1
|
Konappa N, Patil RH, Kariyappa AS, Krishnamurthy S, Ramachandrappa NS, Krishnappa R, Chowdappa S. Green synthesis of silver nanoparticles using Amomum nilgiricum leaf extracts: preparation, physicochemical characterization and ameliorative effect against human cancer cell lines. Cytotechnology 2025; 77:16. [PMID: 39669689 PMCID: PMC11631834 DOI: 10.1007/s10616-024-00674-7] [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: 10/03/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The present study to production of silver nanoparticles (AgNPs) by leaf extracts of A. nilgiricum and to evaluate the activity of anticancer by using AgNPs against cancer cell lines such as MCF-7, HEPG2, H9C2, HEK293 and H1975. The synthesized AgNPs were characterized by using UV-Vis spectroscopy, EDS, FT-IR, XRD, DLS, SEM and HRTEM with SAED patterns. The surface plasmon resonance (SPR) of AgNPs formed a peak centered at 427 nm by UV-Vis analysis. FTIR analysis reveals that existence of functional groups subjected to silver ions reduction to metallic silver. Crystalline form of the AgNPs was assessed by XRD analysis, four spectral peaks at 111, 200, 220, and 311 were formed and zeta potential peak was found at 28.5 mV indicating the higher stability. The size average diameter of the AgNPs was between 27 and 30 nm by TEM and SEM analysis was reveals the morphology of AgNPs as elongated, irregular and aggregated and some particles are spherical. EDX analysis confirmed the elemental composition of AgNPs with 81.43% Ag. The average diameter of AgNPs was found 21.49 nm in diameter and width was about 12.01 nm by DLS analysis. Cytotoxicity of AgNPs was investigated by using MTT, SRB assay and comet assay was performed as a genotoxicity. The results revealed that AgNPs decreased the viability of cancer cells in a concentration dependent pattern (50 to 350 µg/ml). The influence of AgNPs on cell cycle stop was studied on H1975, HEP-G2 and MCF-7 cells and found that AgNPs could induce sub G0 cell cycle arrest. The AgNPs was also induced DNA fragmentation confirms the DNA damage in nanoparticles treated cell lines. The anticancer action of nanoparticles was analyzed using proapoptotic and antiapoptotic caspase 8 and caspase 3 mRNA expression levels. Finally the results suggested that AgNPs is an effective anticancer agent which induces apoptosis in H1975, HEP-G2 and MCF-7 cells. Based on our studies, further identification of the major compounds of leaf extracts is acceptable. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00674-7.
Collapse
Affiliation(s)
- Narasimhamurthy Konappa
- Department of Microbiology and Biotechnology, Jnanabharathi Campus, Bangalore University, Bengaluru, Karnataka 560 056 India
| | - Rajeshwari H. Patil
- Department of Microbiology and Biotechnology, Jnanabharathi Campus, Bangalore University, Bengaluru, Karnataka 560 056 India
| | - Anupama S. Kariyappa
- Department of Microbiology and Biotechnology, Jnanabharathi Campus, Bangalore University, Bengaluru, Karnataka 560 056 India
| | - Soumya Krishnamurthy
- Department of Microbiology, Field Marshal K. M. Cariappa College, A Constituent College of Mangalore University, Madikeri, Karnataka 571201 India
| | | | - Rahul Krishnappa
- Department of Microbiology and Biotechnology, Jnanabharathi Campus, Bangalore University, Bengaluru, Karnataka 560 056 India
| | - Srinivas Chowdappa
- Department of Microbiology and Biotechnology, Jnanabharathi Campus, Bangalore University, Bengaluru, Karnataka 560 056 India
| |
Collapse
|
2
|
Zhou T, Niu Y, Li Y. Advances in research on malignant tumors and targeted agents for TOP2A (Review). Mol Med Rep 2025; 31:50. [PMID: 39670307 DOI: 10.3892/mmr.2024.13415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The DNA topoisomerase isoform topoisomerase IIα (TOP2A) is essential for the condensation and segregation of cellular mitotic chromosomes and the structural maintenance. It has been demonstrated that TOP2A is highly expressed in various malignancies, including lung adenocarcinoma (LUAD), hepatocellular carcinoma (HCC) and breast cancer (BC), associating with poor prognosis and aggressive tumor behavior. Additionally, TOP2A has emerged as a promising target for cancer therapy, with widespread clinical application of associated chemotherapeutic agents. The present study explored the impact of TOP2A on malignant tumor growth and the advancements in research on its targeted drugs. The fundamental mechanisms of TOP2A have been detailed, its specific roles in tumor cells are analyzed, and its potential as a biomarker for tumor prognosis and therapeutic targeting is highlighted. Additionally, the present review compiles findings from the latest clinical trials of relevant targeted agents, information on newly developed inhibitors, and discusses future research directions and clinical application strategies in cancer therapy, aiming to propose novel ideas and methods.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yiting Niu
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yanjun Li
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
3
|
Aldoghachi FEH, Oraibi A, Hamid Mohsen N, Hassan SS. Repurposing Phytochemicals against Breast Cancer (MCF-7) using Classical Structure-Based Drug Design. Curr Drug Discov Technol 2025; 22:e280324228430. [PMID: 38551041 DOI: 10.2174/0115701638295736240315105737] [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: 01/04/2024] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The significant public health effect of breast cancer is demonstrated by its high global prevalence and the potential for severe health consequences. The suppression of the proliferative effects facilitated by the estrogen receptor alpha (ERα) in the MCF-7 cell line is significant for breast cancer therapy. OBJECTIVE The current work involves in-silico techniques for identifying potential inhibitors of ERα. METHODS The method combines QSAR models based on machine learning with molecular docking to identify potential binders for the ERα. Further, molecular dynamics simulation studied the stability of the complexes, and ADMET analysis validated the compound's properties. RESULTS Two compounds (162412 and 443440) showed significant binding affinities with ERα, with binding energies comparable to the established binder RL4. The ADMET qualities showed advantageous characteristics resembling pharmaceutical drugs. The stable binding of these ligands in the active region of ERα during dynamic conditions was confirmed by molecular dynamics simulations. RMSD plots and conformational stability supported the ligands' persistent occupancy in the protein's binding site. After simulation, two hydrogen bonds were found within the protein-ligand complexes of 162412 and 443440, with binding free energy values of -27.32 kcal/mol and -25.00 kcal/mol. CONCLUSION The study suggests that compounds 162412 and 443440 could be useful for developing innovative anti-ERα medicines. However, more research is needed to prove the compounds' breast cancer treatment efficacy. This will help develop new treatments for ERα-associated breast cancer.
Collapse
Affiliation(s)
| | - Amjad Oraibi
- Department of Pharmacy, Al-Manara College for Medical Sciences, Amarah, Iraq
| | | | | |
Collapse
|
4
|
Nalla K, Chatterjee B, Poyya J, Swain A, Ghosh K, Pan A, Joshi CG, Manavathi B, Kanade SR. Epigallocatechin-3-gallate inhibit the protein arginine methyltransferase 5 and enhancer of Zeste homolog 2 in breast cancer both in vitro and in vivo. Arch Biochem Biophys 2025; 763:110223. [PMID: 39581340 DOI: 10.1016/j.abb.2024.110223] [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: 06/06/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE Histone methyltransferases are enzymes that selectively methylate lysine or arginine residues on both histone and non-histone proteins, categorized into lysine methyltransferases and arginine methyltransferases. Notably, EZH2 and PRMT5 are known for catalyzing trimethylation of H3 at K27 and symmetric dimethylation of H4 at R3, respectively. These methylation events are recognized as characteristic histone-repressive marks in cancer. The over expression of PRMT5 and EZH2 were reported in various cancers and recognized as a drug target. The study aims to explore the inhibitory potential of phytocompound, Epigallocatechin-3-gallate (EGCG), against PRMT5 and EZH2 in the breast cancer model. METHODS Screening of an array of phytocompounds was conducted through a combination of in-silico and in-vitro assays. Interactions between EGCG and human PRMT5: MEP50 and EZH2 were evaluated using molecular docking. Binding efficiency was validated, by Surface Plasmon Resonance studies and inhibitory potential was accessed by in vitro methylation followed by western blots, ELISA, and cell-based assays. In-vivo efficacy of EGCG was carried on cell line derived mice xenograft model. RESULTS EGCG demonstrated robust interactions with PRMT5:MEP50 complex and EZH2, particularly within the SAM binding site. Surface Plasmon Resonance analysis revealed strong binding affinity in nanomolar concentrations, particularly with PRMT5-MEP50 compared to EZH2. In-vitro assays confirmed EGCG's ability to inhibit PRMT5 and EZH2, leading to a decrease in their catalytic products, namely H4R3me2s and H3K27me3, respectively. EGCG treatment induced both autophagy and apoptosis invitro. In-vivo studies demonstrated significant reductions in tumor size and the proliferation marker ki67, accompanied by a decrease in histone repressive marks. CONCLUSION The findings suggest that EGCG effectively inhibits PRMT5 and EZH2, underscoring its potential for combined therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Kirankumar Nalla
- Department of Plant Sciences, School of Life Sciences University of Hyderabad PO Central University Gachibowli, Hyderabad Telangana, 500046, India
| | - Biji Chatterjee
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Centre, Houston, TX, USA
| | - Jagadeesha Poyya
- SDM Research Institute for Biomedical Sciences, A Constituent Unit of Shri Dharmasthala, Manjunatheshwara University, Dharwad, 580009, Karnataka, India
| | - Aishwarya Swain
- Department for Bioinformatics, Pondicherry University, Puducherry, 605014, India
| | - Krishna Ghosh
- Department of Anaesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Centre, Houston, TX, USA
| | - Archana Pan
- Department for Bioinformatics, Pondicherry University, Puducherry, 605014, India
| | - Chandrashekhar G Joshi
- Department of Studies in Biochemistry, Mangalore University PG Centre, Jnana Kaveri, Chikka Aluvara, Thorenoor Post Kushalnagar, Somawarpet TQ, Kodagu, 571232, India
| | - Bramanandam Manavathi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad PO Central University Gachibowli, Hyderabad Telangana 500046, India
| | - Santosh R Kanade
- Department of Plant Sciences, School of Life Sciences University of Hyderabad PO Central University Gachibowli, Hyderabad Telangana, 500046, India.
| |
Collapse
|
5
|
Joković N, Pešić S, Vitorović J, Bogdanović A, Sharifi-Rad J, Calina D. Glucosinolates and Their Hydrolytic Derivatives: Promising Phytochemicals With Anticancer Potential. Phytother Res 2024. [PMID: 39726346 DOI: 10.1002/ptr.8419] [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: 06/19/2024] [Revised: 10/29/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Recent research has increasingly focused on phytochemicals as promising anticancer agents, with glucosinolates (GSLs) and their hydrolytic derivatives playing a central role. These sulfur-containing compounds, found in plants of the Brassicales order, are converted by myrosinase enzymes into biologically active products, primarily isothiocyanates (ITCs) and indoles, which exhibit significant anticancer properties. Indole-3-carbinol, diindolylmethane, sulforaphane (SFN), phenethyl isothiocyanate (PEITC), benzyl isothiocyanate, and allyl isothiocyanate have shown potent anticancer effects in animal models, particularly in breast, prostate, lung, melanoma, bladder, hepatoma, and gastrointestinal cancers. Clinical studies further support the chemopreventive effects of SFN and PEITC, particularly in detoxifying carcinogens and altering biochemical markers in cancer patients. These compounds have demonstrated good bioavailability, low toxicity, and minimal adverse effects, supporting their potential therapeutic application. Their anticancer mechanisms include the modulation of reactive oxygen species, suppression of cancer-related signaling pathways, and direct interaction with tumor cell proteins. Additionally, semi-synthetic derivatives of GSLs have been developed to enhance anticancer efficacy. In conclusion, GSLs and their derivatives offer significant potential as both chemopreventive and therapeutic agents, warranting further clinical investigation to optimize their application in cancer treatment.
Collapse
Affiliation(s)
- Nataša Joković
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Strahinja Pešić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Jelena Vitorović
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Andrija Bogdanović
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador
- Department of Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
6
|
Lakshmi S, Priya S. Molecular insights into epoxyazadiradione induced death in triple-negative breast cancer cells: A system biology approach. Gene 2024; 930:148814. [PMID: 39116958 DOI: 10.1016/j.gene.2024.148814] [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: 03/28/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Epoxyazadiradione is an important limonoid with immense pharmacological potential. We have reported previously that epoxyazadiradione (EAD) induces apoptosis in triple negative breast cancer cells (MDA-MB 231) by modulating diverse cellular targets. Here, we identify the key genes/pathways responsible for this effect through next-generation sequencing of the transcriptome from EAD treated cells and integrated molecular data analysis using bioinformatics. In silico analysis indicated that EAD displayed favourable drug-like properties and could target multiple macromolecules relevant to TNBC. RNA sequencing revealed that EAD treatment results in the differential expression of 1838 genes in MDA-MB 231 cells, with 752 downregulated and 1086 upregulated. Gene set enrichment analysis of these genes suggested that EAD disrupts protein folding in the endoplasmic reticulum, triggering the unfolded protein response (UPR) and potentially leading to cell death. EAD also induced oxidative stress and DNA damage, downregulated pathways linked to metabolism, cell cycle progression, pro-survival signalling, cell adhesion, motility and inflammatory response. The identification of protein cluster and hub genes were also done. The validation of the identified hub genes gave an inverse correlation between their expression in EAD treated cells and TNBC patient samples. Thus, the identified hub genes could be explored as therapeutic or diagnostic markers for TNBC. Hence, EAD appears to be a promising therapeutic candidate for TNBC by targeting various hallmarks of cancer, including cell death resistance, uncontrolled proliferation and metastasis. To conclude, the identified pathways and validated targets for EAD will provide a roadmap for further in vivo studies and preclinical/clinical validation required for potential drug development.
