1
|
Aqib M, Khatoon S, Ali M, Sajid S, Assiri MA, Ahamad S, Saquib M, Hussain MK. Exploring the anticancer potential and mechanisms of action of natural coumarins and isocoumarins. Eur J Med Chem 2025; 282:117088. [PMID: 39608206 DOI: 10.1016/j.ejmech.2024.117088] [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: 10/21/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
Natural coumarins and isocoumarins show significant therapeutic potential against cancer in preclinical studies by targeting multiple pathways and processes. These compounds influence several critical cellular processes, such as apoptosis, autophagy, and cell cycle regulation, which are pivotal in cancer development and progression. Their capability to target multiple signalling pathways provides a strategic advantage over single-target therapies, which are often limited by drug resistance. Notably, coumarins have the potential to inhibit angiogenesis, the process through which tumours develop new blood vessels, thereby potentially restricting tumour growth and metastasis. Additionally, coumarins may enhance anticancer effects by modulating immune responses and reducing inflammation, thus offering a dual approach to combating cancer. They also show promise in addressing multidrug resistance, a significant challenge in cancer therapy, by targeting drug efflux proteins and potentially improving the efficacy of existing treatments. While preclinical studies are promising, further research is required to elucidate the pharmacokinetics, toxicity, and potential side effects of coumarins in humans. Continued clinical evaluation will be crucial to confirm their effectiveness in cancer patients. Nonetheless, their ability to target multiple pathways positions coumarin based molecules as potential candidates for future anti-cancer drug development.
Collapse
Affiliation(s)
- Mohd Aqib
- Department of Chemistry, Govt. Raza P.G. College, Rampur, M. J. P. Rohilkhand University, Bareilly, UP, India
| | | | - Mujahid Ali
- Department of Physical Education, Govt. Raza P.G. College, Rampur, M. J. P. Rohilkhand University, Bareilly, UP, India
| | - Shabana Sajid
- Department of Chemistry, Gandhi Faiz-e-Aam College, Shahjahanpur, M. J. P. Rohilkhand University, Bareilly, UP, India
| | - Mohammed Ali Assiri
- Research Center for Advanced Materials Science (RCAMS), Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha, 61413, Saudi Arabia
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India.
| | - Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj (Allahabad), 211002, UP, India; Department of Chemistry, G. R. P. B. Degree College, P. R. S. University, Prayagraj (Allahabad), 211010, UP, India.
| | - Mohd Kamil Hussain
- Department of Chemistry, Govt. Raza P.G. College, Rampur, M. J. P. Rohilkhand University, Bareilly, UP, India.
| |
Collapse
|
2
|
Menon NA, Kumar CD, Ramachandran P, Blaize B, Gautam M, Cordani M, Lekha Dinesh Kumar. Small-molecule inhibitors of WNT signalling in cancer therapy and their links to autophagy and apoptosis. Eur J Pharmacol 2025; 986:177137. [PMID: 39551337 DOI: 10.1016/j.ejphar.2024.177137] [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: 08/01/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Cancer represents an intricate and heterogeneous ailment that evolves from a multitude of epigenetic and genetic variations that disrupt normal cellular function. The WNT/β-catenin pathway is essential in maintaining the balance between cell renewal and differentiation in various tissues. Abnormal activation of this pathway can lead to uncontrolled cell growth and initiate cancer across a variety of tissues such as the colon, skin, liver, and ovary. It enhances characteristics that lead to cancer progression, including angiogenesis, invasion and metastasis. Processes like autophagy and apoptosis which regulate cell death and play a crucial role in maintaining cellular equilibrium are also intimately linked with WNT/ β-catenin pathway. Thus, targeting WNT pathway has become a key strategy in developing antitumor therapies. Employing small molecule inhibitors has emerged as a targeted therapy to improve the clinical outcome compared to conventional cancer treatments. Many strategies using small molecule inhibitors for modulating the WNT/β-catenin pathway, such as hindering WNT ligands' secretion or interaction, disrupting receptor complex, and blocking the nuclear translocation of β-catenin have been investigated. These interventions have shown promise in both preclinical and clinical settings. This review provides a comprehensive understanding of the role of WNT/β-catenin signalling pathway's role in cancer, emphasizing its regulation of autophagy and apoptosis. Our goal is to highlight the potential of specific small molecule inhibitors targeting this pathway, fostering the development of novel, tailored cancer treatments.
Collapse
Affiliation(s)
- Nayana A Menon
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Chethana D Kumar
- Department of Surgical ICU, Christian Medical College, IDA Scudder Road, Vellore, 632004, Tamil Nadu, India
| | - Pournami Ramachandran
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Britny Blaize
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Mridul Gautam
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Lekha Dinesh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
3
|
El-Said KS, Attia MS, Abdelmoaty BE, Salim EI. Synergistic antitumor effects of atorvastatin and chemotherapies: In vitro and in vivo studies. Biochem Biophys Res Commun 2025; 742:151078. [PMID: 39632292 DOI: 10.1016/j.bbrc.2024.151078] [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: 06/14/2024] [Revised: 10/23/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Atorvastatin (ATOR) acts on certain antitumor pathways; the consequences of chemotherapies continue to be a major concern, notwithstanding the increased efficacy provided by contemporary therapies. This study investigated the synergistic effects and underlying mechanisms of different treatment protocols using ATOR on the THP-1 cell line and on lung cancer in mice. For the in vitro study, an MTT assay was performed, and then different combinations against the THP-1 cell line were used as follows: non-treated cells, THP-1/ATOR IC50, THP-1/cytarabine (CYT) IC50, THP-1/doxorubicin (DOX) IC50, THP-1/DOX/CYT, THP-1/ATOR/CYT, THP-1/ATOR/DOX, and THP-1/ATOR/CYT/DOX. For the in vivo study, CD-1 male mice were used; G1 was the normal control. Gs2-5 were administered with urethane (Ure) and butylated hydroxytoluene (BHT). G2 was the positive control. G3 was treated with ATOR (20 mg/kg). G4 was treated with Bevacizumab (Bev) (5 mg/kg). G5 was co-treated with ATOR/Bev. Histopathological and immunohistochemical investigations, flow cytometry and molecular analysis of PI3K, Akt, and mTOR genes were performed after different treatment protocols. The results showed that different combinatorial treatment settings of ATOR in vitro increase the apoptotic-inducing capacity and cell cycle arrest. Co-treatment with ATOR and Bev led to a significant decrease in S-phase and G2/M percentages. Furthermore, in vivo co-treatment with ATOR/Bev decreased tumor incidence and size with a significant reduction of the immunohistochemical PCNA (LI%) in lung parenchyma, targeting PI3K/Akt/mTOR, and VEGF-A signaling pathways. Co-treatment with ATOR and chemotherapies led to cell cycle arrest, modulation of the PI3K/Akt/mTOR, and VEGF-A signaling pathways in tumor cells.
Collapse
Affiliation(s)
- Karim Samy El-Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Merna Saied Attia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Bassant Ezzat Abdelmoaty
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Elsayed Ibrahim Salim
- Research Lab. of Molecular Carcinogenesis, Zoology Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
4
|
Jiang X, Nik Nabil WN, Ze Y, Dai R, Xi Z, Xu H. Unlocking Natural Potential: Antibody-Drug Conjugates With Naturally Derived Payloads for Cancer Therapy. Phytother Res 2024. [PMID: 39688127 DOI: 10.1002/ptr.8407] [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: 08/15/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Natural compound-derived chemotherapies remain central to cancer treatment, however, they often cause off-target side effects that negatively impact patients' quality of life. In contrast, antibody-drug conjugates (ADCs) combine cytotoxic payloads with antibodies to specifically target cancer cells. Most approved and clinically investigated ADCs utilize naturally derived payloads, while those with conventional synthetic molecular payloads remain limited. This review focuses on approved ADCs that enhance the efficacy of naturally derived payloads by linking them with antibodies. We provide an overview of the core components of ADCs, their working mechanisms, and FDA-approved ADCs featuring naturally derived payloads, such as calicheamicin, camptothecin, dolastatin 10, maytansine, pyrrolbenzodiazepine (PBD), and the immunotoxin Pseudomonas exotoxin A. This review also explores recent clinical advancements aimed at broadening the therapeutic potential of ADCs, their applicability in treating heterogeneously composed tumors and their potential use beyond oncology. Additionally, this review highlights naturally derived payloads that are currently being clinically investigated but have not yet received approval. By summarizing the current landscape, this review provides insights into promising avenues for exploration and contributes to the refinement of treatment protocols for improved patient outcomes.
Collapse
Affiliation(s)
- Xue Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Pharmaceutical Regulatory Agency, Ministry of Health, Selangor, Malaysia
| | - Yufei Ze
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Loo CY, Traini D, Young PM, Yeung S, Leong CR, Lee WH. Evaluation of curcumin nanoparticles of various sizes for targeting multidrug-resistant lung cancer cells via inhalation. Nanomedicine (Lond) 2024:1-13. [PMID: 39660666 DOI: 10.1080/17435889.2024.2439241] [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: 07/30/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Inhalation drug delivery can deliver high doses of chemotherapeutic drugs to the lung tumor. This study evaluates the efficacy and the mechanistic pathways of nebulized Cur NPs at various sizes to treat multidrug resistant lung cancer. METHODS AND RESULTS Cur-NPs (30 nm and 200 nm) were nebulized separately onto the multidrug-resistant lung cancer cells (H69AR). Smaller NPs induced significantly higher cell death owing to a higher rate of particle internalization via dynamin-dependent clathrin-mediated endocytosis. Owing to the higher lysosome trafficking of Cur-NP30 nm compared to Cur-NP200 nm, oxidation of lysosome was higher (0.47 ± 0.08 vs 0.38 ± 0.08), contributing to significantly higher mitochondrial membrane potential loss (1.57 ± 0.17 vs 1.30 ± 0.11). MRP1 level in H69AR cells was reduced from 352 ± 12.3 ng/µg of protein (untreated cells) to 287 ± 12 ng/µg of protein (Cur-NP30 nm) and 303 ± 13.4 ng/µg of protein (Cur-NP200 nm). NF-κB, and various cytokine expressions were reduced after treatment with nebulized Cur-NPs. CONCLUSIONS Nebulized Cur-NPs formulations could be internalized into the H69AR cells. The Cur-NPs toxicity toward the H69AR was size and time-dependent. Cur-NP30 nm was more effective than Cur-NP200 nm to retain within the cells to exert higher oxidative stresss-induced cell death.
Collapse
Affiliation(s)
- Ching-Yee Loo
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Ipoh, Malaysia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Department of Marketing, Macquarie Business School, Macquarie University, Sydney, Australia
| | - Stewart Yeung
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Chean Ring Leong
- Malaysian Institute of Chemical and Bioengineering Technology, Universiti Kuala Lumpur, Melaka, Malaysia
| | - Wing-Hin Lee
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Ipoh, Malaysia
| |
Collapse
|
6
|
Wang XC, Huang XY, Zhang FX, Muhire J, Di DL, Hai J, Pei D. Recent Advances in Screening and Separating Active Components From Natural Products Based on High-Speed Countercurrent Chromatography. J Sep Sci 2024; 47:e70051. [PMID: 39681966 DOI: 10.1002/jssc.70051] [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: 08/30/2024] [Revised: 11/07/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Natural products play a vital role in human health because people find that they can reduce the incidence of many diseases, such as cancer and cardiovascular diseases. Therefore, researchers have developed many effective techniques for screening and separating active compounds from natural products. High-speed countercurrent chromatography is a unique unsupported liquid-liquid chromatography technology that has been widely used in the separation of natural products. In this paper, the research progress of screening and separating active components from natural products by combining high-speed countercurrent chromatography technology with other technologies is reviewed from two aspects: pre-column activity assay and post-column activity assay. The purpose of this paper is to provide some theoretical support for the screening and separation of natural active compounds by high-speed countercurrent chromatography. I hope that the contents summarized in this paper can provide more basis for the development of screening and separation of natural active compounds to realize large-scale preparation.
Collapse
Affiliation(s)
- Xing-Cui Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Chemistry Department, University of Chinese Academy of Sciences, Beijing, China
| | - Xin-Yi Huang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Chemistry Department, University of Chinese Academy of Sciences, Beijing, China
| | - Fu-Xin Zhang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Chemistry Department, University of Chinese Academy of Sciences, Beijing, China
| | - Jules Muhire
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Chemistry Department, University of Chinese Academy of Sciences, Beijing, China
| | - Duo-Long Di
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Chemistry Department, University of Chinese Academy of Sciences, Beijing, China
| | - Jun Hai
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Chemistry Department, University of Chinese Academy of Sciences, Beijing, China
| | - Dong Pei
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory of Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou, China
- Research Department, Yunnan Olive Health Industry Innovation Research and Development Co., Ltd, Lijiang, China
| |
Collapse
|
7
|
Atmaca H, Çamli Pulat Ç, Ilhan S, Kalyoncu F. Hericium erinaceus Extract Induces Apoptosis via PI3K/AKT and RAS/MAPK Signaling Pathways in Prostate Cancer Cells. Chem Biodivers 2024; 21:e202400905. [PMID: 39183463 DOI: 10.1002/cbdv.202400905] [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: 04/09/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Prostate cancer (PCa) is increasing globally, surpassing lung cancer in incidence. Despite available treatment options, prostate cancer remains incurable. Hence, novel therapeutic strategies are urgently needed to treat PCa. Hericium erinaceus (HE), a medicinal mushroom, offers diverse therapeutic benefits. We examined HE's effects on PCa cells, preparing an ethanol extract and identifying its volatile compounds through GC-MS. MTT assay assessed cell viability, while specific inhibitors and western blotting explored HE's impact on PI3K/AKT and RAS/MAPK pathways. Flow cytometry and ELISA evaluated apoptosis induction. HE showed concentration- and time-dependent cytotoxicity on PCa cells with minimal effects on normal cells. Mechanistically, HE suppressed PI3K/AKT and RAS/MAPK pathways, reducing phosphorylated protein levels. Moreover, it induced PCa cell apoptosis. These findings suggest HE as a potential therapeutic for prostate cancer, shedding light on its cytotoxic and apoptotic effects for further investigation.
