Chemo-photodynamic synergistic therapy (CPST) holds tremendous promise for treating cancers. Unfortunately, existing CPST applications suffer from complex synthetic procedures, low drug co-loading efficiency, and carrier-related toxicity. To address these issues, we have developed a supramolecular carrier-free self-sensitized nanoassemblies by co-assembling podophyllotoxin (PTOX) and chlorin e6 (Ce6) to enhance CPST efficiency against tumors. The nanoassemblies show stable co-assembly performance in simulative vivo neural environment (∼150 nm), with high co-loading ability for PTOX (72.2 wt%) and Ce6 (27.8 wt%). In vivo, the nanoassemblies demonstrate a remarkable ability to accumulate at tumor sites by leveraging the enhanced permeability and retention (EPR) effect. The disintegration of nanoassemblies following photosensitizer bioactivation triggered by the acidic tumor environment effectively resolves the challenge of aggregation-caused quenching (ACQ) effect. Upon exposure to external light stimulation, the disintegrated nanoassemblies not only illuminate cancer cells synergistically but also exert a more potent antitumor effect when compared with PTOX and Ce6 administered alone. This self-sensitized strategy represents a significant step forward in CPST, offering a unique co-delivery paradigm for clinic cancer treatment.
Collapse
Affiliation(s)
- Xu Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Xinyu Lou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Han Qiao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Zhouyu Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Hang Sun
- Hong Kong Education University, Hong Kong SAR, 999077, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
49
|
Santos W, Katchborian-Neto A, Viana GS, Ferreira MS, Martins LC, Vale TC, Murgu M, Dias DF, Soares MG, Chagas-Paula DA, Paula ACC. Metabolomics Unveils Disrupted Pathways in Parkinson's Disease: Toward Biomarker-Based Diagnosis. ACS Chem Neurosci 2024; 15:3168-3180. [PMID: 39177430 PMCID: PMC11378289 DOI: 10.1021/acschemneuro.4c00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by diverse symptoms, where accurate diagnosis remains challenging. Traditional clinical observation methods often result in misdiagnosis, highlighting the need for biomarker-based diagnostic approaches. This study utilizes ultraperformance liquid chromatography coupled to an electrospray ionization source and quadrupole time-of-flight untargeted metabolomics combined with biochemometrics to identify novel serum biomarkers for PD. Analyzing a Brazilian cohort of serum samples from 39 PD patients and 15 healthy controls, we identified 15 metabolites significantly associated with PD, with 11 reported as potential biomarkers for the first time. Key disrupted metabolic pathways include caffeine metabolism, arachidonic acid metabolism, and primary bile acid biosynthesis. Our machine learning model demonstrated high accuracy, with the Rotation Forest boosting model achieving 94.1% accuracy in distinguishing PD patients from controls. It is based on three new PD biomarkers (downregulated: 1-lyso-2-arachidonoyl-phosphatidate and hypoxanthine and upregulated: ferulic acid) and surpasses the general 80% diagnostic accuracy obtained from initial clinical evaluations conducted by specialists. Besides, this machine learning model based on a decision tree allowed for visual and easy interpretability of affected metabolites in PD patients. These findings could improve the detection and monitoring of PD, paving the way for more precise diagnostics and therapeutic interventions. Our research emphasizes the critical role of metabolomics and machine learning in advancing our understanding of the chemical profile of neurodegenerative diseases.
Collapse
Affiliation(s)
- Wanderleya
T. Santos
- Department
of Pharmaceutical Sciences, Federal University
of Juiz de Fora, Juiz de
Fora 36036-900, Brazil
| | | | - Gabriel S. Viana
- Chemistry
Institute, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Miller S. Ferreira
- Chemistry
Institute, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Luiza C. Martins
- Department
of Pharmaceutical Sciences, Federal University
of Juiz de Fora, Juiz de
Fora 36036-900, Brazil
- Faculty
of Medicine, Federal University of Juiz
de Fora, Juiz de
Fora 36036-900, Brazil
| | - Thiago C. Vale
- Faculty
of Medicine, Federal University of Juiz
de Fora, Juiz de
Fora 36036-900, Brazil
| | | | - Danielle F. Dias
- Chemistry
Institute, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Marisi G. Soares
- Chemistry
Institute, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | | | - Ana C. C. Paula
- Department
of Pharmaceutical Sciences, Federal University
of Juiz de Fora, Juiz de
Fora 36036-900, Brazil
| |
Collapse
|
50
|
Gulia S, Chandra P, Das A. Combating anoikis resistance: bioactive compounds transforming prostate cancer therapy. Anticancer Drugs 2024; 35:687-697. [PMID: 38743565 DOI: 10.1097/cad.0000000000001616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The study aims to discuss the challenges associated with treating prostate cancer (PCa), which is known for its complexity and drug resistance. It attempts to find differentially expressed genes (DEGs), such as those linked to anoikis resistance and circulating tumor cells, in PCa samples. This study involves analyzing the functional roles of these DEGs using gene enrichment analysis, and then screening of 102 bioactive compounds to identify a combination that can control the expression of the identified DEGs. In this study, 53 DEGs were identified from PCa samples including anoikis-resistant PCa cells and circulating tumor cells in PCa. Gene enrichment analysis with regards to functional enrichment of DEGs was performed. An inclusive screening process was carried out among 102 bioactive compounds to identify a combination capable of affecting and regulating the expression of selected DEGs. Eventually, gastrodin, nitidine chloride, chenodeoxycholic acid, and bilobalide were selected, as their combination demonstrated ability to modulate expression of 50 out of the 53 genes targeted. The subsequent analysis focused on investigating the biological pathways and processes influenced by this combination. The findings revealed a multifaceted and multidimensional approach to tumor regression. The combination of bioactive compounds exhibited effects on various genes including those related to production of inflammatory cytokines, cell proliferation, autophagy, apoptosis, angiogenesis, and metastasis. The current study has made a valuable contribution to the development of a combination of bioactive natural compounds that can significantly impede the development of treatment resistance in prostate tumor while countering the tumors' evasion of the immune system. The implications of this study are highly significant as it suggests the creation of an enhanced immunotherapeutic, natural therapeutic concoction with combinatorial potential.
Collapse
Affiliation(s)
- Shweta Gulia
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | | | | |
Collapse
|