Collapse
Affiliation(s)
- Sreerenjini Lakshmi
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695 019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sulochana Priya
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695 019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
7
|
Mandal T, Shukla D, Pattanayak S, Barman R, Ashraf R, Dixit AK, Kumar S, Kumar D, Srivastava AK. Ellagic Acid Induces DNA Damage and Apoptosis in Cancer Stem-like Cells and Overcomes Cisplatin Resistance. ACS OMEGA 2024; 9:48988-49000. [PMID: 39713677 PMCID: PMC11656259 DOI: 10.1021/acsomega.3c08819] [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: 11/06/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 12/24/2024]
Abstract
Cancer stem cells (CSCs) are responsible for chemoresistance and tumor relapse in many solid malignancies, including lung and ovarian cancer. Ellagic acid (EA), a natural polyphenol, exhibits anticancer effects on various human malignancies. However, its impact and mechanism of action on cancer stem-like cells (CSLCs) are only partially understood. In this study, we evaluated the therapeutic potential and underlying molecular mechanism of EA isolated from tropical mango against CSLCs. Herein, we observed that EA treatment reduces the stem-like phenotypes in cancer cells, thereby lowering the cell survival and self-renewal potential of ovarian and lung CSLCs. Additionally, EA treatment limits the populations of lung and ovarian CSLCs characterized by CD133+ and CD44+CD117+, respectively. A mechanistic investigation showed that EA treatment induces ROS generation by altering mitochondrial dynamics, causing changes in the levels of Drp1 and Mfn2, which lead to an increased level of accumulation of DNA damage and eventually trigger apoptosis in CSLCs. Moreover, pretreatment with EA sensitizes CSLCs to cisplatin treatment by enhancing DNA damage accumulation and impairing the DNA repair ability of the CSLCs. Furthermore, EA pretreatment significantly reduces cisplatin-induced mutation frequency and improves drug retention in CSLCs, potentially suppressing the development of acquired drug resistance. Taken together, our results demonstrate an unreported finding that EA inhibits CSLCs by targeting mitochondrial function and triggering apoptosis. Thus, EA can be used either alone or in combination with other chemotherepeutic drugs for the management of cancer.
Collapse
Affiliation(s)
- Tanima Mandal
- Cancer
Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Devendra Shukla
- Cancer
Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Subhamoy Pattanayak
- Organic
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Biology, Kolkata, West Bengal 700032, India
| | - Raju Barman
- Organic
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Biology, Kolkata, West Bengal 700032, India
| | - Rahail Ashraf
- Division
of Biology, Indian Institute of Science
Education & Research Tirupati, Tirupati, Andhra Pradesh 517507, India
| | - Amit Kumar Dixit
- CCRAS-Central
Ayurveda Research Institute, Kolkata, West Bengal 700091, India
| | - Sanjay Kumar
- Division
of Biology, Indian Institute of Science
Education & Research Tirupati, Tirupati, Andhra Pradesh 517507, India
| | - Deepak Kumar
- Organic
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amit Kumar Srivastava
- Cancer
Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
8
|
Suha H, Tasnim SA, Rahman S, Alodhayb A, Albrithen H, Poirier RA, Uddin KM. Evaluating the Anticancer Properties of Novel Piscidinol A Derivatives: Insights from DFT, Molecular Docking, and Molecular Dynamics Studies. ACS OMEGA 2024; 9:49639-49661. [PMID: 39713673 PMCID: PMC11656217 DOI: 10.1021/acsomega.4c07808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024]
Abstract
Cancer is characterized by uncontrolled cell growth and spreading throughout the body. This study employed computational approaches to investigate 18 naturally derived anticancer piscidinol A derivatives (1-18) as potential therapeutics. By examining their interactions with 15 essential target proteins (HIF-1α, RanGAP, FOXM1, PARP2, HER2, ERα, NGF, FAS, GRP78, PRDX2, SCF complex, EGFR, Bcl-xL, ERG, and HSP70) and comparing them with established drugs such as camptothecin, docetaxel, etoposide, irinotecan, paclitaxel, and teniposide, compound 10 emerged as noteworthy. In molecular dynamics simulations, the protein with the strongest binding to the crucial 1A52 protein exceeded druglikeness criteria and displayed extraordinary stability within the enzyme's pocket over varied temperatures (300-320 K). Additionally, density functional theory was used to calculate dipole moments and molecular orbital characteristics, as well as analyze the thermodynamic stability of the putative anticancer derivatives. This finding reveals a well-defined, potentially therapeutic relationship supported by theoretical analysis, which is in good agreement with subsequent assessments of their potential in vitro cytotoxic effects of piscidinol A derivatives (6-18) against various cancer cell lines. Future in vivo and clinical studies are required to validate these findings further. Compound 10 thus emerges as an intriguing contender in the fight against cancer.
Collapse
Affiliation(s)
- Humaera
Noor Suha
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Syed Ahmed Tasnim
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Shofiur Rahman
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah Alodhayb
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Department
of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hamad Albrithen
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Department
of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Raymond A. Poirier
- Department
of Chemistry, Memorial University, St. John’s, Newfoundland
and Labrador A1C 5S7, Canada
| | - Kabir M. Uddin
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| |
Collapse
|
9
|
Ritika, Liao ZY, Chen PY, Rao NV, Mathew J, Sharma R, Grewal AS, Singh G, Mehan S, Liou JP, Pan CH, Nepali K. Rationally designed febuxostat-based hydroxamic acid and its pH-Responsive nanoformulation elicits anti-tumor activity. Eur J Med Chem 2024; 279:116866. [PMID: 39293244 DOI: 10.1016/j.ejmech.2024.116866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/20/2024]
Abstract
Attempts to furnish antitumor structural templates that can prevent the occurrence of drug-induced hyperuricemia spurred us to generate xanthine oxidase inhibitor-based hydroxamic acids and anilides. Specifically, the design strategy involved the insertion of febuxostat (xanthine oxidase inhibitor) as a surface recognition part of the HDAC inhibitor pharmacophore model. Investigation outcomes revealed that hydroxamic acid 4 elicited remarkable antileukemic effects mediated via HDAC isoform inhibition. Delightfully, the adduct retained xanthine oxidase inhibitory activity, though xanthine oxidase inhibition was not the underlying mechanism of its cell growth inhibitory effects. Also, compound 4 demonstrated significant in-vivo anti-hyperuricemic (PO-induced hyperuricemia model) and antitumor activity in an HL-60 xenograft mice model. Compound 4 was conjugated with poly (ethylene glycol) poly(aspartic acid) block copolymer to furnish pH-responsive nanoparticles (NPs) in pursuit of circumventing its cytotoxicity towards the normal cell lines. SEM analysis revealed that NPs had uniform size distributions, while TEM analysis ascertained the spherical shape of NPs, indicating their ability to undergo self-assembly. HDAC inhibitor 4 was liberated from the matrix due to the polymeric nanoformulation's pH-responsiveness, and the NPs demonstrated selective cancer cell targeting ability.
Collapse
Affiliation(s)
- Ritika
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Zi-Yi Liao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan
| | - Pin-Yu Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan
| | - N Vijayakamasewara Rao
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Jacob Mathew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan
| | - Ajmer Singh Grewal
- Department of Pharmaceutical Sciences, Guru Gobind Singh College of Pharmacy, Near Guru Nanak Khalsa College, Yamuna Nagar, 135001, Haryana, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India; Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Chun Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
10
|
Gowda BJ, Ahmed MG, Thakur RRS, Donnelly RF, Vora LK. Microneedles as an Emerging Platform for Transdermal Delivery of Phytochemicals. Mol Pharm 2024; 21:6007-6033. [PMID: 39470172 PMCID: PMC11615954 DOI: 10.1021/acs.molpharmaceut.4c00894] [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: 08/08/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Phytochemicals, which are predominantly found in plants, hold substantial medicinal value. Despite their potential, challenges such as poor oral bioavailability and instability in the gastrointestinal tract have limited their therapeutic use. Traditional intra/transdermal drug delivery systems offer some advantages over oral administration but still suffer from issues such as limited penetration depth, slow drug release rates, and inconsistent drug absorption. In contrast, microneedles (MNs) represent a significant advancement in intra/transdermal drug delivery by providing precise control over phytochemical delivery and enhanced penetration capabilities. By circumventing skin barriers, MNs directly access dermal layers rich in blood vessels and lymphatics, thus facilitating efficient phytochemical delivery. This review extensively discusses the obstacles of traditional oral delivery and the benefits of intra/transdermal delivery routes with a particular focus on the transformative potential of MNs for phytochemical delivery. This review explores the complexities of delivering phytochemicals through intra/transdermal routes, the development and types of MNs as innovative delivery tools, and the optimal design and properties of MNs for effective phytochemical delivery. Additionally, this review examines the versatile applications of MN-mediated phytochemical delivery, including its role in administering phytophotosensitizers for photodynamic therapy, and concludes with insights into relevant patents and future perspectives.
Collapse
Affiliation(s)
- B.H. Jaswanth Gowda
- School
of Pharmacy, Queen’s University Belfast,
Medical Biology Centre, Belfast BT9 7BL, United
Kingdom
- Department
of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department
of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Raghu Raj Singh Thakur
- School
of Pharmacy, Queen’s University Belfast,
Medical Biology Centre, Belfast BT9 7BL, United
Kingdom
| | - Ryan F. Donnelly
- School
of Pharmacy, Queen’s University Belfast,
Medical Biology Centre, Belfast BT9 7BL, United
Kingdom
| | - Lalitkumar K. Vora
- School
of Pharmacy, Queen’s University Belfast,
Medical Biology Centre, Belfast BT9 7BL, United
Kingdom
| |
Collapse
|
11
|
Liu Y, Yu X, Shen H, Hong Y, Hu G, Niu W, Ge J, Xuan J, Qin JJ, Li Q. Mechanisms of traditional Chinese medicine in the treatment and prevention of gastric cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156003. [PMID: 39305742 DOI: 10.1016/j.phymed.2024.156003] [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: 04/03/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Gastric cancer (GC) ranks as the fifth most prevalent malignancy worldwide. Conventional treatments, including radiotherapy and chemotherapy, often induce severe side effects and significant adverse reactions, and they may also result in drug resistance. Consequently, there is a critical need for the development of new therapeutic agents. Traditional Chinese Medicine (TCM) and natural products are being extensively researched due to their low toxicity, multi-targeted approaches, and diverse pathways. Scholars are increasingly focusing on identifying active anticancer components within TCM. PURPOSE This review aims to summarise research conducted over the past 14 years on the treatment of GC using TCM. The focus is on therapeutic targets, mechanisms, and efficacy of Chinese medicine and natural products, including monomer compounds, extracts or analogues, and active ingredients. METHODS Relevant articles on TCM and GC were retrieved from PubMed using appropriate keywords. The collected articles were screened and classified according to the types of TCM, with an emphasis on the molecular mechanisms underlying the treatment of GC. RESULTS The research on TCM indicates that TCM and natural products can effectively inhibit the metastasis, proliferation, and invasion of tumour cells. They can also induce apoptosis, autophagy and improve the chemosensitivity of drug-resistant cells. Additionally, injections derived from Chinese herbal medicine, when used as an adjunct to conventional chemotherapy, can significantly improve the prognosis of GC patients by reducing chemotherapy toxicity. CONCLUSION This review summarises the progress of TCM treatment of GC over the past 14 years, and discusses its therapeutic application of GC, which proves that TCM is a promising treatment strategy for GC in the future.
Collapse
Affiliation(s)
- Yanyang Liu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuefei Yu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China
| | - Huize Shen
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yangjian Hong
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaofeng Hu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenyuan Niu
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiaming Ge
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Xuan
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiang-Jiang Qin
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1, Banshan east road, Gongshu district, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Gibbs RJ, Chambers AC, Hill DJ. The emerging role of Fusobacteria in carcinogenesis. Eur J Clin Invest 2024; 54 Suppl 2:e14353. [PMID: 39674881 DOI: 10.1111/eci.14353] [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: 09/27/2024] [Accepted: 11/04/2024] [Indexed: 12/17/2024]
Abstract
The Fusobacterium genus comprises Gram-negative, obligate anaerobic bacteria that typically reside in the periodontium of the oral cavity, gastrointestinal tract, and female genital tract. The association of Fusobacterial spp. with colorectal tumours is widely accepted, with further evidence that this pathogen may also be implicated in the development of other malignancies. Fusobacterial spp. influence malignant cell behaviours and the tumour microenvironment in various ways, which can be related to the multiple surface adhesins expressed. These adhesins include Fap2 (fibroblast-activated protein 2), CpbF (CEACAM binding protein of Fusobacteria), FadA (Fusobacterium adhesin A) and FomA (Fusobacterial outer membrane protein A). This review outlines the influence of Fusobacteria in promoting cancer initiation and progression, impacts of therapeutic outcomes and discusses potential therapeutic interventions where appropriate.