Collapse
Affiliation(s)
- Harika Atmaca
- Faculty of Engineering and Natural Sciences, Department of Biology, Manisa Celal Bayar University, Manisa, Türkiye
| | - Çisil Çamli Pulat
- Applied Science Research Center, Manisa Celal Bayar University, Manisa, Türkiye
| | - Suleyman Ilhan
- Faculty of Engineering and Natural Sciences, Department of Biology, Manisa Celal Bayar University, Manisa, Türkiye
| | - Fatih Kalyoncu
- Faculty of Engineering and Natural Sciences, Department of Biology, Manisa Celal Bayar University, Manisa, Türkiye
| |
Collapse
|
8
|
Yeniocak S, Karaduman-Yeşildal T, Arslan ME, Toraman GC, Yücetepe A. Effect of In Vitro Digestion on Anticancer and Antioxidant Activity of Phenolic Extracts From Latex of Fig Fruit (Ficus carica L.). Chem Biodivers 2024:e202401624. [PMID: 39590517 DOI: 10.1002/cbdv.202401624] [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: 07/04/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 11/28/2024]
Abstract
In this study, changes in total phenolic content (TPC), phenolic profile, and antioxidant activity and anticancer activity against cervix cancer and colorectal cancer cell lines of phenolic extracts of black and white fig (Ficus carica L.) latex (milk) were investigated during in vitro gastrointestinal digestions for the first time. The findings indicated that the in vitro digestion process exerted a significant effect on TPC of the phenolic extract from white fig milk (WFM-PE) and phenolic extract from black fig milk (BFM-PE), and TPC tended to decrease after in vitro digestion (p < 0.05). As consistent with these findings, antioxidant activity (by the CUPRAC method) of the samples decreased (p < 0.05) during in vitro digestion. The IC50 value of the undigested BFM-PE was significantly lower than that of the undigested WFM-PE (p < 0.05). The undigested and the digested WFM-PE and BFM-PE did not show any cytotoxic activity against normal cells. However, anticancer activity of WFM-PE on cervix and colorectal cancer cell lines (p < 0.05) and anticancer activity of BFM-PE against colorectal cancer cell lines decreased after in vitro digestion (p < 0.01). On the other hand, the dominant phenolic was catechin hydrate and was syringic acid.
Collapse
Affiliation(s)
- Salih Yeniocak
- Department of Food Engineering, Faculty of Engineering, Aksaray University, Aksaray, Turkey
| | - Tuğçe Karaduman-Yeşildal
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Aksaray University, Aksaray, Turkey
| | - Mustafa Enes Arslan
- Department of Food Engineering, Faculty of Engineering, Aksaray University, Aksaray, Turkey
| | - Gizem Cansu Toraman
- Department of Food Engineering, Faculty of Engineering, Aksaray University, Aksaray, Turkey
| | - Aysun Yücetepe
- Department of Food Engineering, Faculty of Engineering, Aksaray University, Aksaray, Turkey
| |
Collapse
|
9
|
Poje G, Šakić D, Marinović M, You J, Tarpley M, Williams KP, Golub N, Dernovšek J, Tomašič T, Bešić E, Rajić Z. Unveiling the antiglioblastoma potential of harmicens, harmine and ferrocene hybrids. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2024:acph-2024-0033. [PMID: 39560310 DOI: 10.2478/acph-2024-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 11/20/2024]
Abstract
The poor prognosis of glioblastoma multiforme, inadequate treatment options, and growing drug resistance urge the need to find new effective agents. Due to the significant anti-cancer potential of harmicens, hybrid compounds which comprise harmine/β-carboline and ferrocene moiety, we investigated their antiglioblastoma potential in vitro and mechanism of action (inhibition of DYRK1A, Hsp90, anti-oxidative activity). The results have shown that triazole-type harmicens, namely 5, with a ferrocene moiety in C-3 position of the β-carboline ring (IC 50 = 3.7 ± 0.1 µmol L-1, SI = 12.6) and 9, the C-6 substituted harmicene (IC 50 = 7.4 ± 0.5 µmol L-1, SI = 5.8) exert remarkable activity and selectivity against human malignant glioblastoma cell line (U251) in vitro. On the other hand, amide-type harmicens 10, 12, and 14 exhibited strong, but non-selective activity, in the low micro-molar range. Mechanistic studies revealed that among active compounds, amide-type harmicens 12 and 14 inhibit DYRK1A and Hsp90 CTD, whereas compound 14 showed pronounced antioxidative activity. Therefore, the antiproliferative activity of harmicens might be a combination of complex molecular interactions.
Collapse
Affiliation(s)
- Goran Poje
- 1University of Zagreb, Faculty of Pharmacy and Biochemistry, 10 000 Zagreb, Croatia
| | - Davor Šakić
- 1University of Zagreb, Faculty of Pharmacy and Biochemistry, 10 000 Zagreb, Croatia
| | - Marina Marinović
- 1University of Zagreb, Faculty of Pharmacy and Biochemistry, 10 000 Zagreb, Croatia
| | | | | | | | - Nikolina Golub
- 1University of Zagreb, Faculty of Pharmacy and Biochemistry, 10 000 Zagreb, Croatia
| | - Jaka Dernovšek
- 4University of Ljubljana, Faculty of Pharmacy, 1000 Ljubljana, Slovenia
| | - Tihomir Tomašič
- 4University of Ljubljana, Faculty of Pharmacy, 1000 Ljubljana, Slovenia
| | - Erim Bešić
- 1University of Zagreb, Faculty of Pharmacy and Biochemistry, 10 000 Zagreb, Croatia
| | - Zrinka Rajić
- 1University of Zagreb, Faculty of Pharmacy and Biochemistry, 10 000 Zagreb, Croatia
| |
Collapse
|
10
|
Li N, Zhu X, Zhang H, Yang X, Shao M, Cui S, Lin C. Exploring the Target Genes of Fucosylated Chondroitin Sulfate in Treating Lung Adenocarcinoma Based on the Integration of Bioinformatics Analysis, Molecular Docking, and Experimental Verification. ACS OMEGA 2024; 9:46312-46322. [PMID: 39583738 PMCID: PMC11579779 DOI: 10.1021/acsomega.4c07295] [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: 08/08/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
Fucosylated chondroitin sulfate (FCS), extracted from sea cucumbers' body walls, has been found to inhibit the proliferation of lung adenocarcinoma (LUAD) cells. However, there have been few studies of the associated drug targets. This study combined bioinformatics analysis and molecular docking to screen the main targets of FCS intervention in LUAD. Moreover, an experimental validation was performed. First, we downloaded the LUAD gene data set from The Cancer Genome Atlas (TCGA) database and the cisplatin (DDP) resistance gene data set of LUAD A549 cells from the Gene Expression Omnibus (GEO) database. Nine significant genes (PLK1, BUB1, CDK1, CDC20, CCNB1, BUB1B, KIF11, CCNB2, and DLAGP5) were identified by bioinformatics analysis, and these nine genes overlapped in both data sets. Then, molecular docking results showed that FCS had a better affinity with target proteins BUB1 and PLK1. Further experimental verification revealed that FCS inhibited the growth of A549 cells and increased the sensitivity of A549 cells to DDP. Quantitative real-time polymerase chain reaction (qRT-PCR) revealed that A549 cells treated with FCS exhibited down-regulated BUB1 and PLK1 mRNA expression. At the same time, FCS+DDP treatment resulted in a more significant reduction in BUB1 and PLK1 mRNA expression than DDP or FCS treatment alone. These findings reveal potential targets of FCS for LUAD and provide clues for the development of FCS as a potential anticancer agent.
Collapse
Affiliation(s)
- Nana Li
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xinhong Zhu
- Department
of International Medicine, Qingdao Municipal
Hospital Group, Qingdao 266071, China
| | - Hua Zhang
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaohui Yang
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Mingju Shao
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Shichao Cui
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Cunzhi Lin
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
11
|
Qian Y, Dong J, Zhang W, Xue X, Xiong Z, Zeng W, Wang Q, Fan Z, Zuo Z, Huang Z, Jiang Y. Deguelin inhibits the glioblastoma progression through suppressing CCL2/NFκB signaling pathway. Neuropharmacology 2024; 259:110109. [PMID: 39128581 DOI: 10.1016/j.neuropharm.2024.110109] [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: 03/25/2024] [Revised: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Glioblastoma multiforme (GBM) is the most common primary intracranial tumor with characteristics of high aggressiveness and poor prognosis. Deguelin, a component from the bark of Leguminosae Mundulea sericea (African plant), displays antiproliferative effects in some tumors, however, the inhibitory effect and mechanism of deguelin on GBM were still poorly understood. At first, we found that deguelin reduced the viability of GBM cells by causing cell cycle arrest in G2/M phase and inducing their apoptosis. Secondly, deguelin inhibited the migration of GBM cells. Next, RNA-seq analysis identified that CCL2 (encoding chemokine CCL2) was downregulated significantly in deguelin-treated GBM cells. As reported, CCL2 promoted the cell growth, and CCL2 was associated with regulating NFκB signaling pathway, as well as involved in modulating tumor microenvironment (TME). Furthermore, we found that deguelin inactivated CCL2/NFκB signaling pathway, and exougous CCL2 could rescue the anti-inhibitory effect of deguelin on GBM cells via upregulating NFκB. Finally, we established a syngeneic intracranial orthotopic GBM model and found that deguelin regressed the tumor growth, contributed to an anti-tumorigenic TME and inhibited angiogenesis of GBM by suppressing CCL2/NFκB in vivo. Taken together, these results suggest the anti-GBM effect of deguelin via inhibiting CCL2/NFκB pathway, which may provide a new strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Yiming Qian
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Jianhong Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Wei Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiumin Xue
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Zhenrong Xiong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Weiquan Zeng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Qian Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Ziwei Fan
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhenxing Zuo
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Yuanyuan Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
12
|
Kuttikrishnan S, Ansari AW, Suleman M, Ahmad F, Prabhu KS, El-Elimat T, Alali FQ, Al Shabeeb Akil AS, Bhat AA, Merhi M, Dermime S, Steinhoff M, Uddin S. The apoptotic and anti-proliferative effects of Neosetophomone B in T-cell acute lymphoblastic leukaemia via PI3K/AKT/mTOR pathway inhibition. Cell Prolif 2024:e13773. [PMID: 39542458 DOI: 10.1111/cpr.13773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
The phosphatidylinositol 3-kinase/Protein Kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signalling pathway is pivotal in various cancers, including T-cell acute lymphoblastic leukaemia (T-ALL), a particularly aggressive type of leukaemia. This study investigates the effects of Neosetophomone B (NSP-B), a meroterpenoid fungal metabolite, on T-ALL cell lines, focusing on its anti-cancer mechanisms and therapeutic potential. NSP-B significantly inhibited the proliferation of T-ALL cells by inducing G0/G1 cell cycle arrest and promoting caspase-dependent apoptosis. Additionally, NSP-B led to the dephosphorylation and subsequent inactivation of the PI3K/AKT/mTOR signalling pathway, a critical pathway in cell survival and growth. Molecular docking studies revealed a strong binding affinity of NSP-B to the active site of AKT, primarily involving key residues crucial for its activity. Interestingly, NSP-B treatment also induced apoptosis and significantly reduced proliferation in phytohemagglutinin-activated primary human CD3+ T cells, accompanied by a G0/G1 cell cycle arrest. Importantly, NSP-B did not affect normal primary T cells, indicating a degree of selectivity in its action, targeting only T-ALL cells and activated T cells. In conclusion, our findings highlight the potential of NSP-B as a novel therapeutic agent for T-ALL, specifically targeting the aberrantly activated PI3K/AKT/mTOR pathway and being selective in action. These results provide a strong basis for further investigation into NSP-B's anti-cancer properties and potential application in T-ALL clinical therapies.