Collapse
|
13
|
Mgijima T, Sibuyi NRS, Fadaka AO, Meyer S, Madiehe AM, Meyer M, Onani MO. Wound healing effects of biogenic gold nanoparticles synthesized using red wine extracts. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:399-410. [PMID: 39069752 DOI: 10.1080/21691401.2024.2383583] [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: 01/26/2024] [Revised: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024]
Abstract
Gold nanoparticles (AuNPs) were synthesized using three red wine extracts (RW-Es); by varying temperature, pH, concentrations of RW-Es and gold salt. The RW-AuNPs were characterized by UV-vis, transmission electron microscopy (TEM), dynamic light scattering (DLS), and the Fourier Transform Infra-red Spectroscopy (FT-IR). Their stability was evaluated in water, foetal bovine serum (FBS), phosphate-buffered saline (PBS), and Dulbecco's Modified Eagle Medium (DMEM) by UV-Vis. The effect of the RW-Es and RW-AuNPs on KMST-6 cell cell viability was evaluated by MTT assay; and their wound healing effects were monitored by scratch assay. RW-AuNPs synthesis was observed by colour change, and confirmed by UV-Vis spectrum, with an absorption peak around 550 nm. The hydrodynamic sizes of the RW-AuNPs ranged between 10 and 100 nm. Polyphenols, carboxylic acids, and amino acids are some of functional groups in the RW-Es that were involved in the reduction of RW-AuNPs. The RW-AuNPs were stable in test solutions and showed no cytotoxicity to the KMST-6 cells up to 72 h. AuNPs synthesized from Pinotage and Cabernet Sauvignon enhanced proliferation of KMST-6 cells and showed potential as wound healing agents. Further studies are required to investigate the molecular mechanisms involved in the potential wound-healing effect of the RW-AuNPs.
Collapse
Affiliation(s)
- Tswellang Mgijima
- Organometallics and Nanomaterials, Department of Chemical Sciences, University of the Western Cape, Bellville, South Africa
| | - Nicole R S Sibuyi
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre (NIC) Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Bellville, South Africa
- Health Platform, Advanced Materials Division, Mintek, Randburg, South Africa
| | - Adewale O Fadaka
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre (NIC) Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Samantha Meyer
- Phytotherapy Research Group, Department of Biomedical Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Abram M Madiehe
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre (NIC) Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Mervin Meyer
- Department of Science and Innovation (DSI)/Mintek Nanotechnology Innovation Centre (NIC) Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Martin O Onani
- Organometallics and Nanomaterials, Department of Chemical Sciences, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
14
|
Hekmatshoar Y, Karabay AZ, Ozkan T, Koc A, Sunguroglu A. Methylsulfonylmethane induces caspase-dependent apoptosis in acute myeloid leukemia cell lines. Fundam Clin Pharmacol 2024; 38:1094-1102. [PMID: 39114894 DOI: 10.1111/fcp.13030] [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: 03/26/2024] [Revised: 06/20/2024] [Accepted: 07/16/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous ailment in both biological and clinical concepts. Numerous efforts have been devoted to discover natural compounds for combating cancer, which showed great potential in cancer management. Methylsulfonylmethane (MSM), an organosulfur dietary supplement, is utilized for improving various clinical conditions, particularly osteoarthritis. MSM can exert antitumor activity in a wide range of cancers. OBJECTIVES The molecular mechanisms of action underlying antileukemic activity of MSM remain unclear. In this regard, we aimed to investigate the anticancer properties of MSM on human AML cell lines (U937 and HL60) with focus on underlying cell death mechanism. METHODS Anticancer activity of the MSM was examined employing MTT assay, Annexin V-PE/7AAD staining, caspase3/7 activity test, and real-time qPCR. Both cell lines were treated with different concentrations (50-400 mM) of MSM for 24 h. Pretreatment of the cells with a caspase inhibitor (i.e., Z-VAD-fmk) was performed for the assessment of apoptosis induction. RESULTS The results of MTT assay revealed that in both cell lines, the MSM markedly reduced cell viability in comparison to the control cells. Additionally, findings of Annexin V-7AAD staining revealed that MSM induced apoptosis and activated caspase 3/7 in both cell lines markedly. Real-time quantitative PCR results also supported the induction of apoptosis in AML cells. MSM altered the expression levels of various apoptotic genes (BAX, BAD, and BIM). CONCLUSION Overall, our results indicated that MSM could induce apoptosis in AML cell lines in a dose-dependent manner, which therefore could be utilized as an antileukemic agent.
Collapse
Affiliation(s)
- Yalda Hekmatshoar
- Department of Medical Biology, Faculty of Medicine, Altinbas University, Istanbul, Turkey
| | - Arzu Zeynep Karabay
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Tulin Ozkan
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Asli Koc
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Asuman Sunguroglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
15
|
Zhao L, Ma H, Jiang Y, Li Y, Guo N, Chen Y, Jiang X, Zhao Y, Yang J, Liu Y, Wen K, Wang L, Jian L, Fan X. Reserpine, a novel N6-methyladenosine regulator, reverses Lenvatinib resistance in hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156002. [PMID: 39326134 DOI: 10.1016/j.phymed.2024.156002] [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: 03/01/2024] [Revised: 05/23/2024] [Accepted: 06/09/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an aggressive malignancy and a growing global health problem. Reserpine (Res), a plant-derived hypertension drug, has been reported to possess anti-tumor efficacy. However, the role and function of Res in N6-methyladenosine (m6A) regulation and Lenvatinib (Len) resistance in HCC have not been clarified. PURPOSE To verify whether Res can be used as a natural small-molecule regulator of m6A to reverse Len resistance in HCC. METHODS Dot blotting, Western blotting and m6A quantification were used to compare and analyze the differential expression of m6A and its methyltransferase METTL3. Western blotting, Real-Time PCR (RT-PCR), cellular thermal shift assay (CETSA) and molecular docking were used to explore the mechanism of interaction between Res and m6A. The effects of Res on the biological characteristics of Lenvatinib-resistant HCC cells were investigated through CCK-8, clone formation, and Transwell assays. Cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models were used to assess the ability of Res to reverse Len resistance in vivo. MeRIP m6A sequencing, PATHWAY analysis and Western blotting were used to analyze the downstream signaling pathways and genes involved in Res-mediated reversal of Len resistance. RESULTS Len resistance in HCC is related to the increased m6A level and the high expression of METTL3. Res affects the activity of METTL3 protein by binding to it, thereby downregulating the level of m6A. In vitro study showed that Res can sensitize HCC cells to the anti-tumor effects of Len treatment, including blocking proliferation, inhibiting migration, and inducing apoptosis. Len-resistant CDX and PDX models revealed that Res can reverse the resistant phenotype, with the tumor inhibition rates of 77.46 % and 62.1 %, respectively, when combined with Len treatment. Analysis of xenograft tissues showed that the combination of Res and Len down-regulates the m6A level, reduces proliferation biomarkers, and induces apoptosis, which is consistent with the in vitro data. Mechanistically, our preliminary results indicate that Res can up-regulate the SMAD3 level by down-regulating m6A in Len-resistant cells. CONCLUSIONS Reserpine, a small-molecule regulator of m6A, reverses Lenvatinib-resistant phenotypes, including proliferation, migration and anti-apoptosis, in vitro and in vivo by targeting SMAD3 and down-regulating the m6A level in HCC.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Heyao Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Yuhui Jiang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yingying Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ning Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yu Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaowen Jiang
- Department of Analysis and Pharmacology of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yunpeng Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jingjing Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Yifei Liu
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Kaishu Wen
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Lingyan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Xinyu Fan
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
16
|
Balaji D, Balakrishnan R, Srinivasan D, Subbarayan R, Shrestha R, Srivastava N, Chauhan A. The Impact of SARS-CoV-2 on Liver Diseases and Potential Phytochemical Treatments. INFECTIOUS MICROBES AND DISEASES 2024; 6:177-188. [DOI: 10.1097/im9.0000000000000161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has brought about numerous challenges. One of these challenges is the impact of SARS-CoV-2 on the liver. Although this virus primarily affects the lungs, it can induce elevated transaminase levels and the development of scar tissue in the liver, exacerbating preexisting liver conditions. Individuals with preexisting conditions, such as nonalcoholic fatty liver disease, alcohol-induced liver disease and hepatocellular carcinoma, face an increased risk of mortality from COVID-19. However, drugs currently used to treat COVID-19 have undesirable side effects, which make them unsuitable for patients with preexisting liver conditions. In this review, we explore the potential of phytochemicals, such as apigenin, berberine, curcumin, epigallocatechin-3-gallate, quercetin, resveratrol and silymarin, for treatment of the liver conditions, including nonalcoholic fatty liver disease, alcohol-induced liver disease and hepatocellular carcinoma. We also discuss significant associations between phytochemicals and COVID-19 by depicting their molecular interactions. Based on the discussed overlapping functions, it is important to assess the therapeutic efficacy of phytochemicals that possess hepatoprotective properties as potential alternative treatments for COVID-19.
Collapse
Affiliation(s)
- Dhanvee Balaji
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Ranjith Balakrishnan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | | | | | | | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
17
|
Hashemi M, Khoushab S, Aghmiuni MH, Anaraki SN, Alimohammadi M, Taheriazam A, Farahani N, Entezari M. Non-coding RNAs in oral cancer: Emerging biomarkers and therapeutic frontier. Heliyon 2024; 10:e40096. [PMID: 39583806 PMCID: PMC11582460 DOI: 10.1016/j.heliyon.2024.e40096] [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: 06/09/2024] [Revised: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Around the world, oral cancer (OC) is a major public health problem, resulting in a significant number of deaths each year. Early detection and treatment are crucial for improving patient outcomes. Recent progress in DNA sequencing and transcriptome profiling has revealed extensive non-coding RNAs (ncRNAs) transcription, underscoring their regulatory importance. NcRNAs influence genomic transcription and translation and molecular signaling pathways, making them valuable for various clinical applications. Combining spatial transcriptomics (ST) and spatial metabolomics (SM) with single-cell RNA sequencing provides deeper insights into tumor microenvironments, enhancing diagnostic and therapeutic precision for OC. Additionally, the exploration of salivary biomarkers offers a non-invasive diagnostic avenue. This article explores the potential of ncRNAs as diagnostic and therapeutic tools for OC.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Hobabi Aghmiuni
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeid Nemati Anaraki
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Operative, Faculty of Dentistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University,Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
18
|
Bhutta ZA, Choi KC. Phytochemicals as Novel Therapeutics for Triple-Negative Breast Cancer: A Comprehensive Review of Current Knowledge. Phytother Res 2024. [PMID: 39533509 DOI: 10.1002/ptr.8376] [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: 01/14/2024] [Revised: 05/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Triple-negative breast cancer is a characteristic subtype of breast cancer that lacks the estrogen receptor, human epidermal growth factor receptor 2, and progesterone receptor. Because of its highly diverse subtypes, increased metastasis capability, and poor prognosis, the risk of mortality for people with triple-negative breast cancers is high as compared with other cancers. Chemotherapy is currently playing a major role in treating triple-negative breast cancer patients; however, poor prognosis due to drug resistance is causing serious concern. Recent studies on several phytochemicals derived from various plants being used in Traditional Chinese Medicine, Traditional Korean Medicine, Ayurveda (Traditional Indian Medicine), and so on, have demonstrated to be a promising agent as a viable therapy against triple-negative breast cancer. Phytochemicals categorized as alkaloids, polyphenols, terpenoids, phytosterols, and organosulfur compounds have been demonstrated to reduce cancer cell proliferation and metastasis by activating various molecular pathways, thereby reducing the spread of triple-negative breast cancer. This review analyzes the molecular mechanisms by which various phytochemicals fight triple-negative breast cancer and offers a perspective on the difficulties and potential prospects for treating triple-negative breast cancer with various phytochemicals.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
19
|
Chauhan A, Salwa, Shedgaonkar GG, Kumar L, Karmakar A, Khajuria S, Raghavendra AP, Verma R. Antioxidant and anticancer activities of hesperetin and its novel formulations in KB cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03581-y. [PMID: 39531045 DOI: 10.1007/s00210-024-03581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
This study aimed to formulate the hesperetin nanostructured lipid carriers (NLCs) containing oro-mucosal gel for its activity assessment on the KB cell line. NLCs were prepared with glyceryl monostearate, oleic acid, and lecithin using a modified constant-temperature emulsification technique. The particle size analysis, in vitro drug release studies, etc., of prepared NLCs were evaluated. The formulated gels were analyzed with respect to spreadability, extrudability, swelling index, texture analysis, etc. The particle size, polydispersity index, zeta potential, and drug entrapment of nanocarriers were recorded to be 221.733 ± 61.536 nm, 0.381 ± 0.091, - 51.433 ± 4.143 mV, and 89.29%, respectively. The optimized NLCs in 24 h released 87.14 ± 6.62% of the drug. The round shape of NLCs was noticed with scanning electron microscopy. The pH, spreadability, extrudability, swelling index, content uniformity, and drug release studies of hesperetin NLCs-containing gel (HNG) were found to be 6.81 ± 0.04, 2.49 ± 0.04 cm.mg/s, 539.04 ± 32.88 g/cm2, 4.27 ± 0.47, 107.98 ± 1.93%, and 90.17 ± 6.67% (in 48 h), respectively. The developed formulations showed promising in vitro anticancer and antioxidant activities. HNP results authorize that the formulation may be beneficial for the treatment of oral cancer.