Collapse
Affiliation(s)
- Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Abdul W Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Muhammad Suleman
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Tamam El-Elimat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Qatar
| | - Feras Q Alali
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Ammira S Al Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- College of Health Sciences, Qatar University, Doha, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology & Venereology, Hamad Medical Corporation, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Roy A, Cheriyan BV, Perumal E, Rengasamy KR, Anandakumar S. Effect of hinokitiol in ameliorating oral cancer: in vitro and in silico evidences. Odontology 2024:10.1007/s10266-024-01020-1. [PMID: 39540968 DOI: 10.1007/s10266-024-01020-1] [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: 02/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The study aimed to evaluate the anticancer potential of hinokitiol in treating oral cancer by using in vitro models and examining its interaction with the Pim-1 protein through in silico methods. Hinokitiol was applied to KB-1 oral squamous carcinoma cells, where the half-maximal inhibitory concentrations (IC50) was determined. Morphologic changes in treated cells were observed using phase contrast microscopy, while acridine orange/ethidium bromide (AO/EB) staining was used to assess nuclear changes and apoptosis. Flow cytometry was employed to analyze the cell-cycle progression. Given the high expression of Pim-1 in oral squamous carcinoma cells, molecular docking and simulation were performed to evaluate hinokitiol's binding affinity and stability with the Pim-1 protein. To compare its effects, hinokitiol was also tested on non-cancerous pre-adipocytes (3T3-L1), providing insights into its selective cytotoxicity between healthy and cancerous cells. Hinokitiol treatment resulted in cytotoxic effects on KB-1 oral squamous carcinoma cells, with an IC50 of 30 µg/mL after 24 and 48 hs of exposure. Morphologic studies showed reduced cell population and density. In contrast, hinokitiol exhibited lower toxicity and caused fewer morphological changes in non-cancerous 3T3-L1 pre-adipocytes. Apoptosis was confirmed through acridine orange/ethidium bromide staining, while flow cytometry revealed cell-cycle arrest in the Synthesis phase (S) and Gap 2 phase/ Mitosis Phase (G2/M) phases. Molecular docking showed strong binding of hinokitiol to Pim-1, and simulations confirmed the interaction's stability. These findings suggest hinokitiol selectively targets cancer cells and effectively inhibit Pim-1, supporting its potential as an oral cancer treatment.
Collapse
Affiliation(s)
- Anitha Roy
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, SIMATS, Chennai, Tamil Nadu, 600077, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, SIMATS, Chennai, Tamil Nadu, 602105, India.
| | - Elumalai Perumal
- Cancer Genomics lab, Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, SIMATS, Chennai, Tamil Nadu, 602105, India
| | - Kannan Rr Rengasamy
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, SIMATS, Chennai, Tamil Nadu, 602105, India
| | - Shanmugam Anandakumar
- Department of Microbiology, Dr. ALM Post Graduate Institute of Basic Medical Science, University of Madras, Chennai, Tamil Nadu, 600113, India
| |
Collapse
|
14
|
Moralev A, Zenkova MA, Markov AV. Complex Inhibitory Activity of Pentacyclic Triterpenoids against Cutaneous Melanoma In Vitro and In Vivo: A Literature Review and Reconstruction of Their Melanoma-Related Protein Interactome. ACS Pharmacol Transl Sci 2024; 7:3358-3384. [PMID: 39539268 PMCID: PMC11555519 DOI: 10.1021/acsptsci.4c00422] [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: 07/18/2024] [Revised: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Pentacyclic triterpenoids (PTs) are a class of plant metabolites with a wide range of pharmacological activities, including strong antitumor potential against skin malignancies. By acting on multiple signaling pathways that control key cellular processes, PTs are able to exert complex effects on melanoma progression in vitro and in vivo. In this review, we have analyzed the works published in the past decade and devoted to the effects of PTs, both natural and semisynthetic, on cutaneous melanoma pathogenesis, including not only their direct action on melanoma cells but also their influence on the tumor microenvironment and abberant melanogenesis, often associated with melanoma aggressiveness. Special attention will be paid to the molecular basis of the pronounced antimelanoma potency of PTs, including a detailed consideration of the pathways sensitive to PTs in melanoma cells, as well as the reconstruction of the melanoma-related protein interactome of PTs using a network pharmacology approach based on previously published experimentally verified protein targets of PTs. The information collected on the primary targets of PTs was compiled in the Protein Interactome of PTs (PIPTs) database, freely available at http://www.pipts-db.ru/, which can be used to further optimize the mechanistic studies of PTs in the context of melanoma and other malignancies. By summarizing recent research findings, this review provides valuable information to scientists working in the fields related to the evaluation of melanoma pathogenesis and development of PTs-based drug candidates.
Collapse
Affiliation(s)
- Arseny
D. Moralev
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| | - Andrey V. Markov
- Institute of Chemical Biology and Fundamental
Medicine, Siberian Branch of the Russian
Academy of Sciences, 630090, Lavrent’ev avenue 8, Novosibirsk, Russia
| |
Collapse
|
15
|
Mitra A, Roy R, Paul S. Modulating the Self-Assembly of a Camptothecin Prodrug with Paclitaxel for Anticancer Combination Therapy: A Molecular Dynamics Approach. J Phys Chem B 2024; 128:10799-10812. [PMID: 39230512 DOI: 10.1021/acs.jpcb.4c04798] [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: 09/05/2024]
Abstract
Camptothecin (CPT) and paclitaxel (PTX), derived from natural products, are recognized for their significant efficacy in clinical cancer treatments. Despite its therapeutic advantages, CPT is challenged by issues of toxicity and solubility, necessitating its use in conjugation with other compounds for enhanced compatibility. This study delves into the coassembly mechanism of Evans blue-conjugated camptothecin (EB-CPT) with PTX, aiming to elucidate their synergistic potential in combination therapy applications, employing all-atom molecular dynamics simulations. The EB-CPT prodrug is reported to form a self-aggregated cluster. Our findings suggest that increasing the PTX concentration induces a dispersion of EB-CPT clusters, thereby disrupting their inherent self-assembly. This disruption is explained to be facilitated by the coassembly of EB-CPT and PTX. With increasing concentration of PTX, a lengthening of the coassembled structures is observed, supporting the experimental findings of tube-like coassembled structures at higher weight ratios of PTX. Hydrophobic interactions and π-π stacking are the primary forces responsible for the formation of both self- and coassembled structures. Interestingly, the structural analysis reveals that the CPT moiety of EB-CPT is less involved in assemblies due to steric hindrances. Instead, the interaction and coassembly processes are predominantly mediated by the EB derivative component of the prodrug. This research underscores the critical role of the solubilizing agent, EB derivative, in mediating the flexibility and interaction of CPT in combination therapy strategies, particularly with PTX, thus emphasizing the importance of conjugates for therapeutic developments. Furthermore, the molecular insights into the interaction sites and mechanisms facilitating coassembly between EB-CPT and PTX contribute valuable knowledge to the field, highlighting the potential of these nanomedicine combinations in advancing cancer treatment modalities.
Collapse
Affiliation(s)
- Anandita Mitra
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| |
Collapse
|
16
|
Wen Y, Li Y, Li BB, Liu P, Qiu M, Li Z, Xu J, Bi B, Zhang S, Deng X, Liu K, Zhou S, Wang Q, Zhao J. Pyroptosis induced by natural products and their derivatives for cancer therapy. Biomater Sci 2024; 12:5656-5679. [PMID: 39429101 DOI: 10.1039/d4bm01023j] [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: 10/22/2024]
Abstract
Natural products, which are compounds extracted and/or refined from plants and microbes in nature, have great potential for the discovery of therapeutic agents, especially for infectious diseases and cancer. In recent years, natural products have been reported to induce multiple cell death pathways to exhibit antitumor effects. Among them, pyroptosis is a unique programmed cell death (PCD) characterized by continuous cell membrane permeability and intracellular content leakage. According to the canonical and noncanonical pathways, the formation of gasdermin-N pores involves a variety of transcriptional targets and post-translational modifications. Thus, tailored control of PCD may facilitate dying cells with sufficient immunogenicity to activate the immune system to eliminate other tumor cells. Therefore, we summarized the currently reported natural products or their derivatives and their nano-drugs that induce pyroptosis-related signaling pathways. We reviewed six main categories of bioactive compounds extracted from natural products, including flavonoids, terpenoids, polyphenols, quinones, artemisinins, and alkaloids. Correspondingly, the underlying mechanisms of how these compounds and their derivatives engage in pyroptosis are also discussed. Moreover, the synergistic effect of natural bioactive compounds with other antitumor therapies is proposed as a novel therapeutic strategy for traditional chemotherapy, radiotherapy, chemodynamic therapy, photodynamic therapy, photothermal therapy, hyperthermal therapy, and sonodynamic therapy. Consequently, we provide insights into natural products to develop a novel antitumor therapy or qualified adjuvant agents by inducing pyroptosis, which may eventually be applied clinically.
Collapse
Affiliation(s)
- Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Bin-Bin Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Peng Liu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Miaojuan Qiu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Zihang Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Jiaqi Xu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Bo Bi
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Xinyi Deng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Kaiyuan Liu
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shangbo Zhou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Qiang Wang
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
17
|
Banjare L, Murmu A, Pandey NK, Matore BW, Banjare P, Bhattacharya A, Gayen S, Singh J, Roy PP. First report on exploration of structural features of natural compounds (NPACT database) for anti-breast cancer activity (MCF-7): QSAR-based virtual screening, molecular docking, ADMET, MD simulation, and DFT studies. In Silico Pharmacol 2024; 12:92. [PMID: 39435346 PMCID: PMC11490471 DOI: 10.1007/s40203-024-00266-5] [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: 08/02/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Due to the high toxicity, poor efficacy and resistance associated with current anti-breast cancer drugs, there's growing interest in natural products (NPs) for their potential anti-cancer properties. Computational modelling of NPs to identify key structural features can aid in developing novel natural inhibitors. In this study, we developed statistically significant QSAR models based on NPs from the NPACT database, which have shown potential anticancer activity against the MCF-7 cancer cell lines. All the developed QSAR models were statistically robust, meeting both internal (R 2 = 0.666-0.669, R 2 adj = 0.657-0.660, Q 2 Loo = 0.636-0.638) and external (Q 2 F n = 0.686-0.714, CCC ext = 0.830-0.847) validation criteria. Consequently, they were utilized to virtually screen a series of NPs from the COCONUT database in the search for novel natural inhibitors. Molecular docking studies were conducted on the identified compounds against the human HER2 protein (PDB ID: 3PP0), which is a crucial target in breast cancer. Molecular docking analysis demonstrated that compounds 4608 and 2710 achieved the highest docking scores, with CDOCKER interaction energies of -72.67 kcal/mol and - 72.63 kcal/mol respectively. Compounds 4608 and 2710 were identified as the most promising candidates upon performing triplicate 100 ns MD simulation study using the CHARMM36 force field. DFT studies was performed to evaluate their stability and reactivity as potential drug molecules. This research contributes to the development of new natural inhibitors for breast cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00266-5.
Collapse
Affiliation(s)
- Lomash Banjare
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| | - Anjali Murmu
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| | - Nilesh Kumar Pandey
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| | - Balaji Wamanrao Matore
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| | - Purusottam Banjare
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| | - Arijit Bhattacharya
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Jagadish Singh
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| | - Partha Pratim Roy
- Laboratory of Drug Discovery and Ecotoxicology, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, 495009 India
| |
Collapse
|
18
|
Arora M, Singh AK, Kumar A, Singh H, Pathak P, Grishina M, Yadav JP, Verma A, Kumar P. Semisynthetic phytochemicals in cancer treatment: a medicinal chemistry perspective. RSC Med Chem 2024; 15:3345-3370. [PMID: 39430100 PMCID: PMC11484407 DOI: 10.1039/d4md00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer is the uncontrolled proliferation of abnormal cells that invade other areas, spread to other organs, and cause metastases, which is the most common cause of death. A review of all FDA-approved new molecular entities (NMEs) shows that natural products and derivatives account for over one-third of all NMEs. Before 1940, unmodified products and derivatives accounted for 43% and 14% of NME registrations, respectively. Since then, the share of unmodified products has decreased to 9.5% of all approved NMEs, while the share of derivatives has increased to 28%. Since the 1940s, semi-synthetic and synthetic derivatives of natural substances have gained importance, and this trend continues to date. In this study, we have discussed in detail isolated phytoconstituents with chemical modifications that are either FDA-approved or under clinical trials, such as podophyllotoxin, Taxol (paclitaxel, docetaxel), vinca alkaloids (vincristine, vinblastine), camptothecin, genistein, cephalotaxine, rohitukine, and many more, which may act as essential leads to the development of novel anticancer agents. Furthermore, we have also discussed recent developments in the most potent semisynthetic phytoconstituents, their unique properties, and their importance in cancer treatment.
Collapse
Affiliation(s)
- Meghna Arora
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Prateek Pathak
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, School of Pharmacy, GITAM (Deemed to be University) Hyderabad Campus India
| | - Maria Grishina
- Laboratory of Computational Modeling of Drugs, Higher Medical and Biological School, South Ural State University Chelyabinsk 454008 Russia
| | - Jagat Pal Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University Kanpur 209217 India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| |
Collapse
|
19
|
Awadelkareem AM, Patel M, Banu H, Adnan M. Integrating computational methods and i n vitro experimental validation reveals the pharmacological mechanism of Selaginella bryopteris (L.) Baker targeting major proteins in breast cancer. Heliyon 2024; 10:e38801. [PMID: 39430520 PMCID: PMC11489316 DOI: 10.1016/j.heliyon.2024.e38801] [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: 08/04/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
Breast cancer remains a significant global health challenge, necessitating the exploration of novel therapeutic options. The present study employs an integrated approach encompassing network pharmacology, molecular docking, molecular dynamics simulations, and in-vitro validation to investigate the potential of Selaginella bryopteris in breast cancer treatment. Initial network pharmacology analysis revealed different potential targets and pathways associated with breast cancer that could be modulated by S. bryopteris phytochemical constituents. Molecular docking and dynamics simulations further elucidated the stability and dynamics of protein-ligand complexes (lanaroflavone-EGFR and sequoiaflavone-CTNNB1). The in-vitro assays demonstrated the ability of S. bryopteris crude extract to inhibit cancer cell growth (IC50 - 78.34 μg/mL) migration and invasion, supporting the computational predictions. The integrated approach employed in the present study offers a robust framework for the systematic exploration of S. bryopteris in drug discovery as a promising candidate for breast cancer treatment.
Collapse
Affiliation(s)
- Amir Mahgoub Awadelkareem
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Mitesh Patel
- Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Humera Banu
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| |
Collapse
|
20
|
Inoue TT, Viana Pereira V, Faria de Sousa G, Nunes Dourado LF, da Silva Cunha-Junior A. Anti-angiogenic activity of polymeric nanoparticles loaded with ursolic acid. J Drug Target 2024:1-10. [PMID: 39325639 DOI: 10.1080/1061186x.2024.2409881] [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: 08/05/2024] [Revised: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Ursolic acid (UA) is an abundant natural product and has shown great promise for treating diseases related to the appearance of new blood vessels. However, its clinical use is limited due to its low solubility in aqueous media, resulting in reduced bioavailability. The present study aimed to synthetize poly(lactic-co-glycolic acid) nanoparticles loaded with UA by nanoprecipitation method and to evaluate the toxicity and anti-angiogenic activity using the in vivo chorioallantoic model. The nanoparticles were obtained in the size range that varied from 103.0 to 169.3 nm, they presented a uniform distribution (polydispersity index <0.2), and a negatively charged surface, with an encapsulation efficiency close to 50%. The release profile of the developed nanoformulation showed an initial burst in the first 2 h and demonstrated no acute toxicity (irritation index <0.9). Moreover, the chorioallantoic assay showed a significant reduction in both geometrical and topological parameters compared to saline control (p < .05). In conclusion, the study revealed a quick and simple way to obtain poly(lactic-co-glycolic) acid nanoparticles, a drug delivery system to UA, which showed potential antiangiogenic action and can be used to treat diseases involving neovascularisation.
Collapse
Affiliation(s)
- Thomas Toshio Inoue
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | |
Collapse
|
21
|
Schott M, Vehlow A, Benka M, Lagies S, Kammerer B, Rieckmann T, Cordes N. Aqueous extracts from Dioscorea sansibarensis Pax show cytotoxic and radiosensitizing potential in 3D growing HPV-negative and HPV-positive human head and neck squamous cell carcinoma models. Biomed Pharmacother 2024; 179:117305. [PMID: 39167841 DOI: 10.1016/j.biopha.2024.117305] [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: 02/07/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Numerous natural substances have anti-cancer properties. Especially indigenous people use aqueous plant extracts for tea or ointments including Dioscorea sansibarensis Pax to treat various diseases. The aim of this study was to evaluate the cytotoxic and radiosensitizing potential of aqueous extracts from Dioscorea sansibarensis Pax collected from Kenya in a panel of HPV-negative and -positive head and neck squamous cell carcinoma (HNSCC) cells grown in three-dimensional laminin-rich extracellular matrix (3D lrECM). The results show cytotoxicity, radiosensitization and increased levels of residual double strand breaks (DBS) by Dioscorea sansibarensis Pax extracts in HPV-negative and -positive HNSCC models in a concentration- and cell model-dependent manner. Application of ROS scavengers indicated an association between ROS-induced DSB and radiosensitization through Dioscorea sansibarensis Pax pretreatment. High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) based characterization of Dioscorea sansibarensis Pax identified the main components of the extract including camptothecin. Overall, Dioscorea sansibarensis Pax aqueous extracts alone and in combination with X-ray irradiation showed effective anticancer properties, which are worthy of further mechanistic investigation.
Collapse
Affiliation(s)
- Mandy Schott
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Anne Vehlow
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Moritz Benka
- Core Competence Metabolomics, Hilde-Mangold-Haus, University of Freiburg, Freiburg 79104, Germany; Institute of Organic Chemistry, University of Freiburg, Freiburg 79104, Germany; Hermann Staudinger Graduate School, University of Freiburg, Freiburg 79104, Germany
| | - Simon Lagies
- Core Competence Metabolomics, Hilde-Mangold-Haus, University of Freiburg, Freiburg 79104, Germany; Department of Pneumology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Bernd Kammerer
- Core Competence Metabolomics, Hilde-Mangold-Haus, University of Freiburg, Freiburg 79104, Germany; Institute of Organic Chemistry, University of Freiburg, Freiburg 79104, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg 79104, Germany
| | - Thorsten Rieckmann
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Otorhinolaryngology, University Medical Center Hamburg Eppendorf, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology - OncoRay, Dresden 01328, Germany; German Cancer Consortium, Partner Site Dresden: German Cancer Research Center, Heidelberg 69120, Germany; Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany.
| |
Collapse
|
22
|
Singh H, Mishra AK, Mohanto S, Kumar A, Mishra A, Amin R, Darwin CR, Emran TB. A recent update on the connection between dietary phytochemicals and skin cancer: emerging understanding of the molecular mechanism. Ann Med Surg (Lond) 2024; 86:5877-5913. [PMID: 39359831 PMCID: PMC11444613 DOI: 10.1097/ms9.0000000000002392] [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: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 10/04/2024] Open
Abstract
Constant exposure to harmful substances from both inside and outside the body can mess up the body's natural ways of keeping itself in balance. This can cause severe skin damage, including basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma. However, plant-derived compounds found in fruits and vegetables have been shown to protect against skin cancer-causing free radicals and other harmful substances. It has been determined that these dietary phytochemicals are effective in preventing skin cancer and are widely available, inexpensive, and well-tolerated. Studies have shown that these phytochemicals possess anti-inflammatory, antioxidant, and antiangiogenic properties that can aid in the prevention of skin cancers. In addition, they influence crucial cellular processes such as angiogenesis and cell cycle control, which can halt the progression of skin cancer. The present paper discusses the benefits of specific dietary phytochemicals found in fruits and vegetables, as well as the signaling pathways they regulate, the molecular mechanisms involved in the prevention of skin cancer, and their drawbacks.
Collapse
Affiliation(s)
- Harpreet Singh
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh
| | | | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka
| | - Arvind Kumar
- School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh
| | - Amrita Mishra
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi
| | - Ruhul Amin
- Faculty of Pharmaceutical Science, Assam downtown University, Panikhaiti, Gandhinagar, Guwahati, Assam
| | | | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
23
|
Mohamed MA, Elsaman T, Mohamed MS, Eltayib EM. Computational investigations of flavonoids as ALDH isoform inhibitors for treatment of cancer. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:837-875. [PMID: 39503629 DOI: 10.1080/1062936x.2024.2415593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024]
Abstract
Human aldehyde dehydrogenases (ALDHs) are a group of 19 isoforms often overexpressed in cancer stem cells (CSCs). These enzymes play critical roles in CSC protection, maintenance, cancer progression, therapeutic resistance, and poor prognosis. Thus, targeting ALDH isoforms offers potential for innovative cancer treatments. Flavonoids, known for their ability to affect multiple cancer-related pathways, have shown anticancer activity by downregulating specific ALDH isoforms. This study aimed to evaluate 830 flavonoids from the PubChem database against five ALDH isoforms (ALDH1A1, ALDH1A2, ALDH1A3, ALDH2, ALDH3A1) using computational methods to identify potent inhibitors. Extra precision (XP) Glide docking and MM-GBSA free binding energy calculations identified several flavonoids with high binding affinities. MD simulation highlighted flavonoids 1, 2, 18, 27, and 42 as potential specific inhibitors for each isoform, respectively. Flavonoid 10 showed high binding affinities for ALDH1A2, ALDH1A3, and ALDH3A1, emerging as a potential multi-ALDH inhibitor. ADMET property evaluation indicated that the promising hits have acceptable drug-like profiles, but further optimization is needed to enhance their therapeutic efficacy and reduce toxicity, making them more effective ALDH inhibitors for future cancer treatment.
Collapse
Affiliation(s)
- M A Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - T Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - M S Mohamed
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - E M Eltayib
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| |
Collapse
|
24
|
Zhang X, Miao J, Song Y, Zhang J, Miao M. Review on effects and mechanisms of plant-derived natural products against breast cancer bone metastasis. Heliyon 2024; 10:e37894. [PMID: 39318810 PMCID: PMC11420494 DOI: 10.1016/j.heliyon.2024.e37894] [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: 05/27/2024] [Revised: 08/16/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Bone metastasis is the prevalent form of metastasis in breast cancer, resulting in severe pain, pathological fractures, nerve compression, hypercalcemia, and other complications that significantly impair patients' quality of life. The infiltration and colonization of breast cancer (BC) cells in bone tissue disrupt the delicate balance between osteoblasts and osteoclasts within the bone microenvironment, initiating a vicious cycle of bone metastasis. Once bone metastasis occurs, conventional medical therapy with bone-modifying agents is commonly used to alleviate bone-related complications and improve patients' quality of life. However, the utilization of bone-modifying agents may cause severe drug-related adverse effects. Plant-derived natural products such as terpenoids, alkaloids, coumarins, and phenols have anti-tumor, anti-inflammatory, and anti-angiogenic pharmacological properties with minimal side effects. Certain natural products that exhibit both anti-breast cancer and anti-bone metastasis effects are potential therapeutic agents for breast cancer bone metastasis (BCBM). This article reviewed the effects of plant-derived natural products against BCBM and their mechanisms to provide a reference for the research and development of drugs related to BCBM.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yagang Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jiawen Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| |
Collapse
|
25
|
Charoensedtasin K, Kheansaard W, Roytrakul S, Tanyong D. Piperine, a black pepper compound, induces autophagy and cellular senescence mediated by NF-κB and IL-6 in acute leukemia. BMC Complement Med Ther 2024; 24:343. [PMID: 39342176 PMCID: PMC11438257 DOI: 10.1186/s12906-024-04641-9] [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: 06/27/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
Acute leukemia is characterized by abnormal white blood cell proliferation with rapid onset and severe complications. Natural compounds, which are alternative treatments, are widely used in cancer treatment. Piperine, an alkaloid compound from black pepper, exerts anticancer effects through the cell death signaling pathway. Autophagy and senescence signaling pathways are considered target signaling pathways for cancer treatment. In this study, we investigated the effects of piperine via autophagy and senescence signaling pathways in NB4 and MOLT-4 cells. The MTT assay results demonstrated that piperine significantly decreased the viability of NB4 and MOLT-4 cells. Piperine induced autophagy by increasing LC3, Beclin-1 and ULK1 and decreasing mTOR and NF-κB1 expression in NB4 and MOLT-4 cells. In addition, piperine increased senescence-associated beta-galactosidase fluorescence intensity by increasing p21 and IL-6 expression while decreasing CDK2 expression in NB4 and MOLT-4 cells. In conclusion, our study provides additional information about the induction of autophagy and senescence by piperine in acute leukemia.
Collapse
Affiliation(s)
- Kantorn Charoensedtasin
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon sai 4 Road, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand
| | - Wasinee Kheansaard
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon sai 4 Road, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology for Development Agency, Pathum Thani, 12120, Thailand
| | - Dalina Tanyong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon sai 4 Road, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
26
|
Aziz IM, Alfuraydi AA, Almarfadi OM, Aboul-Soud MAM, Alshememry AK, Alsaleh AN, Almajhdi FN. Phytochemical analysis, antioxidant, anticancer, and antibacterial potential of Alpinia galanga (L.) rhizome. Heliyon 2024; 10:e37196. [PMID: 39286191 PMCID: PMC11403495 DOI: 10.1016/j.heliyon.2024.e37196] [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: 07/28/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Alpinia galanga (L.) Willd. (A. galanga) is extremely significant and is utilized extensively in traditional medicine throughout many nations. This study aimed to determine the chemical composition of A. galanga rhizome extract (AgRE) and to evaluate its antioxidant, anticancer, and antibacterial activities. The total phenolic content (TPC) and total flavonoid content (TFC) of AgRE were determined. The antioxidant activity, cytotoxic capability, and antibacterial of were assessed, as well as anti-apoptotic genes. Molecular docking (MD) was used to assess the binding affinity of the most enriched constituents in AgRE toward the active sites of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and p53 tumor suppressor protein (TP53). Gas chromatography-mass spectrometry (GC-MS) analysis demonstrated that AgRE is a rich source of turmerone. AgRE had moderate 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) radical scavenging properties, with the half-maximal inhibitory concentration (IC50) values of 79.34 ± 1.78 and 88.94 ± 2.28 μg/ml, respectively. AgRE preferentially reduced the viability of a subset of malignant MCF-7 and HepG2 cell lines, with IC50 of 125.35 ± 4.28 and 182.49 ± 3.19 μg/ml, respectively. AgRE exhibited considerable antimicrobial activity against all bacterial strains, with MIC values ranging from 7.81 ± 1.53 to 62.5 ± 3.28 μg/ml. The MD results revealed that ethyl-4-(2-methylpropyl)-benzene had the greatest binding energy with NADPH oxidase, with a Glide score of -6848 kcal/mol, followed by 2-methoxy-phenol (-5111 kcal/mol). Taken together, we report the interesting antioxidant, antibacterial, and anticancer properties of AgRE, which warrant further investigation. AgRE is a promising natural resource that could be used to combat complicated diseases such as cancer and bacterial infections.