Collapse
Affiliation(s)
- Arunima Chauhan
- Faculty of Dentistry, Melaka-Manipal Medical College, Manipal Academy of Higher Education, Manipal, India
- Faculty of Dentistry, Manipal University College Malaysia, Jalan Batu Hampar, Bukit Baru, Melaka, Malaysia
| | - Salwa
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Gayatri Gopal Shedgaonkar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India.
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur 844 102, Vaishali, Bihar, India.
| | - Arka Karmakar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur 844 102, Vaishali, Bihar, India
| | - Salil Khajuria
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur 844 102, Vaishali, Bihar, India
| | | | - Ruchi Verma
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
20
|
Chan SW, Mahmoud VL, Wang X, Teoh ML, Loh KM, Ng CH, Wong WF, Looi CY. Chemical profiling and cytotoxicity screening of agarwood essential oil (Aquilaria sinensis) in brine shrimp nauplii and cancer cell lines. PLoS One 2024; 19:e0310770. [PMID: 39509364 PMCID: PMC11542896 DOI: 10.1371/journal.pone.0310770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/30/2024] [Indexed: 11/15/2024] Open
Abstract
Agarwood essential oil (AEO) has gained attention from healthcare industries due to its numerous pharmacological properties. However, a comprehensive understanding of the chemical composition and its cytotoxic property is lacking. The objective of this study was to investigate the chemical profile as well as the cytotoxic concentration range of AEO derived from Aquilaria sinensis agarwood. Gas chromatography-mass spectrometry (GC-MS) was employed to identify the AEO components. Results showed that sesquiterpenes and sesquiterpenoids constitute 95.85% of the AEO. Among the major compounds identified are allo-aromadendrene (13.04%), dihydro-eudesmol (8.81%), α-eudesmol (8.48%), bulnesol (7.63%), τ-cadinol (4.95%), dehydrofukinone (3.83%), valerenol (3.54%), cis-nerolidol (2.75%), agarospirol (2.72%), dehydrojinkoh-eremol (2.53%), selina-3,11-dien-9-al (2.36%), guaiol (2.12%) and caryophyllene oxide (2.0%). The presence of volatile quality marker compounds such as 10-epi-ϒ-eudesmol, aromadendrane, β-agarofuran, α-agarofuran, γ-eudesmol, agarospirol and guaiol, with no contaminants detected, indicates that the extracted AEO is of high purity. Interestingly, the AEO displayed moderate to high toxicity in brine shrimp lethality test (BLST). All studied tumor cell lines (MDA-MB-231, HepG2, B16F10) exhibited varying degrees of sensitivity to AEO, which resulted in time and dose-dependent reduction of cell proliferation. Moreover, flow cytometry analysis revealed that AEO could induce apoptosis in treated HepG2 cells. Our findings showed that AEO contains bioactive components that may be exploited in future studies for the development of anti-cancer therapeutics.
Collapse
Affiliation(s)
- Sook Wah Chan
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
- Food Security & Nutrition Impact Lab, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Valizadeh Lakeh Mahmoud
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Xin Wang
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Ming-Li Teoh
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
- Clean Technology Impact Lab, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Kar Min Loh
- Faculty of Medicine, Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chun Howe Ng
- Faculty of Medicine, Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Won Fen Wong
- Faculty of Medicine, Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
- Digital Health and Medical Advancements Impact Lab, Taylor’s University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
21
|
Song L, Qiu Q, Ju F, Zheng C. Mechanisms of doxorubicin-induced cardiac inflammation and fibrosis; therapeutic targets and approaches. Arch Biochem Biophys 2024; 761:110140. [PMID: 39243924 DOI: 10.1016/j.abb.2024.110140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Doxorubicin plays a pivotal role in the treatment of various malignancies. Despite its efficacy, the cardiotoxicity associated with doxorubicin limits its clinical utility. The cardiotoxic nature of doxorubicin is attributed to several mechanisms, including its interference with mitochondrial function, the generation of reactive oxygen species (ROS), and the subsequent damage to cardiomyocyte DNA, proteins, and lipids. Furthermore, doxorubicin disrupts the homeostasis of cardiac-specific transcription factors and signaling pathways, exacerbating cardiac dysfunction. Oxidative stress, cell death, and other severe changes, such as mitochondrial dysfunction, activation of pro-oxidant enzymes, the renin-angiotensin system (RAS), endoplasmic reticulum (ER) stress, and infiltration of immune cells in the heart after treatment with doxorubicin, may cause inflammatory and fibrotic responses. Fibrosis and inflammation can lead to a range of disorders in the heart, resulting in potential cardiac dysfunction and disease. Various adjuvants have shown potential in preclinical studies to mitigate these challenges associated with cardiac inflammation and fibrosis. Antioxidants, plant-based products, specific inhibitors, and cardioprotective drugs may be recommended to alleviate cardiotoxicity. This review explores the complex mechanisms of doxorubicin-induced heart inflammation and fibrosis, identifies possible cellular and molecular targets, and investigates potential substances that could help reduce these harmful effects.
Collapse
Affiliation(s)
- Linghua Song
- Department of Pharmacy, Yantai Mountain Hospital, Yantai City, Shandong Province, 264001, China
| | - Qingzhuo Qiu
- Medical Imaging Department of Qingdao Women and Children's Hospital, 266000, China
| | - Fei Ju
- Department of Critical Care, Medicine East Hospital of Qingdao Municipal Hospital, 266000, China
| | - Chunyan Zheng
- Cadre Health Office of Zibo Central Hospital in Shandong Province, 255000, China.
| |
Collapse
|
22
|
Czerwińska K, Radziejewska I. Rosmarinic Acid: A Potential Therapeutic Agent in Gastrointestinal Cancer Management-A Review. Int J Mol Sci 2024; 25:11704. [PMID: 39519255 PMCID: PMC11546295 DOI: 10.3390/ijms252111704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Gastrointestinal cancers are still the leading cause of death worldwide. This is related, among other things, to the non-specific symptoms, especially in the initial stages, and also to the limited possibilities for treatment. Therefore, research is still being conducted to improve the detection of this type of cancer and increase the effectiveness of therapy. The potential application of natural compounds in cancer management deserves special attention. In the group of such products, there are polyphenolic compounds that reveal, e.g., anti-oxidative, anti-carcinogenic, anti-inflammatory, anti-diabetic, and neuroprotective properties. One of these polyphenols is rosmarinic acid, commonly found in plants such as the Boraginaceae and Nepetoideae subfamilies of the Lamiaceae (mint) family. A number of studies have considered the positive effects of rosmarinic acid in the treatment of many cancers, including gastrointestinal ones such as oral, stomach, pancreas, colon, and liver cancers. The main aim of this paper was to summarize the mechanisms of action of rosmarinic acid in gastrointestinal cancers.
Collapse
Affiliation(s)
| | - Iwona Radziejewska
- Department of Medical Chemistry, Medical University of Białystok, ul. Mickiewicza 2a, 15-222 Białystok, Poland;
| |
Collapse
|
23
|
He L, Su Z, Wang S. The anti-obesity effects of polyphenols: a comprehensive review of molecular mechanisms and signal pathways in regulating adipocytes. Front Nutr 2024; 11:1393575. [PMID: 39539361 PMCID: PMC11557335 DOI: 10.3389/fnut.2024.1393575] [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] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Excess weight gain is a growing concern worldwide, fueled by increased consumption of calorie-dense foods and more sedentary lifestyles. Obesity in China is also becoming increasingly problematic, developing into a major public health concern. Obesity not only increases the risk of associated disease but also imposes a burden on health care systems, and it is thus imperative that an effective intervention approach be identified. Recent studies have demonstrated that the polyphenol-rich Mediterranean diet has considerable potential in this regard. Polyphenols can inhibit the production of adipocytes and reduce adverse reactions, such as inflammation, insulin resistance, and gut microflora imbalance. In this review, we examine four polyphenols (curcumin, ellagic acid, ferulic acid, and quercetin) in terms of their potential as interventions targeting obesity. The mechanisms that help promote adipocyte browning, increase thermogenic factors, increase thermogenesis, and regulate adipocyte differentiation are summarized, and key signaling pathways, including PPARγ, C/EBP-, and others, are reviewed.
Collapse
Affiliation(s)
- Lan He
- Department of Cardiology, The First People’s Hospital of Wenling, Taizhou University Affiliated Wenling Hospital, Zhejiang, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Zhan Su
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Shuangshuang Wang
- Department of Cardiology, The First People’s Hospital of Wenling, Taizhou University Affiliated Wenling Hospital, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
24
|
Chatterjee A, Khan R, Mukherjee T, Sahoo PP, Tiwari LN, Singh BN, Kumari R, Kumari A, Rai A, Ray S. Harnessing bacterial metabolites for enhanced cancer chemotherapy: unveiling unique therapeutic potentials. Arch Microbiol 2024; 206:449. [PMID: 39472338 DOI: 10.1007/s00203-024-04179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
Cancer poses a serious threat to health globally, with millions diagnosed every year. According to Global Cancer Statistics 2024, about 20 million new cases were reported in 2022, and 9.7 million people worldwide died of this condition. Advanced therapies include combination of one or more treatment procedures, depending on the type, stage, and particular genetic constitution of the cancer, which may include surgery, radiotherapy, chemotherapy, immunotherapy, hormone therapy, targeted therapy, and stem cell transplant. Also, awareness about lifestyle changes, preventive measures and screening at early stages has reduced the incidence of the disease; still, there is a major failure in controlling the incidence of cancer because of its complex and multifaceted nature. With increasing interest in bacterial metabolites as possible novel and effective treatment options in cancer therapy, their main benefits include not only direct anticancer effects but also the modulation of the immune system and potential for targeted and combination therapies. They can therefore be used in combination with chemotherapy, radiotherapy, or immunotherapy to improve outcomes or reduce side effects. Furthermore, nanoparticle-based delivery systems have the potential to enhance the potency and safety of anticancer drugs by providing improved stability, targeted release, and controlled delivery.
Collapse
Affiliation(s)
- Aroni Chatterjee
- Department of Biotechnology, School of Biotechnology and Biosciences, Brainware University, Barasat, Kolkata, 700125, West Bengal, India
| | - Rajni Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, 844102, Bihar, India
| | - Triparna Mukherjee
- Department of Biotechnology, School of Biotechnology and Biosciences, Brainware University, Barasat, Kolkata, 700125, West Bengal, India
| | - Preity Pragnya Sahoo
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Laxmi Narayan Tiwari
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Basant Narain Singh
- Department of Botany, Pandit Deendayal Upadhyaya Shekhawati University, Sikar, Nawalgarh Road, Katrathal, Rajasthan, 332024, India
| | - Rashmi Kumari
- Department of Zoology, ZA Islamia College Siwan, Affiliated Unit of Jai Prakash University, Chapra, Bihar, 841226, India
| | - Anisha Kumari
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India
| | - Ankit Rai
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India.
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India.
| |
Collapse
|
25
|
Statilko O, Tsiaka T, Sinanoglou VJ, Strati IF. Overview of Phytochemical Composition of Brassica oleraceae var. capitata Cultivars. Foods 2024; 13:3395. [PMID: 39517179 PMCID: PMC11544802 DOI: 10.3390/foods13213395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
The Brassicaceae family includes a wide range of horticultural crops of economic and traditional importance, consumed either fresh, cooked, or fermented. Cabbage (Brassica oleraceae var. capitata) is one of the most important crops of the family. The present review analyzes the most important phytochemicals present in cabbage, focusing on variation of phytochemical composition between cultivars of B. oleraceae var. capitata f. alba, B. oleraceae var. capitata f. rubra, B. oleraceae var. capitata f. acuta, and B. oleraceae var. capitata f. sabauda. Cabbage form and cultivars significantly affect phytochemical compositions. B. oleraceae var. capitata f. rubra cultivars are generally great sources of phenolic compounds, especially anthocyanins, whereas B. oleraceae var. capitata f. alba cultivars display the highest concentration of glucosinolates; nevertheless, their levels are also dependent on the specific cultivar. B. oleraceae var. capitata f. acuta cultivars may be considered advantageous due to their high glucosinolate content and consistent phytochemical composition. Recognizing the benefits of specific cultivars can be valuable for consumers seeking a healthier lifestyle, as well as for scientists aiming to enhance cultivars through breeding programs or use plants' extracts to produce high quality pigments and dietary supplements.