Collapse
Affiliation(s)
- Ibrahim M Aziz
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Akram A Alfuraydi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Omer M Almarfadi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Mourad A M Aboul-Soud
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Abdullah K Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Asma N Alsaleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fahad N Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
27
|
Yadav G, Megha, Yadav S, Tomar R. An overview: total synthesis of arborisidine, and arbornamine. Mol Divers 2024:10.1007/s11030-024-10978-7. [PMID: 39242485 DOI: 10.1007/s11030-024-10978-7] [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: 07/09/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Arborisidine and Arbornamine are two monoterpenoid indole alkaloids that were isolated from the Malayan Kopsia arborea plant. This review provides valuable information about the total and formal syntheses of these alkaloids. The synthesis strategies discussed in this review, such as Pictet-Spengler cyclization, chemo- and stereoselective oxidative cyclization, Michael/Mannich cascade process, and intramolecular N-alkylation, can be useful for developing new methods to synthesize these and other similar compounds.
Collapse
Affiliation(s)
- Gitanjali Yadav
- Department of Chemistry, Indira Gandhi University, Meerpur, Rewari, Haryana, 122502, India
- Department of Chemistry, Baba Mastnath University Asthal bohar, Rohtak, Haryana, 124021, India
| | - Megha
- Department of Chemistry, Indira Gandhi University, Meerpur, Rewari, Haryana, 122502, India
| | - Sangeeta Yadav
- Department of Chemistry, Netaji Subhas University of Technology, Dwarka, Delhi, 110078, India
| | - Ravi Tomar
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Modinagar, Ghaziabad, 201204, India.
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Delhi, 110016, India.
| |
Collapse
|
28
|
Fayyaz A, Basit M, Farooq A, Khan T, Ayub U, Khan S, Armaghan M, Mati-Ur-Rahman, Ammad M, Büsselberg D, Khan K, Habtemariam S, Sharifi-Rad J. Therapeutic potential of ethoxy mansonone G: A comprehensive exploration of its anticancer actions in breast cancer, colorectal cancer, and non-small cell lung carcinoma. Cell Biol Int 2024; 48:1229-1239. [PMID: 38924324 DOI: 10.1002/cbin.12207] [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: 01/30/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
Mansonone G (MG), a 1,2-naphthoquinones with antiestrogenic, antimicrobial, and anti-adipogenic activities, is derived from the heartwood of Mansonia gagei Drumm. Ethoxy mansonone G (EMG), an essential derivative of MG, has anticancer and antioxidant agent. EMG also has antiestrogen activity and is demonstrated to lower estrogen receptor expression in endocrine-resistant cells. EMG significantly inhibits cell division, invasion, and anchorage-dependent growth in all cancer types. Through the stimulation of the tumor protein (p53) and extracellular signal-regulated kinase (ERK) signaling cascades, it also causes apoptosis. Moreover, it manifests its anti-cancerous effects in toll-like receptor pathways, c-Jun N-terminal kinase (c-JNK), and nuclear factor kappa B (NF-κB). EMG inhibits the phosphorylation of glycogen synthase kinase (GSK3), Erk, protein kinase B (Akt), and mammalian target of rapamycin (mTOR). By interfering with molecular cascades, EMG significantly reduces the metabolism of cancer cells. This paper focuses on the potential use of EMG in cancer treatment. Moreover, it states the methodology by which specific assays establish the anti-cancerous role of EMG. Breast cancer, non-small cell lung cancer, and colorectal cancer are only a few of the cancers for which EMG was shown to be effective. Through further research, EMG may be developed as a therapeutic solution to complications caused by cancer. This study presents EMG as a novel candidate for cancer therapy, offering a unique combination of pharmacological advantages and mechanistic insights that warrant further exploration and development toward addressing the complexities of cancer treatment.
Collapse
Affiliation(s)
- Amna Fayyaz
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Mahnoor Basit
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Andleeb Farooq
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Tooba Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Umama Ayub
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Somia Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Mati-Ur-Rahman
- Pure Health Laboratory, Mafraq Hospital, Abu Dhabi, United Arab Emirates
| | - Muhammad Ammad
- Pure Health Laboratory, Mafraq Hospital, Abu Dhabi, United Arab Emirates
| | | | | | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, Central Avenue, Chatham-Maritime, Kent, UK
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
29
|
Ortigosa-Palomo A, Fuentes-Ríos D, Quiñonero F, Melguizo C, Ortiz R, López-Romero JM, Prados J. Evaluation of cytotoxic effect of siphonochilone from African ginger: an in vitro analysis. ENVIRONMENTAL TOXICOLOGY 2024; 39:4333-4346. [PMID: 38742918 DOI: 10.1002/tox.24308] [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: 09/11/2023] [Revised: 03/05/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024]
Abstract
Plants provide a wide array of compounds that can be explored for potential anticancer properties. Siphonochilone, a furanoterpene that represents one of the main components of the African plant Siphonochilus aethiopicus, shows numerous health benefits. However, to date, its antiproliferative properties have not been tested. The aim of this study was to analyze the cytotoxic effects of siphonochilone on a panel of cancer cell lines and its underlying mechanism of action. Our results demonstrated that siphonochilone exhibited significant cytotoxic effects on pancreatic, breast, lung, colon, and liver cancer cell lines showing a IC50 ranging from 22 to 124 μM at 72 h of treatment and highlighting its cytotoxic effect against MCF7 and PANC1 breast and pancreas cancer cell lines (22.03 and 39.03 μM, respectively). Cell death in these tumor lines was mediated by apoptosis by the mitochondrial pathway, as evidenced by siphonochilone-induced depolarization of the mitochondrial membrane potential. In addition, siphonochilone treatment involves the generation of reactive oxygen species that may contribute to apoptosis induction. In this work, we described for the first time the cytotoxic properties of siphonochilone and provided data about the molecular processes of cell death. Although future studies will be necessary, our results support the interest in this molecule in relation to their clinical application in cancer, and especially in breast and pancreatic cancer.
Collapse
Affiliation(s)
- Alba Ortigosa-Palomo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, Granada, Spain
| | - David Fuentes-Ríos
- Department of Organic Chemistry, Faculty of Sciences, University of Malaga, Málaga, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, Granada, Spain
| | - Raul Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, Granada, Spain
| | - Juan M López-Romero
- Department of Organic Chemistry, Faculty of Sciences, University of Malaga, Málaga, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. Granada, Granada, Spain
| |
Collapse
|
30
|
Xu P, Zhou J, Xing X, Hao Y, Gao M, Li Z, Li X, Li M, Xiao Y. Melitoxin Inhibits Proliferation, Metastasis, and Invasion of Glioma U251 Cells by Down-regulating F2RL1. Appl Biochem Biotechnol 2024; 196:6234-6252. [PMID: 38252207 DOI: 10.1007/s12010-023-04841-y] [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] [Accepted: 12/19/2023] [Indexed: 01/23/2024]
Abstract
As the principal active component of bee venom, melittin has an anti-cancer effect in different cancers. This study was aimed to investigate the effect of melittin in glioma and explore whether F2RL1 is closely involved in glioblastoma cells proliferation. TCGA and GES databases were used to evaluate the role of F2RL1 in gliomas. The U251 cells were divided into a control lentivirus + PBS group (NC-PBS), F2RL1 intervention lentivirus + PBS group (KD-PBS), control lentivirus + melittin group (NC-melittin), and F2RL1 intervention lentivirus + melittin group (KD-melittin). Cell proliferation was detected by MTT and EDU staining assays. The apoptosis rate was assessed by flow cytometry. Expressions of genes related to apoptosis, cycle arrest, migration, and invasion were detected by qRT-PCR. Cellular LDH concentrations were detected by ELISA. The subcutaneous tumor volume of nude mice was analyzed by xenograft method. F2RL1 was significantly overexpressed in glioma tissues and were reduced in the melittin-treated group compared to the blank group. F2RL1 knockdown and melittin alone or in combination increased the proportion of cells in the G1-phase, and the combination was more pronounced. The KD-melittin group showed a decrease in the number of viable cells at 24, 48, 72, and 96 h compared to the NC-PBS group. The number of cell migration and invasion was decreased in the KD-melittin group compared to the other groups. Moreover, the genes related to cell cycle arrest and apoptosis were significantly changed in the KD-melittin group. At weeks 4, 5, and 6, the tumor volume in the KD-melittin group was smaller than that in the KD-PBS group and NC-melittin group. Interference with the target gene F2RL1 inhibited the proliferation of glioma U251 cells, and melittin treatment inhibited the proliferation of glioma U251 cells. Melittin inhibited the proliferation of glioma U251 cells by suppressing the expression of target gene F2RL1.
Collapse
Affiliation(s)
- Peng Xu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Jie Zhou
- Department of Nursing, Liaocheng Vocational and Technical College, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Yuan Hao
- Department of Pathology, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Mingxu Gao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Xin Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China
| | - Mengyou Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China.
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, 252000, People's Republic of China.
| |
Collapse
|
31
|
Ding XJ, Cai XM, Wang QQ, Liu N, Zhong WL, Xi XN, Lu YX. Vitexicarpin suppresses malignant progression of colorectal cancer through affecting c-Myc ubiquitination by targeting IMPDH2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155833. [PMID: 39008915 DOI: 10.1016/j.phymed.2024.155833] [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: 03/05/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is the second most common cause of cancer-related mortality and is characterised by extensive invasive and metastatic potential. Previous studies have shown that vitexicarpin extracted from the fruits of Vitex rotundifolia can impede tumour progression. However, the molecular mechanisms involved in CRC treatment are still not fully established. PURPOSE Our study aimed to investigate the anticancer activity, targets, and molecular mechanisms of vitexicarpin in CRC hoping to provide novel therapies for patients with CRC. STUDY DESIGN/METHODS The impact of vitexicarpin on CRC was assessed through various experiments including MTT, clone formation, EDU, cell cycle, and apoptosis assays, as well as a tumour xenograft model. CETSA, label-free quantitative proteomics, and Biacore were used to identify the vitexicarpin targets. WB, Co-IP, Ubiquitination assay, IF, molecular docking, MST, and cell transfection were used to investigate the mechanism of action of vitexicarpin in CRC cells. Furthermore, we analysed the expression patterns and correlation of target proteins in TCGA and GEPIA datasets and clinical samples. Finally, wound healing, Transwell, tail vein injection model, and tissue section staining were used to demonstrate the antimetastatic effect of vitexicarpin on CRC in vitro and in vivo. RESULTS Our findings demonstrated that vitexicarpin exhibits anticancer activity by directly binding to inosine monophosphate dehydrogenase 2 (IMPDH2) and that it promotes c-Myc ubiquitination by disrupting the interaction between IMPDH2 and c-Myc, leading to epithelial-mesenchymal transition (EMT) inhibition. Vitexicarpin hinders the migration and invasion of CRC cells by reversing EMT both in vitro and in vivo. Additionally, these results were validated by the overexpression and knockdown of IMPDH2 in CRC cells. CONCLUSION These results demonstrated that vitexicarpin regulates the interaction between IMPDH2 and c-Myc to inhibit CRC proliferation and metastasis both in vitro and in vivo. These discoveries introduce potential molecular targets for CRC treatment and shed light on new mechanisms for c-Myc regulation in tumours.
Collapse
Affiliation(s)
- Xiao-Jing Ding
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Xue-Mei Cai
- Huabei Petroleum Administration Bureau General Hospital, Renqiu 062550, PR China
| | - Qian-Qian Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300350, PR China
| | - Ning Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300350, PR China
| | - Wei-Long Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, PR China.
| | - Xiao-Nan Xi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300350, PR China.
| | - Ya-Xin Lu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300350, PR China; College of Chemistry, Nankai University, Tianjin 300350, PR China.
| |
Collapse
|
32
|
Wang J, Wang L, Zhang Y, Pan S, Lin Y, Wu J, Bu M. Design, Synthesis, and Anticancer Activity of Novel Enmein-Type Diterpenoid Derivatives Targeting the PI3K/Akt/mTOR Signaling Pathway. Molecules 2024; 29:4066. [PMID: 39274913 PMCID: PMC11396751 DOI: 10.3390/molecules29174066] [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: 07/27/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
The enmein-type diterpenoids are a class of anticancer ent-Kaurane diterpnoids that have received much attention in recent years. Herein, a novel 1,14-epoxy enmein-type diterpenoid 4, was reported in this project for the first time. A series of novel enmein-type diterpenoid derivatives were also synthesized and tested for anticancer activities. Among all the derivatives, compound 7h exhibited the most significant inhibitory effect against A549 cells (IC50 = 2.16 µM), being 11.03-folds better than its parental compound 4. Additionally, 7h exhibited relatively weak anti-proliferative activity (IC50 > 100 µM) against human normal L-02 cells, suggesting that it had excellent anti-proliferative selectivity for cancer cells. Mechanism studies suggested that 7h induced G0/G1 arrest and apoptosis in A549 cells by inhibiting the PI3K/AKT/mTOR pathway. This process was associated with elevated intracellular ROS levels and collapsed MMP. In summary, these data identified 7h as a promising lead compound that warrants further investigation of its anticancer properties.