Collapse
Affiliation(s)
| | | | | | - Irini F. Strati
- Laboratory of Chemistry, Analysis & Design of Food Processes, Department of Food Science and Technology, University of West Attica, Agiou Spyridonos, 12243 Egaleo, Greece; (O.S.); (T.T.); (V.J.S.)
| |
Collapse
|
26
|
Orozco-Barocio A, Sánchez-Sánchez MA, Rojas-Mayorquín AE, Godínez-Rubí M, Reyes-Mata MP, Ortuño-Sahagún D. Phytochemicals from Cactaceae family for cancer prevention and therapy. Front Pharmacol 2024; 15:1421136. [PMID: 39512831 PMCID: PMC11540781 DOI: 10.3389/fphar.2024.1421136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024] Open
Abstract
Cancer is a global health issue, increasingly prevalent and a leading cause of mortality. Despite extensive research, conventional treatments remain aggressive, often damaging healthy cells, and exhibit limited efficacy. Addressing drug resistance and enhancing treatment effectiveness are critical challenges in advancing cancer therapy. This review examines the potential of natural plant compounds, particularly phytochemicals and their derivatives, in developing novel anticancer agents. These metabolites have a long history in traditional medicine, with 42% of molecules approved for cancer treatment between 1981 and 2019 being either natural products or derivatives. The Cactaceae family, which comprises more than 1,500 species, represents a largely untapped source of potentially useful chemopreventive and anticancer agents. Although more than 3,000 plants and their derivatives have contributed to chemotherapeutic development, cactus species have received limited attention until recently. Emerging evidence highlights the anticancer potential of fruits, stems, and cladodes from various cactus species. This review provides a comprehensive and current overview of experimental studies on Cactaceae in cancer research, aiming to pave the way for the development of innovative, natural cancer therapeutics and contribute to the ongoing battle against this formidable disease.
Collapse
Affiliation(s)
- Arturo Orozco-Barocio
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Mexico
| | - Marina A. Sánchez-Sánchez
- Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Argelia E. Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Mexico
| | - Marisol Godínez-Rubí
- Laboratorio de Patología Diagnóstica e Inmunohistoquímica, Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - María Paulina Reyes-Mata
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, GuadalajaraMexico
| |
Collapse
|
27
|
Dey S, Dinakar YH, R S, Jain V, Jain R. Navigating the therapeutic landscape for breast cancer: targeting breast cancer stem cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03542-5. [PMID: 39441235 DOI: 10.1007/s00210-024-03542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Breast cancer is a common and deadly malignancy that affects women globally, and breast cancer stem cells (BCSCs) play an important role in tumorigenesis, development, metastasis, and recurrence. Traditional therapies often fail to eliminate BCSCs, leading to treatment resistance and relapse. This review explores the therapeutic strategies which are designed to target BCSCs, including inhibition of key signaling pathway and targeting receptor. This paper also explores the approaches to targeting BCSCs including chemotherapy, phytomedicines, and nanotechnology. Nanotechnology has gained a lot of importance in cancer therapy because of its ability to deliver therapeutic agents with more precision and minimal side effects. Various chemotherapeutic drugs, siRNAs, or gene editing tools are delivered efficiently with the use of nanocarriers which target pathways, receptors, and proteins associated with BCSCs. Over the past few years, stimuli-responsive and receptor-targeted nanocarriers have been explored for better therapeutic effects. In recent times, strategies such as chimeric antigen receptor (CAR) T-cell therapy, ablation therapy, and cell-free therapies are explored for targeting these stem cells. This review provides a recent developmental overview of strategies to attack BCSCs from conventional chemotherapeutic agents to nanotechnological platforms such as polymeric, lipidic, and metal-based nanoparticles and advanced technologies like CAR T cell therapies.
Collapse
Affiliation(s)
- Soudeep Dey
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Yirivinti Hayagreeva Dinakar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Soundarya R
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| |
Collapse
|
28
|
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
|
29
|
Adetunji TL, Olisah C, Acho MA, Oyetunde-Joshua F, Amoo SO. Global Research Trends and Recent Advances in Medicinal Plant-Synthesized Nanoparticles for Cancer Treatment. PLANTS (BASEL, SWITZERLAND) 2024; 13:2836. [PMID: 39458783 PMCID: PMC11511196 DOI: 10.3390/plants13202836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
Worldwide, cancer ranks among the foremost contributors to mortality despite recent medical progress. Alternative approaches in controlling various forms of cancer are being highly explored by researchers. This study provides the global research trends in the utilization of medicinal plant-synthesized nanoparticles for cancer treatment over the span of 18 years using scientometric analysis. Recent research advances on medicinal plant-derived nanoparticles for cancer treatment and their possible mechanisms of action were described. Relevant articles published between 2005 and 2023 were retrieved from Scopus and Web of Science and analyzed using RStudio and VOSViewer. Scientometric indicators were employed to analyze the results. The initial search returned 5695 articles, with a publication growth rate of 3.71% annually. Countries from Asia contributed the most (61.37%) to the total number of publications. The therapeutic effects of nanoparticles derived from medicinal plants can be attributed to various mechanistic pathways, including induced apoptosis from reactive oxygen species generation, as well as mitochondrial and cell membrane disruption, amongst others. Although some reported studies demonstrated promising safety and efficacy against certain cancer cells in vivo and in vitro, the little to no clinical data on medicinal plant-synthesized nanoparticles hinder the ability to make informed decisions about their clinical potential in cancer treatment.
Collapse
Affiliation(s)
- Tomi Lois Adetunji
- Agricultural Research Council—Vegetables, Industrial and Medicinal Plants, Private Bag X293, Pretoria 0001, South Africa;
- Unit for Environmental Sciences and Management (UESM), Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Chijioke Olisah
- Institute for Coastal and Marine Research (CMR), Nelson Mandela University, P.O. Box 77000, Gqeberha 6031, South Africa;
| | | | - Funsho Oyetunde-Joshua
- Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa;
| | - Stephen O. Amoo
- Agricultural Research Council—Vegetables, Industrial and Medicinal Plants, Private Bag X293, Pretoria 0001, South Africa;
- Unit for Environmental Sciences and Management (UESM), Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom 2520, South Africa
| |
Collapse
|
30
|
Thozhukkad Moosaripparambil S, Vadakkadath Meethal K. Unveiling the anticancer potential of Anamirta cocculus (L.) Wight& Arn.: Evidences from cytotoxicity studies, apoptosis analysis, and molecular docking. 3 Biotech 2024; 14:245. [PMID: 39345961 PMCID: PMC11424601 DOI: 10.1007/s13205-024-04096-2] [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: 05/25/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Anamirta cocculus, a woody climber, is extensively utilised in traditional Asian medicine. This study investigates the cytotoxic effects of A. cocculus leaf extracts on various cancer cell lines as well as on a normal cell line. The ethyl acetate extract exhibited potent anticancer activity, with the highest cytotoxicity observed against ovarian cancer cell line (PA1) (IC50 = 8.30 ± 0.38 µg/mL) and colorectal adenocarcinoma cell line (HT29) (IC50 = 17.97 ± 0.63 µg/mL). Notably, the extract displayed low toxicity (18.72 ± 0.73%) on the normal human keratinocyte cell line (HaCaT) at a concentration of 100 µg/mL, indicating selective cytotoxicity towards cancer cells. The acetone extract also demonstrated significant cytotoxicity against various cancer cell lines, including A498, MG63, PA1, and UM-SCC-83B. The ethyl acetate extract of A. cocculus demonstrated potent inhibition of colony formation in HT29 and PA1 cancer cell lines while inducing apoptosis, as evidenced by membrane blebbing, chromatin condensation, and DNA fragmentation. The number of late apoptotic cells increased with an increase in concentrations of ACLE. Molecular docking studies of compounds identified through GC-MS analysis revealed strong interactions with key apoptotic proteins, including caspase-8, p53, caspase-3, and caspase-9. Compounds such as vitamin E, epoxylathyrol, squalene, and phytol showed high binding affinity to these proteins, suggesting their role in apoptosis induction. The possibility of induction of apoptotic proteins through indirect interaction by binding to other proteins or receptors cannot be ruled out. The cytotoxic effects may result from individual, combined, or synergistic actions of these compounds. Among these, epoxylathyrol emerged as a particularly promising anticancer drug candidate based on ADME analysis and binding affinity assessments, warranting further investigation. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04096-2.
Collapse
Affiliation(s)
| | - Kannan Vadakkadath Meethal
- Department of Zoology, University of Calicut, Calicut University PO, Malappuram District, Malappuram, Kerala 673635 India
- Centre for Advances in Molecular Biology, University of Calicut, Calicut University PO, Malappuram District, Malappuram, Kerala 673635 India
| |
Collapse
|
31
|
Das T, Mondal S, Das S, Das S, Das Saha K. Enhanced anticancer activity of (-)-epigallocatechin-3-gallate (EGCG) encapsulated NPs toward colon cancer cell lines. Free Radic Res 2024; 58:565-582. [PMID: 38810269 DOI: 10.1080/10715762.2024.2360013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/08/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
(-)-Epigallocatechin-3-gallate (EGCG), a bioactive polyphenol of green tea, has chemo-preventive effects against various cancer cells. Nanoparticles (NPs) carrying different ligands are able to specifically interact with their receptors on different cancer cells that can provide effective release of cytotoxic drugs. In the present study, we have prepared EGCG entrapped NPs using PLGA (poly(d,l-lactide-co-glycolide)). Polyethylene glycol (PEG) and folic acid (FA) via double emulsion solvent evaporation (DESE) method obtained PLGA-EGCG (P-E), PLGA-PEG-EGCG (PP-E), and PLGA-PEG-FA-EGCG (PPF-E). Nanoformulations had been characterized with 1H NMR and FT-IR techniques, AFM, and DLS. PPF-E NPs showed an average size of 220 nm. Analysis of zeta potential confirmed the stability of NPs. HCT-116, HT-29, HCT-15, and HEK 293 cells were treated with both the prepared NPs and free EGCG (0-140 μM). Result showed PPF-E NPs had improved delivery, uptake and cell cytotoxicity toward human folic acid receptor-positive (FR+) colorectal cancer (CRC) cells as mainly on HCT-116 compared to HT-29, but not on the folic acid-negative cells (FR-) as HCT-15. PPF-E NPs enhanced intracellular reactive oxygen species (ROS) level in absence of N-acetyl-l-cysteine (NAC), elevated DNA fragmentation level, and increased apoptotic cell death at higher doses compared to other two NPs and free EGCG. In conclusion, PPF-E NPs exerted greater efficacy than PP-E, P-E, and free EGCG in HCT-116 cells.
Collapse
Affiliation(s)
- Tanushree Das
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sanchaita Mondal
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sujata Das
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sanjib Das
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Krishna Das Saha
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
32
|
Hasan‐Abad A, Atapour A, Sobhani‐Nasab A, Motedayyen H, ArefNezhad R. Plant-Based Anticancer Compounds With a Focus on Breast Cancer. Cancer Rep (Hoboken) 2024; 7:e70012. [PMID: 39453820 PMCID: PMC11506041 DOI: 10.1002/cnr2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 07/11/2024] [Accepted: 08/11/2024] [Indexed: 10/27/2024] Open
Abstract
Breast cancer is a common form of cancer among women characterized by the growth of malignant cells in the breast tissue. The most common treatments for this condition include chemotherapy, surgical intervention, radiation therapy, hormone therapy, and biological therapy. The primary issues associated with chemotherapy and radiation therapy are their adverse events and significant financial burden among patients in underdeveloped countries. This highlights the need to explore and develop superior therapeutic options that are less detrimental and more economically efficient. Plants provide an abundant supply of innovative compounds and present a promising new avenue for investigating cancer. Plants and their derivations are undergoing a revolution due to their reduced toxicity, expediency, cost-effectiveness, safety, and simplicity in comparison to conventional treatment methods. Natural products are considered promising candidates for the development of anticancer drugs, due perhaps to the diverse pleiotropic effects on target events. The effects of plant-derived products are limited to cancer cells while leaving healthy cells unaffected. Identification of compounds with strong anticancer properties and development of plant-based medications for cancer treatment might be crucial steps in breast cancer therapy. Although bioactive compounds have potent anticancer properties, they also have drawbacks that need to be resolved before their application in clinical trials and improved for the approved drugs. This study aims to give comprehensive information on known anticancer compounds, including their sources and molecular mechanisms of actions, along with opportunities and challenges in plant-based anticancer therapies.
Collapse
Affiliation(s)
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Ali Sobhani‐Nasab
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
| | - Hossein Motedayyen
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
| | - Reza ArefNezhad
- Department of Anatomy, School of MedicineShiraz University of Medical SciencesShirazIran
| |
Collapse
|
33
|
Doan NQH, Tran HN, Nguyen NTM, Pham TM, Nguyen QDK, Vu TT. Synthesis, Antimicrobial - Cytotoxic Evaluation, and Molecular Docking Studies of Quinolin-2-one Hydrazones Containing Nitrophenyl or Isonicotinoyl/Nicotinoyl Moiety. Chem Biodivers 2024; 21:e202401142. [PMID: 39032128 DOI: 10.1002/cbdv.202401142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 07/22/2024]
Abstract
By applying the hybrid molecular strategy, in this study, we reported the synthesis of fifteen quinolin-2-one hydrazones containing nitrophenyl or nicotinonyl/isonicotinoyl moiety, followed by in vitro and in silico evaluations of their potential antimicrobial and anticancer activities. In vitro antimicrobial evaluation of the target compounds on seven pathogenic strains, applying the broth microdilution method, revealed that compound 4a demonstrated the most potential antifungal activity against C. albicans (MIC 512 μg mL-1) and C. krusei (MIC 128 μg mL-1). In vitro cytotoxic evaluation of the target compounds on three human cancer cell lines, employing the MTT method, suggested that compound 5c exhibited the most potential cytotoxicities against HepG2 (IC50 10.19 μM), A549 (IC50 20.43 μM), and MDA-MB-231 (IC50 16.82 μM) cells. Additionally, molecular docking studies were performed to investigate the binding characteristics of compounds 4a and 5c with fungal lanosterol 14α-demethylase and human topoisomerase I-II, respectively, thereby contributing to the elucidation of their in vitro antifungal and cytotoxic properties. Furthermore, compounds 4a and 5c, via SwissADME prediction, could exhibit favorable physicochemical and pharmacokinetic properties. In conclusion, this study provides valuable insights into the potential of quinolin-2-one hydrazones as promising candidates for the development of novel antimicrobial and anticancer agents in the future.
Collapse
Affiliation(s)
- Nam Q H Doan
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Hoan N Tran
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Nhu T M Nguyen
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Thu M Pham
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Quyen D K Nguyen
- Faculty of Pharmacy, Van Lang University, 69/68 Dang Thuy Tram Street, Ward 13, Binh Thanh District, Ho Chi Minh City, 70000, Vietnam
| | - Thanh-Thao Vu
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 41-43 Dinh Tien Hoang Street, Ben Nghe Ward, District 1, Ho Chi Minh City, 70000, Vietnam
| |
Collapse
|
34
|
Alqarni A, Hosmani J, Alassiri S, Alqahtani AMA, Assiri HA. A Network Pharmacology Identified Metastasis Target for Oral Squamous Cell Carcinoma Originating from Breast Cancer with a Potential Inhibitor from F. sargassaceae. Pharmaceuticals (Basel) 2024; 17:1309. [PMID: 39458948 PMCID: PMC11510435 DOI: 10.3390/ph17101309] [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: 08/13/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
This study aimed to identify specific therapeutic targets for oral squamous cell carcinoma (OSCC) that metastasize from breast cancer (BC) by using network pharmacology. The Gene Expression Omnibus for OSCC and BC served as the source of gene expression datasets and their analysis. Upregulated genes and the common intersecting genes of these cancers were determined along with that of the phytochemicals of F. sargassum to predict the pharmacological targets. Further, gene enrichment analysis revealed that their metastasis signature and metastasis targets were determined via a protein interaction network. Molecular docking and pharmacokinetic screening determined the potential therapeutic phytochemicals against the targets. The interaction network of 39 genes thus identified encoding proteins revealed HIF1A as a prominent metastasis target due to its high degree of connectivity and its involvement in cancer-related pathways. Molecular docking showed a strong binding affinity of isonahocol D2, a sargassum-derived compound with HIF1A, presenting a binding energy of -7.1 kcal/mol. Further, pharmacokinetic screening showed favorable ADME properties and molecular dynamics simulations showed stable interactions between isonahocol D2 and HIF1A, with significant stability over 100 ns. This study's results emphasized that isonahocol D2 is a promising therapeutic candidate against HIF1A in OSCC metastasized from breast cancer in translational medicine.
Collapse
Affiliation(s)
| | - Jagadish Hosmani
- Department of Diagnostic Dental Sciences & Oral Biology, College of Dentistry, King Khalid University, Abha 61421, Saudi Arabia; (A.A.); (S.A.); (A.M.A.A.); (H.A.A.)
| | | | | | | |
Collapse
|
35
|
Chakraborty A, Midde A, Chakraborty P, Adhikary S, Kumar S, Arri N, Chandra Das N, Sen Gupta PS, Banerjee A, Mukherjee S. Revisiting Luteolin Against the Mediators of Human Metastatic Colorectal Carcinoma: A Biomolecular Approach. J Cell Biochem 2024:e30654. [PMID: 39300917 DOI: 10.1002/jcb.30654] [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: 05/16/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
Metastatic colorectal carcinoma (mCRC) is one of the prevalent subtypes of human cancers and is caused by the alterations of various lifestyle and diet-associated factors. β-catenin, GSK-3β, PI3K-α, AKT1, and NF-κB p50 are known to be the critical regulators of tumorigenesis and immunopathogenesis of mCRC. Unfortunately, current drugs have limited efficacy, side effects and can lead to chemoresistance. Therefore, searching for a nontoxic, efficacious anti-mCRC agent is crucial and of utmost interest. The present study demonstrates the identification of a productive and nontoxic anti-mCRC agent through a five-targets (β-catenin, GSK-3β, PI3K-α, AKT1, and p50)-based and three-tier (binding affinity, pharmacokinetics, and pharmacophore) screening strategy involving a series of 30 phytocompounds having a background of anti-inflammatory/anti-mCRC efficacy alongside 5-fluorouracil (FU), a reference drug. Luteolin (a phyto-flavonoid) was eventually rendered as the most potent and safe phytocompound. This inference was verified through three rounds of validation. Firstly, luteolin was found to be effective against the different mCRC cell lines (HCT-15, HCT-116, DLD-1, and HT-29) without hampering the viability of non-tumorigenic ones (RWPE-1). Secondly, luteolin was found to curtail the clonogenicity of CRC cells, and finally, it also disrupted the formation of colospheroids, a characteristic of metastasis. While studying the mechanistic insights, luteolin was found to inhibit β-catenin activity (a key regulator of mCRC) through direct physical interactions, promoting its degradation by activating GSK3-β and ceasing its activation by inactivating AKT1 and PI3K-α. Luteolin also inhibited p50 activity, which could be useful in mitigating mCRC-associated proinflammatory milieu. In conclusion, our study provides evidence on the efficacy of luteolin against the critical key regulators of immunopathogenesis of mCRC and recommends further studies in animal models to determine the effectiveness efficacy of this natural compound for treating mCRC in the future.
Collapse
Affiliation(s)
- Ankita Chakraborty
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Advaitha Midde
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pritha Chakraborty
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Sourin Adhikary
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
- Food Toxicology Laboratory, Food, Drug, and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
| | - Simran Kumar
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Navpreet Arri
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nabarun Chandra Das
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Parth Sarthi Sen Gupta
- School of Biosciences and Bioengineering, D. Y. Patil International University, Pune, Maharashtra, India
| | - Aditi Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Suprabhat Mukherjee
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
36
|
Li Z, Yao L, Saravanakumar K, Thuy NTT, Kim Y, Xue C, Zheng X, Cho N. Lespedeza bicolor root extract exerts anti-TNBC potential by regulating FAK-related signalling pathways. Am J Cancer Res 2024; 14:4265-4285. [PMID: 39417178 PMCID: PMC11477838 DOI: 10.62347/mypg4066] [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: 07/14/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lespedeza bicolor is a shrub plant that has been widely distributed in East Asia. The methanol extract from its LBR has been shown to exhibit anticancer and anti-bacterial effects. However, its anticancer efficacy in TNBC remains uncertain. This work aimed to study the anti-TNBC effect of LBR ethanol extract and its underlying mechanism. LBR triggered the cell death in TNBC through inhibiting cell proliferation, S-phase cell arrest, and induction of apoptosis. RNA-seq analysis revealed that the genes altered by LBR treatment were predominantly enriched in the cell adhesion. Notably, LBR inhibited phosphorylation and distribution of FAK. Furthermore, LBR demonstrated significant anticancer activity in xenograft tumors in mice through inhibiting cancer cell growth and inducing apoptosis. This work demonstrated the anticancer efficiency of LBR in TNBC without causing significant adverse effect, which providing a foundation for developing LBR based chemotherapeutic agents for breast cancer therapy.
Collapse
Affiliation(s)
- Zijun Li
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Lulu Yao
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Kandasamy Saravanakumar
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Nguyen Thi Thanh Thuy
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Yunyeong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Chang Xue
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| | - Xiaohui Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University1210 University Town, Wenzhou 325035, Zhejiang, China
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National UniversityGwangju 61186, Korea
| |
Collapse
|
37
|
Prasad K, Saggam A, Guruprasad KP, Tillu G, Patwardhan B, Satyamoorthy K. Molecular mechanisms of Asparagus racemosus willd. and Withania somnifera (L.) Dunal as chemotherapeutic adjuvants for breast cancer treatment. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118261. [PMID: 38685363 DOI: 10.1016/j.jep.2024.118261] [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: 02/07/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Despite various treatment modalities, the progression and metastasis of breast cancer (BC) are grave concerns due to the alarming disease-free survival rate (DFS) and overall survival rate (OS) of affected patients. Over the years, many antibiotics, synthetic compounds, medicinal plant isolates and polyherbal combinations have been used as adjuvants in therapy for the management of primary and secondary tumors. Paclitaxel (PTX)-based chemotherapy for breast cancer causes multiple adverse side effects in patients. Withania somnifera (L.) Dunal (WS) and Asparagus racemosus Willd. (AR) as Ayurveda-inspired plant-based adjuvants were investigated for their anticancer effects on MDA-MB-231 and 4T1 cells in mouse model systems. AIM OF THE STUDY This study focused on evaluating the adjuvant properties of WS and AR plant extracts with PTX and their effectiveness over PTX alone in terms of tumor inhibition. MATERIALS AND METHODS The effects of WS and AR on DNA double-strand breaks (DSBs), senescence induction and mitochondrial functions were evaluated in BC cells in vitro. The potential for cancer stem cell (CSC) inhibition was evaluated via mammosphere formation assays and CD44/CD24 immunostaining. In vivo tumor growth studies were conducted in athymic BALB/c mice for MDA-MB-231 cells and in BALB/c mice for 4T1 cells. RESULTS Induction of senescence was evident due to DSBs induced by the WS and AR extracts. Mammosphere formation and CD44/CD24 CSC markers were reduced after treatment with WS, AR or the combination of both in MCF-7 cells. WS or AR inhibited epithelial-to-mesenchymal transition (EMT). In vivo studies demonstrated that tumor growth inhibition was more pronounced in the treated group than in the PTX alone group and the untreated control group. CONCLUSION Our study showed that the use of WS or AR plant hydroalcoholic extracts in combination with paclitaxel (PTX) has better effects on sensitivity and efficacy than PTX alone, as demonstrated in in vitro BC cells and mouse models with BC cell grafts. Hence, scheduling adjuvant therapy with WS or AR alone or combined with PTX can be advantageous for the management of triple-negative BC (TNBC). Further studies are warranted in human clinical conditions to ascertain the efficacy of these treatments.
Collapse
Affiliation(s)
- Keshava Prasad
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| | - Akash Saggam
- Ayush Center of Excellence, Center for Complementary and Integrative Health, School of Health Sciences, Savitribai Phule Pune University, Pune, 411007, India.
| | - Kanive Parashiva Guruprasad
- Centre for Ayurvedic Biology, Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| | - Girish Tillu
- Ayush Center of Excellence, Center for Complementary and Integrative Health, School of Health Sciences, Savitribai Phule Pune University, Pune, 411007, India.
| | - Bhushan Patwardhan
- Ayush Center of Excellence, Center for Complementary and Integrative Health, School of Health Sciences, Savitribai Phule Pune University, Pune, 411007, India.
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India; SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Manjushree Nagar, Sattur, Dharwad, Karnataka, 580009, India.
| |
Collapse
|
38
|
Kim D, Go SH, Song Y, Lee DK, Park JR. Decursin Induces G1 Cell Cycle Arrest and Apoptosis through Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress in Human Colorectal Cancer Cells in In Vitro and Xenograft Models. Int J Mol Sci 2024; 25:9939. [PMID: 39337425 PMCID: PMC11432441 DOI: 10.3390/ijms25189939] [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: 08/26/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Decursin, a coumarin isolated from Angelica gigas Nakai, possesses anti-inflammatory and anti-cancer properties. However, the molecular mechanisms underlying its anti-cancer effects against human colorectal cancer (CRC) are unclear. Therefore, this study aimed to evaluate the biological activities of decursin in CRC in vitro and in vivo and to determine its underlying mechanism of action. Decursin exhibited anti-tumor activity in vitro, accompanied by an increase in G1 cell cycle arrest and apoptosis in HCT-116 and HCT-8 CRC cells. Decursin also induced the production of reactive oxygen species (ROS), thereby activating the endoplasmic reticulum (ER) stress apoptotic pathway in CRC cells. Furthermore, the role of ROS in decursin-induced apoptosis was investigated using the antioxidant N-acetyl-L-cysteine. Inhibiting ROS production reversed decursin-induced ER stress. Moreover, decursin significantly suppressed tumor growth in a subcutaneous xenograft mouse model of HCT-116 and HCT-8 CRC cells without causing host toxicity. Decursin also decreased cell proliferation, as documented by Ki-67, and partly increased cleaved caspase 3 expression in tumor tissues by activating ER stress apoptotic pathways. These findings suggest that decursin induces cell cycle arrest and apoptosis in human CRC cells via ROS-mediated ER stress, suggesting that decursin could be a therapeutic agent for CRC.