Collapse
Affiliation(s)
- Jiafeng Wang
- College of Pathology, Qiqihar Medical University, Qiqihar 161006, China; (J.W.); (Y.Z.); (S.P.)
| | - Lu Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (L.W.); (Y.L.)
| | - Yingbo Zhang
- College of Pathology, Qiqihar Medical University, Qiqihar 161006, China; (J.W.); (Y.Z.); (S.P.)
| | - Siwen Pan
- College of Pathology, Qiqihar Medical University, Qiqihar 161006, China; (J.W.); (Y.Z.); (S.P.)
| | - Yu Lin
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (L.W.); (Y.L.)
| | - Jiale Wu
- College of Life and Health, Hainan University, Haikou 570228, China;
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (L.W.); (Y.L.)
| |
Collapse
|
33
|
Swain S, Narayan RK, Mishra PR. Unraveling the interplay: exploring signaling pathways in pancreatic cancer in the context of pancreatic embryogenesis. Front Cell Dev Biol 2024; 12:1461278. [PMID: 39239563 PMCID: PMC11374643 DOI: 10.3389/fcell.2024.1461278] [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: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Pancreatic cancer continues to be a deadly disease because of its delayed diagnosis and aggressive tumor biology. Oncogenes and risk factors are being reported to influence the signaling pathways involved in pancreatic embryogenesis leading to pancreatic cancer genesis. Although studies using rodent models have yielded insightful information, the scarcity of human pancreatic tissue has made it difficult to comprehend how the human pancreas develops. Transcription factors like IPF1/PDX1, HLXB9, PBX1, MEIS, Islet-1, and signaling pathways, including Hedgehog, TGF-β, and Notch, are directing pancreatic organogenesis. Any derangements in the above pathways may lead to pancreatic cancer. TP53: and CDKN2A are tumor suppressor genes, and the mutations in TP53 and somatic loss of CDKN2A are the drivers of pancreatic cancer. This review clarifies the complex signaling mechanism involved in pancreatic cancer, the same signaling pathways in pancreas development, the current therapeutic approach targeting signaling molecules, and the mechanism of action of risk factors in promoting pancreatic cancer.
Collapse
Affiliation(s)
- Sashikanta Swain
- Department of Anatomy, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Ravi Kant Narayan
- Department of Anatomy, All India Institute of Medical Sciences, Bhubaneswar, India
| | | |
Collapse
|
34
|
Joseph AG, Biji M, Murali VP, Sherin DR, Valsan A, Sukumaran VP, Radhakrishnan KV, Maiti KK. A comprehensive apoptotic assessment of niloticin in cervical cancer cells: a tirucallane-type triterpenoid from Aphanamixis polystachya (Wall.) Parker. RSC Med Chem 2024:d4md00318g. [PMID: 39246746 PMCID: PMC11378019 DOI: 10.1039/d4md00318g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Pharmacologically active small organic molecules derived from natural resources are prominent drug candidates due to their inherent structural diversity. Herein, we explored one such bioactive molecule, niloticin, which is a tirucallane-type triterpenoid isolated from the stem barks of Aphanamixis polystachya (Wall.) Parker. After initial screening with other isolated compounds from the same plant, niloticin demonstrated selective cytotoxicity against cervical cancer cells (HeLa) with an IC50 value of 11.64 μM. Whereas the compound exhibited minimal cytotoxicity in normal epithelial cell line MCF-10A, with an IC50 value of 83.31 μM. Subsequently, in silico molecular docking studies of niloticin based on key apoptotic proteins such as p53, Fas, FasL, and TNF β revealed striking binding affinity, reflecting docking scores of -7.2, -7.1, -6.8, and -7.2. Thus, the binding stability was evaluated through molecular dynamic simulation. In a downstream process, the apoptotic capability of niloticin was effectively validated through in vitro fluorimetric assays, encompassing nuclear fragmentation. Additionally, an insightful approach involving surface-enhanced Raman spectroscopy (SERS) re-establishes the occurrence of DNA cleavage during cellular apoptosis. Furthermore, niloticin was observed to induce apoptosis through both intrinsic and extrinsic pathways. This was evidenced by the upregulation of upstream regulatory molecules such as CD40 and TNF, which facilitate the activation of caspase 8. Concurrently, niloticin-induced p53 activation augmented the expression of proapoptotic proteins Bax and Bcl-2 and downregulation of IAPs, leading to the release of cytochrome C and subsequent activation of caspase 9. Therefore, the reflection of mitochondrial-mediated apoptosis is in good agreement with molecular docking studies. Furthermore, the anti-metastatic potential was evidenced by wound area closure and Ki67 expression patterns. This pivotal in vitro assessment confirms the possibility of niloticin being a potent anti-cancer drug candidate, and to the best of our knowledge, this is the first comprehensive anticancer assessment of niloticin in HeLa cells.
Collapse
Affiliation(s)
- Anuja Gracy Joseph
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Mohanan Biji
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Vishnu Priya Murali
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
| | - Daisy R Sherin
- School of Digital Sciences, Kerala University of Digital Sciences, Innovation and Technology Thiruvananthapuram-695317 India
| | - Alisha Valsan
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Vimalkumar P Sukumaran
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Kokkuvayil Vasu Radhakrishnan
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
| | - Kaustabh Kumar Maiti
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
| |
Collapse
|
35
|
Dalavaye N, Nicholas M, Pillai M, Erridge S, Sodergren MH. The Clinical Translation of α-humulene - A Scoping Review. PLANTA MEDICA 2024; 90:664-674. [PMID: 38626911 PMCID: PMC11254484 DOI: 10.1055/a-2307-8183] [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: 09/30/2023] [Accepted: 04/16/2024] [Indexed: 07/19/2024]
Abstract
α-humulene, a sesquiterpene found in essential oils of various plant species, has garnered interest due to its potential therapeutic applications. This scoping review aims to consolidate α-humulene's evidence base, informing clinical translation, and guiding future research directions. A scoping review was conducted of EMBASE, MEDLINE, and PubMed databases up to 14th July 2023. All studies describing original research on α-humulene extraction, as well as pre-clinical and clinical research, were included for review. Three hundred and forty articles were analysed. α-humulene yields ranged from negligible to 60.90% across plant species. In vitro experiments demonstrated cytotoxicity against adenocarcinomas (such as colorectal, pulmonary, breast, prostatic, lung, and ovarian), with varying responses in other cell models. Mechanistic insights revealed its involvement in mitochondrial dysfunction, diminished intracellular glutathione levels, and the induction of oxidative stress. In rodent studies, oral administration of α-humulene at 50 mg/kg reduced inflammation markers in paw oedema and ovalbumin-induced airway inflammation. Intraperitoneal administration of α-humulene (50 - 200 mg/kg) exhibited cannabimimetic properties through cannabinoid 1 and adenosine A2a receptors. α-humulene also exhibited a multitude of properties with potential scope for therapeutic utilisation. However, there is a paucity of studies that have successfully translated this research into clinical populations with the associated disease. Potential barriers to clinical translation were identified, including yield variability, limited isolation studies, and challenges associated with terpene bioavailability. Consequently, rigorous pharmacokinetic studies and further mechanistic investigations are warranted to effectively uncover the potential of α-humulene.
Collapse
Affiliation(s)
- Nishaanth Dalavaye
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
| | - Martha Nicholas
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
| | - Manaswini Pillai
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
| | - Simon Erridge
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
- Curaleaf Clinic, London, UK
| | - Mikael H. Sodergren
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, UK
- Curaleaf International, London, UK
| |
Collapse
|
36
|
Kaur C, Sahu SK, Bansal K, DeLiberto LK, Zhang J, Tewari D, Bishayee A. Targeting Peroxisome Proliferator-Activated Receptor-β/δ, Reactive Oxygen Species and Redox Signaling with Phytocompounds for Cancer Therapy. Antioxid Redox Signal 2024; 41:342-395. [PMID: 38299535 DOI: 10.1089/ars.2023.0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Significance: Peroxisome proliferator-activated receptors (PPARs) have a moderately preserved amino-terminal domain, an extremely preserved DNA-binding domain, an integral hinge region, and a distinct ligand-binding domain that are frequently encountered with the other nuclear receptors. PPAR-β/δ is among the three nuclear receptor superfamily members in the PPAR group. Recent Advances: Emerging studies provide an insight on natural compounds that have gained increasing attention as potential anticancer agents due to their ability to target multiple pathways involved in cancer development and progression. Critical Issues: Modulation of PPAR-β/δ activity has been suggested as a potential therapeutic strategy for cancer management. This review focuses on the ability of bioactive phytocompounds to impact reactive oxygen species (ROS) and redox signaling by targeting PPAR-β/δ for cancer therapy. The rise of ROS in cancer cells may play an important part in the initiation and progression of cancer. However, excessive levels of ROS stress can also be toxic to the cells and cancer cells with increased oxidative stress are likely to be more vulnerable to damage by further ROS insults induced by exogenous agents, such as phytocompounds and therapeutic agents. Therefore, redox modulation is a way to selectively kill cancer cells without causing significant toxicity to normal cells. However, use of antioxidants together with cancer drugs may risk the effect of treatment as both act through opposite mechanisms. Future Directions: It is advisable to employ more thorough and detailed methodologies to undertake mechanistic explorations of numerous phytocompounds. Moreover, conducting additional clinical studies is recommended to establish optimal dosages, efficacy, and the impact of different phytochemicals on PPAR-β/δ.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sanjeev Kumar Sahu
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Keshav Bansal
- Department of Pharmaceutics, Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Lindsay K DeLiberto
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| |
Collapse
|
37
|
Chauhan A, Pathak VM, Yadav M, Chauhan R, Babu N, Chowdhary M, Ranjan A, Mathkor DM, Haque S, Tuli HS, Ramniwas S, Yadav V. Role of ursolic acid in preventing gastrointestinal cancer: recent trends and future perspectives. Front Pharmacol 2024; 15:1405497. [PMID: 39114347 PMCID: PMC11303223 DOI: 10.3389/fphar.2024.1405497] [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: 03/23/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Gastrointestinal malignancies are one of the major worldwide health concerns. In the present review, we have assessed the plausible therapeutic implication of Ursolic Acid (UA) against gastrointestinal cancer. By modulating several signaling pathways critical in cancer development, UA could offer anti-inflammatory, anti-proliferative, and anti-metastatic properties. However, being of low oral bioavailability and poor permeability, its clinical value is restricted. To deliver and protect the drug, liposomes and polymer micelles are two UA nanoformulations that can effectively increase medicine stability. The use of UA for treating cancers is safe and appropriate with low toxicity characteristics and a predictable pharmacokinetic profile. Although the bioavailability of UA is limited, its nanoformulations could emerge as an alternative to enhance its efficacy in treating GI cancers. Further optimization and validation in the clinical trials are necessary. The combination of molecular profiling with nanoparticle-based drug delivery technologies holds the potential for bringing UA to maximum efficacy, looking for good prospects with GI cancer treatment.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, Uttar Pradesh, India
| | | | - Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Neelesh Babu
- Department of Microbiology, Baba Farid Institute of Technology, Dehradun, Uttarakhand, India
| | - Manish Chowdhary
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Ambala, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Mohali, India
| | - Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
38
|
Elzanaty KA, Omran GA, Elmahallawy EK, Albrakati A, Saleh AA, Dahran N, Alhegaili AS, Salahuddin A, Abd-El-Azim H, Noreldin A, Okda TM. Design and Optimization of Sesamol Nanosuspensions to Potentiate the Anti-Tumor Activity of Epirubicin against Ehrlich Solid Carcinoma-Bearing Mice. Pharmaceutics 2024; 16:937. [PMID: 39065634 PMCID: PMC11279961 DOI: 10.3390/pharmaceutics16070937] [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: 06/03/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
There is a growing interest in discovering natural sources of anti-cancer drugs. Sesamol (SES) is a phenolic compound with antitumor effects. The present study aimed to investigate the anticancer properties of SES and its nano-suspensions (SES-NS) combined with Epirubicin (EPI) in breast cancer (BC) using mice bearing a solid Ehrlich tumor. The study involved 35 female albino mice and investigated the effects of SES and EPI on tumor growth, proliferation, apoptosis, autophagy, angiogenesis, and oxidative stress. Methods including ELISA, qRT-PCR, and immunohistochemistry were utilized. The findings revealed reductions in tumor growth and proliferation using SES either alone or combined and evidenced by decreased AKT (AKT Serine/Threonine kinase1) levels, angiogenesis indicated by lower levels of VEGFR (vascular endothelial growth factor), and apoptosis demonstrated by elevated caspase3 and BAX levels. Furthermore, autophagy increased and was indicated by increased levels of beclin1 and lc3, along with decreased oxidative stress as evidenced by elevated TAC (total antioxidant capacity) and reduced MDA (malondialdehyde) levels. Interestingly, SES-NS demonstrated more significant effects at lower doses. In summary, this study underscores the potential of SES as a promising agent for BC treatment. Moreover, SES-NS potentiated the beneficial effects of EPI while mitigating its adverse effects.
Collapse
Affiliation(s)
- Kholoud A. Elzanaty
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt (T.M.O.)
| | - Gamal A. Omran
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt (T.M.O.)
| | - Ehab Kotb Elmahallawy
- Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), Departamento de Sanidad Animal, Universidad de Córdoba, 14071 Córdoba, Spain
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Ayman A. Saleh
- Department of Pathology, College of Medicine, University of Hail, Hail 55428, Saudi Arabia;
| | - Naief Dahran
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah 21959, Saudi Arabia
| | - Alaa S. Alhegaili
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ahmad Salahuddin
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt (T.M.O.)
- Department of Biochemistry, College of Pharmacy, Al-Ayen Iraqi University, Nasiriyah 64001, Iraq
| | - Heba Abd-El-Azim
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt;
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ahmed Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Tarek M. Okda
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt (T.M.O.)
| |
Collapse
|
39
|
Li Y, Xu C, Han H, Pascual-Sabater S, Fillat C, Goel A. Aronia Berry Extract Modulates MYD88/NF-kB/P-Glycoprotein Axis to Overcome Gemcitabine Resistance in Pancreatic Cancer. Pharmaceuticals (Basel) 2024; 17:911. [PMID: 39065761 PMCID: PMC11279572 DOI: 10.3390/ph17070911] [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: 06/09/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with poor survival rates, primarily due to the limited effectiveness of gemcitabine (Gem)-based chemotherapy, as well as the acquisition of chemotherapeutic resistance. Aronia berry extracts (ABEs), abundant in phenolic constituents, have been recently recognized for their anticancer properties as well as their encouraging potential to help overcome chemoresistance in various cancers. In the present study, we explored ABE's potential to overcome Gem resistance in PDAC and identify specific growth regulatory pathways responsible for its anticancer activity. Through a series of in vitro experiments in gemcitabine-resistant (Gem-R) cells, we elucidated the synergistic interactions between Gem and ABE treatments. Using advanced transcriptomic analysis and network pharmacology, we revealed key molecular pathways linked to chemoresistance and potential therapeutic targets of ABE in Gem-R PDAC cells. Subsequently, the findings from cell culture studies were validated in patient-derived 3D tumor organoids (PDOs). The combination treatment of ABE and Gem demonstrated significant synergism and anticancer effects on cell viability, proliferation, migration, and invasion in Gem-R cells. Transcriptomic analysis revealed a correlation between the NF-Κb signaling pathway and Gem-R (p < 0.05), exhibiting a marked upregulation of MYD88. Additionally, MYD88 exhibited a significant correlation with the overall survival rates in patients with PDAC patients in the TCGA cohort (HR = 1.58, p < 0.05). The MYD88/NF-Κb pathway contributes to chemoresistance by potentially upregulating efflux transporters like P-glycoprotein (P-gp). Our findings revealed that the combined treatment with ABE suppressed the NF-Κb pathway by targeting MYD88 and reducing P-gp expression to overcome Gem resistance. Lastly, the combination therapy proved highly effective in PDOs in reducing both their number and size (p < 0.05). Our study offers previously unrecognized insights into the ability of ABE to overcome Gem resistance in PDAC cells through its targeting of the MYD88/NF-κb/P-gp axis, hence providing a safe and cost-effective adjunctive therapeutic strategy to improve treatment outcomes in PDAC.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA; (Y.L.); (C.X.)
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA; (Y.L.); (C.X.)
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116004, China
| | - Haiyong Han
- Division of Molecular Medicine, The Translational Genomics Research Institute, Phoenix, AZ 85004, USA;
| | - Silvia Pascual-Sabater
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-S.); (C.F.)
| | - Cristina Fillat
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-S.); (C.F.)
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA; (Y.L.); (C.X.)
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
40
|
Drif AI, Yücer R, Damiescu R, Ali NT, Abu Hagar TH, Avula B, Khan IA, Efferth T. Anti-Inflammatory and Cancer-Preventive Potential of Chamomile ( Matricaria chamomilla L.): A Comprehensive In Silico and In Vitro Study. Biomedicines 2024; 12:1484. [PMID: 39062057 PMCID: PMC11275008 DOI: 10.3390/biomedicines12071484] [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: 03/01/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIM Chamomile tea, renowned for its exquisite taste, has been appreciated for centuries not only for its flavor but also for its myriad health benefits. In this study, we investigated the preventive potential of chamomile (Matricaria chamomilla L.) towards cancer by focusing on its anti-inflammatory activity. METHODS AND RESULTS A virtual drug screening of 212 phytochemicals from chamomile revealed β-amyrin, β-eudesmol, β-sitosterol, apigenin, daucosterol, and myricetin as potent NF-κB inhibitors. The in silico results were verified through microscale thermophoresis, reporter cell line experiments, and flow cytometric determination of reactive oxygen species and mitochondrial membrane potential. An oncobiogram generated through comparison of 91 anticancer agents with known modes of action using the NCI tumor cell line panel revealed significant relationships of cytotoxic chamomile compounds, lupeol, and quercetin to microtubule inhibitors. This hypothesis was verified by confocal microscopy using α-tubulin-GFP-transfected U2OS cells and molecular docking of lupeol and quercetin to tubulins. Both compounds induced G2/M cell cycle arrest and necrosis rather than apoptosis. Interestingly, lupeol and quercetin were not involved in major mechanisms of resistance to established anticancer drugs (ABC transporters, TP53, or EGFR). Performing hierarchical cluster analyses of proteomic expression data of the NCI cell line panel identified two sets of 40 proteins determining sensitivity and resistance to lupeol and quercetin, further pointing to the multi-specific nature of chamomile compounds. Furthermore, lupeol, quercetin, and β-amyrin inhibited the mRNA expression of the proinflammatory cytokines IL-1β and IL6 in NF-κB reporter cells (HEK-Blue Null1). Moreover, Kaplan-Meier-based survival analyses with NF-κB as the target protein of these compounds were performed by mining the TCGA-based KM-Plotter repository with 7489 cancer patients. Renal clear cell carcinomas (grade 3, low mutational rate, low neoantigen load) were significantly associated with shorter survival of patients, indicating that these subgroups of tumors might benefit from NF-κB inhibition by chamomile compounds. CONCLUSION This study revealed the potential of chamomile, positioning it as a promising preventive agent against inflammation and cancer. Further research and clinical studies are recommended.
Collapse
Affiliation(s)
- Assia I. Drif
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Roxana Damiescu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Nadeen T. Ali
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Tobias H. Abu Hagar
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Bharati Avula
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Ikhlas A. Khan
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| |
Collapse
|
41
|
Chauhan R, Malhotra L, Gupta A, Dagar G, Mendiratta M, Masoodi T, Hashem S, Al Marzooqi S, Das D, Uddin S, Ethayathulla AS, Macha MA, Akil AAS, Sahoo RK, Rai E, Bhat AA, Singh M. Bergenin inhibits growth of human cervical cancer cells by decreasing Galectin-3 and MMP-9 expression. Sci Rep 2024; 14:15287. [PMID: 38961106 PMCID: PMC11222472 DOI: 10.1038/s41598-024-64781-3] [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: 12/04/2023] [Accepted: 06/13/2024] [Indexed: 07/05/2024] Open
Abstract
Cervical cancer is still the leading cause of cancer mortality worldwide even after introduction of vaccine against Human papillomavirus (HPV), due to low vaccine coverage, especially in the developing world. Cervical cancer is primarily treated by Chemo/Radiotherapy, depending on the disease stage, with Carboplatin/Cisplatin-based drug regime. These drugs being non-specific, target rapidly dividing cells, including normal cells, so safer options are needed for lower off-target toxicity. Natural products offer an attractive option compared to synthetic drugs due to their well-established safety profile and capacity to target multiple oncogenic hallmarks of cancer like inflammation, angiogenesis, etc. In the current study, we investigated the effect of Bergenin (C-glycoside of 4-O-methylgallic acid), a natural polyphenol compound that is isolated from medicinal plants such as Bergenia crassifolia, Caesalpinia digyna, and Flueggea leucopyrus. Bergenin has been shown to have anti-inflammatory, anti-ulcerogenic, and wound healing properties but its anticancer potential has been realized only recently. We performed a proteomic analysis of cervical carcinoma cells treated with bergenin and found it to influence multiple hallmarks of cancers, including apoptosis, angiogenesis, and tumor suppressor proteins. It was also involved in many different cellular processes unrelated to cancer, as shown by our proteomic analysis. Further analysis showed bergenin to be a potent-angiogenic agent by reducing key angiogenic proteins like Galectin 3 and MMP-9 (Matrix Metalloprotease 9) in cervical carcinoma cells. Further understanding of this interaction was carried out using molecular docking analysis, which indicated MMP-9 has more affinity for bergenin as compared to Galectin-3. Cumulatively, our data provide novel insight into the anti-angiogenic mechanism of bergenin in cervical carcinoma cells by modulation of multiple angiogenic proteins like Galectin-3 and MMP-9 which warrant its further development as an anticancer agent in cervical cancer.
Collapse
Affiliation(s)
- Ravi Chauhan
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Lakshay Malhotra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Gunjan Dagar
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Mohini Mendiratta
- Department of Medical Oncology, Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Sara Al Marzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Dayasagar Das
- Department of Medicine, NYU Langone Health, New York, 10016, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Ammira Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Ekta Rai
- School of Life Sciences Jawahar Lal Nehru University, New Delhi, India
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India.
| |
Collapse
|
42
|
Sachdeva S, Sarethy IP. Diving into freshwater microbial metabolites: Pioneering research and future prospects. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024:1-19. [PMID: 38887995 DOI: 10.1080/09603123.2024.2351153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
43
|
de Lima E Souza Mesquita GC, Da Cruz ER, Corrêa DS, de Barros Falcão Ferraz A, Miri JM, Farias IV, Reginatto FH, Boaretto FBM, Dos Santos DM, da Silva J, Grivicich I, Picada JN. Genotoxic and antiproliferative properties of Endopleura uchi bark aqueous extract. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:516-531. [PMID: 38619152 DOI: 10.1080/15287394.2024.2340069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The bark extract from Endopleura uchi has been widely used in traditional medicine to treat gynecological-related disorders, diabetes, and dyslipidemias albeit without scientific proof. In addition, E. uchi bark extract safety, especially regarding mutagenic activities, is not known. The aim of this study was to determine the chemical composition, antitumor, and toxicological parameters attributed to an E. uchi bark aqueous extract. The phytochemical constitution was assessed by colorimetric and chromatographic analyzes. The antiproliferative effect was determined using sulforhodamine B (SRB) assay using 4 cancer cell lines. Cytotoxic and genotoxic activities were assessed utilizing MTT and comet assays, respectively, while mutagenicity was determined through micronucleus and Salmonella/microsome assays. The chromatographic analysis detected predominantly the presence of gallic acid and isoquercitrin. The antiproliferative effect was more pronounced in human colon adenocarcinoma (HT-29) and human breast cancer (MCF-7) cell lines. In the MTT assay, the extract presented an IC50 = 39.1 µg/ml and exhibited genotoxic (comet assay) and mutagenic (micronucleus test) activities at 20 and 40 µg/ml in mouse fibroblast cell line (L929) and mutagenicity in the TA102 and TA97a strains in the absence of S9 mix. Data demonstrated that E. uchi bark possesses bioactive compounds which exert cytotoxic and genotoxic effects that might be associated with its antitumor potential. Therefore, E. uchi bark aqueous extract consumption needs to be approached with caution in therapeutic applications.
Collapse
Affiliation(s)
| | - Elkejer Ribeiro Da Cruz
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
- Center for Research in Product and Development (CEPPED), Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Dione Silva Corrêa
- Center for Research in Product and Development (CEPPED), Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Alexandre de Barros Falcão Ferraz
- Pharmacognosy Laboratory, Department of Industrial Pharmaceutical, Health Sciences Center, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Jéssica Machado Miri
- Laboratory of Cancer Biology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Ingrid Vicente Farias
- Pharmacognosy Laboratory, Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Flávio Henrique Reginatto
- Pharmacognosy Laboratory, Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | | | - Duani Maria Dos Santos
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | - Juliana da Silva
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
- Laboratory of Genetics Toxicology, La Salle University, Canoas, RS, Brazil
| | - Ivana Grivicich
- Laboratory of Cancer Biology, Lutheran University of Brazil (ULBRA), Canoas, RS, Brazil
| | | |
Collapse
|
44
|
Afarin R, Ahmadpour F, Hatami M, Monjezi S, Igder S. Combination of Etoposide and quercetin-loaded solid lipid nanoparticles Potentiates apoptotic effects on MDA-MB-231 breast cancer cells. Heliyon 2024; 10:e31925. [PMID: 38841445 PMCID: PMC11152947 DOI: 10.1016/j.heliyon.2024.e31925] [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: 12/30/2023] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Background Breast cancer is a major global cancer, for which radiation and chemotherapy are the main treatments. Natural remedies are being studied to reduce the side effects. Etoposide (ETO), a chemo-drug, and quercetin (QC), a phytochemical, are considered potential factors for adaptation to conventional treatments. Objectives The anticancer effect of the synergy between ETO and Quercetin-loaded solid lipid nanoparticles (QC-SLNs), was investigated in MDA-MB-231 cells. Methods We developed QC-SLNs for efficient cellular delivery, characterizing their morphology, particle size, and zeta potential. We assessed the cytotoxicity of QC-SLNs and ETO on breast cancer cells via the MTT assay. Effects on apoptosis intensity in MDA-MB-231 cells have been detected utilizing annexin V-FITC, PI, and caspase activities. Real-time PCR assessed Bax gene and Bcl-2 gene fold change expression, while Western blot analysis determined p53 and p21 protein levels. Results Spherical, negatively charged QC-SLNs, when combined with ETO, significantly enhanced inhibition of MDA-MB-231 cell proliferation compared to ETO or QC-SLNs alone. The combined treatment also notably increased the apoptosis pathway. QC-SLNs + ETO increased the Bax/Bcl-2 gene ratio, elevated p53 and p21 proteins, and activated caspase 3 and 9 enzymes. These results indicate the potential for QC-SLNs + ETO as a strategy for breast cancer treatment, potentially overcoming ETO-resistant breast cancer chemoresistance. Conclusion These results suggest that QC-SLN has the potential to have a substantial impact on the breast cancer cure by improving the efficacy of ETO. This enhancement could potentially help overcome chemoresistance observed in ETO-resistant breast cancer.