Collapse
Affiliation(s)
| | | | | | - Dong-Keon Lee
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea; (D.K.); (S.-H.G.); (Y.S.)
| | - Jeong-Ran Park
- Division of Research Center, Scripps Korea Antibody Institute, Chuncheon 24341, Republic of Korea; (D.K.); (S.-H.G.); (Y.S.)
| |
Collapse
|
39
|
Affranchi F, Di Liberto D, Lauricella M, D’Anneo A, Calvaruso G, Pratelli G, Carlisi D, De Blasio A, Tesoriere L, Giuliano M, Notaro A, Emanuele S. The Antitumor Potential of Sicilian Grape Pomace Extract: A Balance between ROS-Mediated Autophagy and Apoptosis. Biomolecules 2024; 14:1111. [PMID: 39334877 PMCID: PMC11430817 DOI: 10.3390/biom14091111] [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: 07/26/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
From the perspective of circular economy, it is extremely useful to recycle waste products for human health applications. Among the health-beneficial properties of bioactive phyto-compounds, grape pomace represents a precious source of bioactive molecules with potential antitumor properties. Here, we describe the effects of a Sicilian grape pomace hydroalcoholic extract (HE) in colon and breast cancer cells. The characterization of HE composition revealed the predominance of anthoxanthins and phenolic acids. HE treatment was more effective in reducing the viability of colon cancer cells, while breast cancer cells appeared more resistant. Indeed, while colon cancer cells underwent apoptosis, as shown by DNA fragmentation, caspase-3 activation, and PARP1 degradation, breast cancer cells seemed to not undergo apoptosis. To elucidate the underlying mechanisms, reactive oxygen species (ROS) were evaluated. Interestingly, ROS increased in both cell lines but, while in colon cancer, cells' ROS rapidly increased and progressively diminished over time, in breast cancer, cells' ROS increase was persistent up to 24 h. This effect was correlated with the induction of pro-survival autophagy, demonstrated by autophagosomes formation, autophagic markers increase, and protection by the antioxidant NAC. The autophagy inhibitor bafilomycin A1 significantly increased the HE effects in breast cancer cells but not in colon cancer cells. Overall, our data provide evidence that HE efficacy in tumor cells depends on a balance between ROS-mediated autophagy and apoptosis. Therefore, inhibiting pro-survival autophagy may be a tool to target those cells that appear more resistant to the effect of HE.
Collapse
Affiliation(s)
- Federica Affranchi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Diana Di Liberto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (D.D.L.); (M.L.); (G.P.); (D.C.)
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (D.D.L.); (M.L.); (G.P.); (D.C.)
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Giuseppe Calvaruso
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Giovanni Pratelli
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (D.D.L.); (M.L.); (G.P.); (D.C.)
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (D.D.L.); (M.L.); (G.P.); (D.C.)
| | - Anna De Blasio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Antonietta Notaro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (F.A.); (A.D.); (G.C.); (A.D.B.); (L.T.); (M.G.)
| | - Sonia Emanuele
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy; (D.D.L.); (M.L.); (G.P.); (D.C.)
| |
Collapse
|
40
|
Fakhri S, Moradi SZ, Moradi SY, Piri S, Shiri Varnamkhasti B, Piri S, Khirehgesh MR, Bishayee A, Casarcia N, Bishayee A. Phytochemicals regulate cancer metabolism through modulation of the AMPK/PGC-1α signaling pathway. BMC Cancer 2024; 24:1079. [PMID: 39223494 PMCID: PMC11368033 DOI: 10.1186/s12885-024-12715-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Due to the complex pathophysiological mechanisms involved in cancer progression and metastasis, current therapeutic approaches lack efficacy and have significant adverse effects. Therefore, it is essential to establish novel strategies for combating cancer. Phytochemicals, which possess multiple biological activities, such as antioxidant, anti-inflammatory, antimutagenic, immunomodulatory, antiproliferative, anti-angiogenesis, and antimetastatic properties, can regulate cancer progression and interfere in various stages of cancer development by suppressing various signaling pathways. METHODS The current systematic and comprehensive review was conducted based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria, using electronic databases, including PubMed, Scopus, and Science Direct, until the end of December 2023. After excluding unrelated articles, 111 related articles were included in this systematic review. RESULTS In this current review, the major signaling pathways of cancer metabolism are highlighted with the promising anticancer role of phytochemicals. This was through their ability to regulate the AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) signaling pathway. The AMPK/PGC-1α signaling pathway plays a crucial role in cancer cell metabolism via targeting energy homeostasis and mitochondria biogenesis, glucose oxidation, and fatty acid oxidation, thereby generating ATP for cell growth. As a result, targeting this signaling pathway may represent a novel approach to cancer treatment. Accordingly, alkaloids, phenolic compounds, terpene/terpenoids, and miscellaneous phytochemicals have been introduced as promising anticancer agents by regulating the AMPK/PGC-1α signaling pathway. Novel delivery systems of phytochemicals targeting the AMPK/PGC-1α pathway in combating cancer are also highlighted in this review.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Yahya Moradi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Sarina Piri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Behrang Shiri Varnamkhasti
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Mohammad Reza Khirehgesh
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | | | - Nicolette Casarcia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
41
|
Iqbal Lone W, Chand J, Kumar P, Garg Y, Ahmed Z, Mukherjee D, Goswami A, Momo H Anãl J. Discovery of colchicine aryne cycloadduct as a potent molecule for the abrogation of epithelial to mesenchymal transition via modulating cell cycle regulatory CDK-2 and CDK-4 kinases in breast cancer cells. Bioorg Chem 2024; 150:107581. [PMID: 38908129 DOI: 10.1016/j.bioorg.2024.107581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
In this study, we synthesized a new-generation library of colchicine derivatives via cycloaddition of colchicine utilizing position C-8 and C-12 diene system regioselectivity with aryne precursor to generate a small, focused library of derivatives. We assessed their anticancer activity against various cancer cell lines like MCF-7, MDA-MB-231, MDA-MB-453, and PC-3. Normal human embryonic kidney cell line HEK-293 was used to determine the toxicity. Among these derivatives, silicon-tethered compound B-4a demonstrated the highest potency against breast cancer cells. Subsequent mechanistic studies revealed that B-4a effectively modulates cell cycle regulatory kinases (CDK-2 and CDK-4) and their associated cyclins (cyclin-B1, cyclin-D1), inducing apoptosis. Additionally, B-4a displayed a noteworthy impact on tubulin polymerization, compared to positive control flavopiridol hydrochloride in a dose-dependent manner, and significantly disrupted the vimentin cytoskeleton, contributing to G1 arrest in breast cancer cells. Moreover, B-4a exhibited substantial anti-metastatic properties by inhibiting breast cancer cell migration and invasion. These effects are attributed to the down-regulation of major epithelial to mesenchymal transition (EMT) factors, including vimentin and Twist-1, and the upregulation of the epithelial marker E-cadherin in an apoptosis-dependent manner.
Collapse
Affiliation(s)
- Waseem Iqbal Lone
- Natural Products and Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jagdish Chand
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Puneet Kumar
- Natural Products and Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Yashi Garg
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debaraj Mukherjee
- Department of Chemical Sciences, Bose Institute, EN-80, Sector V, Kolkata 700091, WB, India
| | - Anindya Goswami
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Jasha Momo H Anãl
- Natural Products and Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
42
|
Al Azzani M, Nizami ZN, Magramane R, Sekkal MN, Eid AH, Al Dhaheri Y, Iratni R. Phytochemical-mediated modulation of autophagy and endoplasmic reticulum stress as a cancer therapeutic approach. Phytother Res 2024; 38:4353-4385. [PMID: 38961675 DOI: 10.1002/ptr.8283] [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: 01/20/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Autophagy and endoplasmic reticulum (ER) stress are conserved processes that generally promote survival, but can induce cell death when physiological thresholds are crossed. The pro-survival aspects of these processes are exploited by cancer cells for tumor development and progression. Therefore, anticancer drugs targeting autophagy or ER stress to induce cell death and/or block the pro-survival aspects are being investigated extensively. Consistently, several phytochemicals have been reported to exert their anticancer effects by modulating autophagy and/or ER stress. Various phytochemicals (e.g., celastrol, curcumin, emodin, resveratrol, among others) activate the unfolded protein response to induce ER stress-mediated apoptosis through different pathways. Similarly, various phytochemicals induce autophagy through different mechanisms (namely mechanistic target of Rapamycin [mTOR] inhibition). However, phytochemical-induced autophagy can function either as a cytoprotective mechanism or as programmed cell death type II. Interestingly, at times, the same phytochemical (e.g., 6-gingerol, emodin, shikonin, among others) can induce cytoprotective autophagy or programmed cell death type II depending on cellular contexts, such as cancer type. Although there is well-documented mechanistic interplay between autophagy and ER stress, only a one-way modulation was noted with some phytochemicals (carnosol, capsaicin, cryptotanshinone, guangsangon E, kaempferol, and δ-tocotrienol): ER stress-dependent autophagy. Plant extracts are sources of potent phytochemicals and while numerous phytochemicals have been investigated in preclinical and clinical studies, the search for novel phytochemicals with anticancer effects is ongoing from plant extracts used in traditional medicine (e.g., Origanum majorana). Nonetheless, the clinical translation of phytochemicals, a promising avenue for cancer therapeutics, is hindered by several limitations that need to be addressed in future studies.
Collapse
Affiliation(s)
- Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rym Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed N Sekkal
- Department of Surgery, Specialty Orthopedic, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
43
|
Yadav M, Kandhari K, Mathan SV, Ali M, Singh RP. Fisetin induces G2/M phase arrest and caspase-mediated cleavage of p21 Cip1 and p27 Kip1 leading to apoptosis and tumor growth inhibition in HNSCC. Mol Carcinog 2024; 63:1697-1711. [PMID: 38801393 DOI: 10.1002/mc.23754] [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: 01/14/2024] [Revised: 04/27/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
The anticancer potential and associated mechanisms of flavonoid fisetin are yet to be fully investigated on human head and neck squamous cell carcinoma (HNSCC). In the present study, fisetin (25-75 µM for 24-48 h) dose-dependently inhibited growth and induced death in HNSCC Cal33 and UM-SCC-22B cells, without showing any death in normal cells. Fisetin (25-50 µM) induced G2/M phase arrest via decrease in Cdc25C, CDK1, cyclin B1 expression, and an increase in p53(S15). A concentration-dependent increase in fisetin-induced DNA damage and apoptosis in HNSCC cells was authenticated by comet assay, gamma-H2A.X(S139) phosphorylation, and marked cleavage of PARP protein. Interestingly, fisetin-induced cell death occurred independently of p53 and reactive oxygen species production. The activation of JNK and inhibition of PI3K/Akt, ERK1/2, EGFR, and STAT-3 signaling were identified. Further, fisetin-induced apoptosis was mediated, in part, via p21Cip1 and p27Kip1 cleavage by caspase, which was reversed by z-VAD-FMK, a pan-caspase inhibitor. Subsequently, fisetin was also found to induce autophagy; nevertheless, autophagy attenuation exaggerated apoptosis. Oral fisetin (50 mg/kg body weight) treatment to establish Cal33 xenograft in mice for 19 days showed 73% inhibition in tumor volume (p < 0.01) along with a decrease in Ki67-positive cells and an increase in cleaved caspase-3 level in tumors. Consistent with the effect of 50 µM fisetin in vitro, the protein levels of p21Cip1 and P27Kip1 were also decreased by fisetin in tumors. Together, these findings showed strong anticancer efficacy of fisetin against HNSCC with downregulation of EGFR-Akt/ERK1/2-STAT-3 pathway and activation of JNK/c-Jun, caspases and caspase-mediated cleavage of p21Cip1 and p27Kip1.