Collapse
Affiliation(s)
- Reza Afarin
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Ahmadpour
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mahdi Hatami
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Monjezi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Igder
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
45
|
Kalsoom A, Altaf A, Sattar H, Maqbool T, Sajjad M, Jilani MI, Shabbir G, Aftab S. Gene expression and anticancer evaluation of Kigelia africana (Lam.) Benth. Extracts using MDA-MB-231 and MCF-7 cell lines. PLoS One 2024; 19:e0303134. [PMID: 38837975 DOI: 10.1371/journal.pone.0303134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/16/2024] [Indexed: 06/07/2024] Open
Abstract
In recent years, a cancer research trend has shifted towards identifying novel therapeutic compounds from natural assets for the management of cancer. In this study, we aimed to assess the cytotoxic activity of Kigelia Africana (KA) extracts on breast cancer (MDA-MB-231 and MCF-7) and noncancerous kidney cells (HEK-293T) to develop an efficient anticancer medication. We used gas chromatography mass spectrometry (GC-MS to analyze the constituents of EKA and HKA extracts meanwhile the crystal violet and the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) assays were used to examine the possible cytotoxic effects of plant extracts on our cancer cell lines along with non-cancerous control. The quantitative real-time PCR (RT-PCR) was run on cell samples to evaluate the differential expression of cell proliferative markers of cancer (BCL-2 and TP53). These phytochemicals have been reported to have binding affinity for some other growth factors and receptors as well which was evaluated by the in-silico molecular docking against Bcl2, EGFR, HER2, and TP53. Our Morphological observation showed a significant difference in the cell morphology and proliferation potential which was decreased under the effect of plant extracts treatment as compared to the control samples. The ethanol extract exhibited a marked antiproliferative activity towards MDA-MB-231 and MCF-7 cell lines with IC50 = 20 and 32 μg/mL, respectively. Quantitative RT-PCR gene expression investigation revealed that the IC50 concentration of ethanolic extract regulated the levels of mRNA expression of apoptotic genes. With the target and active binding site amino acids discovered in the molecular docking investigation, TP53/Propanoic acid, 3-(2, 3, 6-trimethyl-1, 4-dioxaspiro [4.4] non-7-yl)-, methyl ester (-7.1 kcal/mol) is the best-docked ligand. The use of this plant in folk remedies justifies its high in vitro anti-cancer capabilities. This work highlights the role of phytochemicals in the inhibition of cancer proliferation. Based on all these findings, it can be concluded that EKA extract has promising anti-proliferative effect on cancerous cells but more study is required in future to further narrow down the active ingredients of total crude extract with specific targets in cancer cells.
Collapse
Affiliation(s)
- Aasia Kalsoom
- Institute of Molecular Biology (IMBB), Center for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Awais Altaf
- Institute of Molecular Biology (IMBB), Center for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Huma Sattar
- Institute of Molecular Biology (IMBB), Center for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Tahir Maqbool
- Institute of Molecular Biology (IMBB), Center for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Muhammad Sajjad
- School of Biological Sciences, Punjab University, Lahore, Pakistan
| | | | - Ghulam Shabbir
- Pakistan Council of Scientific and Industrial Research (PCSIR), Islamabad, Pakistan
| | - Saira Aftab
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
46
|
Siddique R, Thangavelu L, S R, Almalki WH, Kazmi I, Kumar A, Mahajan S, Kalra H, Alzarea SI, Pant K. lncRNAs and cyclin-dependent kinases: Unveiling their critical roles in cancer progression. Pathol Res Pract 2024; 258:155333. [PMID: 38723325 DOI: 10.1016/j.prp.2024.155333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024]
Abstract
Long non-coding RNAs (lncRNAs) are a diverse class of RNA molecules that do not code for proteins but play critical roles in gene regulation. One such role involves the modulation of cell cycle progression and proliferation through interactions with cyclin-dependent kinases (CDKs), key regulators of cell division. Dysregulation of CDK activity is a hallmark of cancer, contributing to uncontrolled cell growth and tumor formation. These lncRNA-CDK interactions are part of a complex network of molecular mechanisms underlying cancer pathogenesis, involving various signaling pathways and regulatory circuits. Understanding the interplay between lncRNAs, CDKs, and cancer biology holds promise for developing novel therapeutic strategies targeting these molecular targets for more effective cancer treatment. Furthermore, targeting CDKs, key cell cycle progression and proliferation regulators, offers another avenue for disrupting cancer pathways and overcoming drug resistance. This can open new possibilities for individualized treatment plans and focused therapeutic interventions.
Collapse
Affiliation(s)
- Raihan Siddique
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Lakshmi Thangavelu
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India.
| | - RenukaJyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Ashwani Kumar
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Shriya Mahajan
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140417, India
| | - Hitesh Kalra
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh 174103, India
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Kumud Pant
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India; Graphic Era Hill University, Clement Town, Dehradun 248002, India
| |
Collapse
|
47
|
Haque S, Hussain A, Almalki AH, Aldawsari MF, Lal B, Rai AK, Srivastava M, Fiołka M. Prospects of earthworm coelomic fluid as a potential therapeutic agent to treat cancer. Cancer Metastasis Rev 2024; 43:621-637. [PMID: 37910294 DOI: 10.1007/s10555-023-10148-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023]
Abstract
Cancer is a major public health concern because it is one of the main causes of morbidity and mortality worldwide. As a result, numerous studies have reported the development of new therapeutic compounds with the aim of selectively treating cancer while having little negative influence on healthy cells. In this context, earthworm coelomic fluid has been acknowledged as a rich source of several bioactive substances that may exhibit promising anticancer activity. Therefore, the objective of the present review is to evaluate the findings of the reported studies exploring the antitumor effects of coelomic fluid in the context of its possible utilization as a natural therapeutic agent to cure different types of cancer. The possible mechanisms underlying the coelomic fluid's anticancerous potential as well as the possibility for future development of cutting-edge therapeutic agents utilizing coelomic fluid-derived natural bioactive compounds to treat cancer disorders have been discussed along with future challenges. In addition, the feasibility of encapsulation of bioactive compounds derived from coelomic fluid with nanomaterials that could be further explored to attain more effective anticancer competence is discussed.
Collapse
Affiliation(s)
- Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, P.O. Box 345050, Dubai, United Arab Emirates
| | - Atiah H Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
- Addiction and Neuroscience Research Unit, College of Pharmacy, Taif University, Al-Hawiah, Taif, 21944, Saudi Arabia
| | - Mohammed F Aldawsari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Basant Lal
- Department of Chemistry, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Ashutosh Kumar Rai
- Department of Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Manish Srivastava
- Department of Chemical Engineering & Technology, Indian Institute of Technology (BHU) Varanasi, Varanasi, Uttar Pradesh, 221005, India.
- LCB Fertilizers Pvt. Ltd., Shyam Vihar Phase 2, Rani Sati Mandir Road, Lachchhipur, Gorakhpur, Uttar Pradesh, 273015, India.
| | - Marta Fiołka
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland.
| |
Collapse
|
48
|
Aljeldah MM. Evaluation of the anticancer and antibacterial activities of moscatilin. Heliyon 2024; 10:e31131. [PMID: 38818150 PMCID: PMC11137398 DOI: 10.1016/j.heliyon.2024.e31131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024] Open
Abstract
Orchids (Dendrobium sp.) have been the subject of extensive research due to their ubiquitous pharmacological, antimicrobial, and anticancer properties. Moscatilin is a bibenzyl secondary metabolite enriched in orchids that exhibits anticancer and antimicrobial properties through mechanisms that have not yet been fully elucidated. The current study aimed to assess the in vitro anticancer and antibacterial potential of moscatilin. The in vitro anti-proliferative effects of moscatilin against breast cancer-MCF-7 and liver-HepG2 cells were assessed using the dimethylthiazol-diphenyltetrazolium bromide assay. Selected six pro-apoptotic (caspase-3, 8, 9, p53, p21 & Bax) and two anti-apoptotic (Bcl-xL & Bcl-2) gene markers were assessed via qPCR and tested antibacterial activity against various bacterial strains using disc diffusion and broth dilution methods. Moscatilin decreased the cellular viabilities of HepG2 and MCF-7 cancer cells, with anti-proliferation rates of 66 % (IC50 51 ± 5.18 μM) and 58 % (IC50 57 ± 4.18 μM), respectively. This effect was selectively observed in cancer cells, and the impact of moscatilin on non-cancerous MCF-12 cells was marginal. Moreover, moscatilin-treated cells exhibited higher mRNA levels of caspase-3,8, 9, Bax, p53, and p21, whereas lower levels of Bcl-2 and Bcl-xL, two anti-apoptotic markers, were observed. Furthermore, moscatilin exhibited varying degrees of antibacterial activity against the bacterial strains investigated. Notably, the highest antibacterial potentials were observed against Staphylococcus epidermidis and Klebsiella pneumonia, while the lowest inhibitory activity was observed in Escherichia coli and Pseudomonas aeruginosa. Overall, these findings demonstrated that moscatilin exerts potent anticancer effects via apoptosis and has antimicrobial properties against Gram-negative and Gram-positive bacteria that are clinically relevant. These findings highlight the potential of moscatilin as a natural therapeutic candidate for the treatment of cancer and clinically important bacterial pathogens.
Collapse
Affiliation(s)
- Mohammed Mubarak Aljeldah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin, 31991, Saudi Arabia
| |
Collapse
|
49
|
Saikia L, Gogoi B, Sen S, Tonk RK, Kumar D, Dutta PP. The recent update and advancements of natural products in targeting the Wnt/β-Catenin pathway for cancer prevention and therapeutics. Med Oncol 2024; 41:164. [PMID: 38816663 DOI: 10.1007/s12032-024-02387-5] [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: 03/13/2024] [Accepted: 04/19/2024] [Indexed: 06/01/2024]
Abstract
The Wnt/β-Catenin pathway (Wnt/β-CatP) is implicated in accelerating carcinogenesis and cancer progression, contributing to increased morbidity and treatment resistance. Even though it holds promise as a focus for cancer treatment, its intricate nature and diverse physiological effects pose significant challenges. Recent years have witnessed significant advancements in this domain, with numerous natural products demonstrating promising preclinical anti-tumor effects and identified as inhibitors of the Wnt/β-CatP through various upstream and downstream mechanisms. This study provides a comprehensive overview of the current landscape of Wnt/β-Cat-targeted cancer therapy, examining the impact of natural products on Wnt/β-Cat signaling in both cancer prevention and therapeutic contexts. A comprehensive search was conducted on scientific databases like SciFinder, PubMed, and Google Scholar to retrieve relevant literature on Wnt-signaling, natural products, β-Catenin (β-Cat), and cancer from 2020 to January 2024. As per the analysis of the relevant reference within the specified period, it has been noted that a total of 58 phytoconstituents, predominantly phenolics, followed by triterpenoids and several other classes, along with a limited number of plant extracts, have exhibited activity targeting the Wnt/β-CatP. Most β-Cat regulating modulators restrict cancer cell development by suppressing β-Cat expression, facilitating proteasomal degradation, and inhibiting nuclear translocation. Multiple approaches have been devised to block the activity of β-Cat in cancer therapy, a key factor in cancer progression, leading to the discovery of various Wnt/β-CatP regulators. However, their exploration remains limited, necessitating further research using clinical models for potential clinical use in cancer prevention and therapeutics.
Collapse
Affiliation(s)
- Lunasmrita Saikia
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India
| | - Bhaskarjyoti Gogoi
- Department of Biotechnology, The Assam Royal Global University, Guwahati, Assam, 781035, India
| | - Saikat Sen
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India
| | - Rajiv K Tonk
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India.
| |
Collapse
|
50
|
Üremis N, Türköz Y, Üremiş MM, Çiğremiş Y, Şalva E. RETRACTED ARTICLE: Investigating EGFR-VEGF-mediated apoptotic effect of cucurbitacin D and I combination with sorafenib via Ras/Raf/MEK/ERK and PI3K/Akt signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3247. [PMID: 37917368 DOI: 10.1007/s00210-023-02811-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Affiliation(s)
- Nuray Üremis
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey.
| | - Yusuf Türköz
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yılmaz Çiğremiş
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Emine Şalva
- Department of Pharmacy Technology, Pharmacy Faculty, Inonu University, Malatya, Turkey
| |
Collapse
|