Collapse
Affiliation(s)
- Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Kushal Kandhari
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sivapar V Mathan
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mansoor Ali
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
44
|
Banerjee S, Banerjee S, Bishayee A, Da Silva MN, Sukocheva OA, Tse E, Casarcia N, Bishayee A. Cellular and molecular mechanisms underlying the potential of betulinic acid in cancer prevention and treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155858. [PMID: 39053249 DOI: 10.1016/j.phymed.2024.155858] [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: 03/02/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Betulinic acid (BA), which is a pentacyclic triterpenoid found in the bark of plane, birch, and eucalyptus trees, has emerged as a compound of significant interest in scientific research due to its potential therapeutic applications. BA has a range of well-documented pharmacological and biological effects, including antibacterial, immunomodulatory, diuretic, antiviral, antiparasitic, antidiabetic, and anticancer activities. Although numerous research studies have explored the potential anticancer effects of BA, there is a noticeable gap in the literature, highlighting the need for a more up-to-date and comprehensive evaluation of BA's anticancer potential. PURPOSE The aim of this work is to critically assess the reported cellular and molecular mechanisms underlying the cancer preventive and therapeutic effects of BA. METHODS Relevant research on the inhibitory effects of BA against cancerous cells was searched using Science Direct, Scopus, Web of Science, and PubMed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS The anticancer properties of BA are mediated by the activation of cell death and cell cycle arrest, production of reactive oxygen species, increased mitochondrial permeability, modulation of nuclear factor-κB and Bcl-2 family signaling. Emerging evidence also underscores the combined anticancer effects of BA with other natural bioactive compounds or approved drugs. Notably, several novel BA nanoformulations have been found to exhibit encouraging antineoplastic activities. CONCLUSION BA, whether used alone or in combination, or as a form of nanoformulation, shows significant potential for cancer prevention and treatment. Nevertheless, further detailed studies are necessary to confirm the therapeutic effectiveness of this natural compound.
Collapse
Affiliation(s)
- Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, West Bengal, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, West Bengal, India
| | | | - Milton Nascimento Da Silva
- Laboratory of Liquid Chromatography, Institute of Exact and Natural Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; Chemistry Post-Graduation Program, Institute of Exact and Natural Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; Pharmaceutical Science Post-Graduation Program, Institute of Health Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Olga A Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Nicolette Casarcia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
45
|
Sofi MA, Sofi MA, Nanda A, Thiruvengadam K, Nayak BK. Investigating the Medicinal Potential of Lavatera cashmeriana Leaf Extract: Phytochemical Profiling and In Vitro Evaluation of Antimicrobial, Antioxidant, and Anticancer Activities. Adv Pharmacol Pharm Sci 2024; 2024:5301687. [PMID: 39220824 PMCID: PMC11366056 DOI: 10.1155/2024/5301687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
This study investigated the medicinal potential of Lavatera cashmeriana, a plant traditionally known for its therapeutic properties. The aim was to identify the phytocompounds in L. cashmeriana leaf extract and evaluate its antibacterial, antioxidant, and anticancer effects. Gas chromatography-mass spectrometry analysis was employed to characterize the phytochemical composition of the ethanol extract derived from L. cashmeriana leaves. The antimicrobial potential was assessed through the well diffusion technique, targeting Escherichia coli, Enterococcus faecalis, Pseudomonas aeruginosa, Staphylococcus aureus, and Candida albicans. The 2,2-diphenyl-1-picrylhydrazyl assay was conducted to assess antioxidant capabilities, while cytotoxicity against the A549 cancer cell line was determined via the MTT assay. GC-MS analysis identified ten different compounds, with phytol, 1-Eicosanol, and 2,6,10-trimethyl,14-ethylene-14-pentadecne being the most prevalent. The extract exhibited notable antimicrobial efficacy against all bacteria with MIC values ranging from 62.5 to 250 µg/mL. However, C. albicans did not respond. The extract exhibited antioxidative properties with an IC50 value of 86 µg/mL and cytotoxicity with an IC50 value of 69.95 µg/mL against the A549 cancer cell line. The results derived from this study supported the historical use of L. cashmeriana as a medicinal plant and suggested that it can potentially treat a wide range of medical ailments. The identified phytocompounds and the demonstrated antibacterial, antioxidant, and anticancer effects provide scientific evidence for its medicinal properties. However, further investigations are needed to fully understand its safety profile, efficacy, and mechanism of action before recommending it for therapeutic purposes.
Collapse
Affiliation(s)
- Mohmmad Ashaq Sofi
- Department of Biomedical EngineeringSathyabama Institute of Science & Technology, Chennai 600119, Tamil Nadu, India
| | - Mohd Abass Sofi
- Department of ChemistrySathyabama Institute of Science & Technology, Chennai 600119, Tamil Nadu, India
| | - Anima Nanda
- Department of Biomedical EngineeringSathyabama Institute of Science & Technology, Chennai 600119, Tamil Nadu, India
| | - Kasi Thiruvengadam
- Biocontrol and Microbial Metabolites LabCentre for Advanced Studies in BotanyUniversity of MadrasGuindy Campus, Chennai, India
| | - B. K. Nayak
- Department of BotanyK. M. Govt. Institute for Postgraduate Studies and Research (Autonomous), Puducherry 605008, India
| |
Collapse
|
46
|
Sharma V, Chaudhary AA, Bawari S, Gupta S, Mishra R, Khan SUD, Ali MAM, Shahid M, Srivastava S, Verma D, Gupta A, Kumar S, Kumar S. Unraveling cancer progression pathways and phytochemical therapeutic strategies for its management. Front Pharmacol 2024; 15:1414790. [PMID: 39246660 PMCID: PMC11377287 DOI: 10.3389/fphar.2024.1414790] [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: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 09/10/2024] Open
Abstract
Cancer prevention is currently envisioned as a molecular-based approach to prevent carcinogenesis in pre-cancerous stages, i.e., dysplasia and carcinoma in situ. Cancer is the second-leading cause of mortality worldwide, and a more than 61% increase is expected by 2040. A detailed exploration of cancer progression pathways, including the NF-kβ signaling pathway, Wnt-B catenin signaling pathway, JAK-STAT pathway, TNF-α-mediated pathway, MAPK/mTOR pathway, and apoptotic and angiogenic pathways and effector molecules involved in cancer development, has been discussed in the manuscript. Critical evaluation of these effector molecules through molecular approaches using phytomolecules can intersect cancer formation and its metastasis. Manipulation of effector molecules like NF-kβ, SOCS, β-catenin, BAX, BAK, VEGF, STAT, Bcl2, p53, caspases, and CDKs has played an important role in inhibiting tumor growth and its spread. Plant-derived secondary metabolites obtained from natural sources have been extensively studied for their cancer-preventing potential in the last few decades. Eugenol, anethole, capsaicin, sanguinarine, EGCG, 6-gingerol, and resveratrol are some examples of such interesting lead molecules and are mentioned in the manuscript. This work is an attempt to put forward a comprehensive approach to understanding cancer progression pathways and their management using effector herbal molecules. The role of different plant metabolites and their chronic toxicity profiling in modulating cancer development pathways has also been highlighted.
Collapse
Affiliation(s)
- Vikas Sharma
- Metro College of Health Sciences and Research, Greater Noida, India
- School of Pharmacy, Sharda University, Greater Noida, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sweta Bawari
- Amity Institute of Pharmacy, Amity University, Noida, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, India
| | - Richa Mishra
- Department of Computer Engineering, Parul University, Vadodara, India
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Devvrat Verma
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| | - Arti Gupta
- Lloyd School of Pharmacy, Greater Noida, India
| | - Sanjay Kumar
- Biological and Bio-computational Laboratory, Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India
- DST-FIST Laboratory, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| |
Collapse
|
47
|
Zhao L, Ma H, Jiang Y, Li Y, Qiao L, Chen Y, Jiang X, Wang L, Wang S, Fan X. Identification of an m6A Natural Inhibitor, Lobeline, That Reverses Lenvatinib Resistance in Hepatocellular Tumors. JOURNAL OF NATURAL PRODUCTS 2024; 87:1983-1993. [PMID: 39136667 DOI: 10.1021/acs.jnatprod.4c00406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive cancer that has an effect on human health. As a first-line drug for HCC, despite its excellent efficacy, lenvatinib (Len) is prone to developing drug resistance in HCC patients. The N6-methyladenosine (m6A) modification is not only related to the development of HCC but also shows great potential in overcoming HCC resistance. Using Dot Blot, our group first screened a small molecule m6A regulator, lobeline (Lob), from a library of 390 compounds (mostly natural products). In vitro experiments demonstrated that Lob could significantly enhance the sensitivity to Len of Len-resistant HCC (HCC/Len) and inhibit migration of resistant cells. In Len-resistant cell-derived and patient-derived xenograft models, Lob could reverse the resistant phenotype, with reductions in tumor volume of 68% and 60%, respectively. Furthermore, MeRIP-m6A sequencing results indicated that the underlying molecular mechanism of Lob reversal of HCC drug resistance was related to UBE3B. Taken together, this study highlighted that Lob, a plant derived natural product, could reverse the resistance of HCC to Len by regulating the m6A levels. It is hoped that this will provide a pharmacological research basis for the clinical treatment of HCC patients.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Heyao Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Yuhui Jiang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingying Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Qiao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaowen Jiang
- Department of Analysis and Pharmacology of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shu Wang
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xinyu Fan
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| |
Collapse
|
48
|
Wang G, Guo C, Pi H, Wang Y, Lin S, Bi K, Zhang M, Wang N, Zhao G. Elucidation of the anti-colorectal cancer mechanism of Atractylodes lancea by network pharmacology and experimental verification. Aging (Albany NY) 2024; 16:12008-12028. [PMID: 39177661 PMCID: PMC11386916 DOI: 10.18632/aging.206075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 07/13/2024] [Indexed: 08/24/2024]
Abstract
Atractylodes lancea which was listed in "Shennong's Materia Medica" and could be used to treat gastrointestinal-associated diseases. However, its roles, core and active ingredients, and mechanisms in treatment of colorectal cancer (CRC) were still unknown. Therefore, network pharmacology and experimental validation were used to clarify the effects, core active ingredients and molecular mechanisms of Atractylodes lancea. We found that Atractylodes lancea has 28 effective active components and 213 potential targets. Seventy-three genes which demonstrate interaction between the Atractylodes lancea and CRC were confirmed. Enrichment analysis showed that 2033 GO biological process items and 144 KEGG pathways. Survival and molecular docking analysis revealed that luteolin as the core component interacted with these genes (Matrix metalloproteinase 3 (MMP3), Matrix metalloproteinase 9 (MMP9), Tissue inhibitor of metalloproteinases 1 (TIMP1), Vascular endothelial growth factor A (VEGFA)) with the lowest binding energy, and these genes were involved in building a prognostic model for CRC. Cellular phenotyping experiments showed that luteolin could inhibit the proliferation and migration of CRC cells and downregulate the expression of MMP3, MMP9, TIMP1, VEGFA probably by Phosphoinositide 3-kinase/ serine/threonine kinase Akt (PI3K/AKT) pathway. To conclude, Atractylodes lancea could inhibit proliferation and migration of CRC cells through its core active ingredient (luteolin) to suppress the expression of MMP3, MMP9, TIMP1, VEGFA probably by PI3K/AKT pathway.
Collapse
Affiliation(s)
- Guangliang Wang
- Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, Guangdong, China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541000, Guangxi, China
| | - Chuangchuang Guo
- Faculty of Public Health, Guilin Medical University, Guilin 541000, Guangxi, China
| | - Hui Pi
- Faculty of Basic Medical Sciences, Dali University, Dali 671003, Yunnan, China
| | - Yu Wang
- Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, Guangdong, China
| | - Shuyun Lin
- Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, Guangdong, China
| | - Keyi Bi
- Department of Pharmacy, Guilin Medical University, Guilin 541000, Guangxi, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, Guangdong, China
| | - Na Wang
- Faculty of Public Health, Guilin Medical University, Guilin 541000, Guangxi, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, Guangdong, China
| |
Collapse
|
49
|
Yıldırım M, Erşatır M, Poyraz S, Amangeldinova M, Kudrina NO, Terletskaya NV. Green Extraction of Plant Materials Using Supercritical CO 2: Insights into Methods, Analysis, and Bioactivity. PLANTS (BASEL, SWITZERLAND) 2024; 13:2295. [PMID: 39204731 PMCID: PMC11359946 DOI: 10.3390/plants13162295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
In recent years, the supercritical CO2 extraction method has gained attention due to its use of environmentally friendly, non-toxic solvents, ability to operate at lower temperatures that do not cause the degradation of bioactive compounds, and capacity for rapid extraction. This method is particularly notable for isolating bioactive compounds from plants. The extracts obtained have shown superior properties due to their activity against diseases such as cancer, which is one of the leading causes of death worldwide. The aim of this study is to provide an in-depth understanding of the supercritical CO2 extraction method, as well as to discuss its advantages and disadvantages. Furthermore, the study includes specific data on various plant materials, detailing the following parameters: plant name and region, bioactive compounds or compound classes, extraction temperature (°C), pressure (bar), time (minutes), co-solvent used, and flow rate. Additionally, this study covers extensive research on the isolation of bioactive compounds and the efficacy of the obtained extracts against cancer.
Collapse
Affiliation(s)
- Metin Yıldırım
- Department of Biochemistry, Faculty of Pharmacy, Harran University, Sanliurfa 63050, Türkiye
| | - Mehmet Erşatır
- Department of Chemistry, Faculty of Art and Science, Cukurova University, Adana 01330, Türkiye;
| | - Samet Poyraz
- Independent Researcher, Nevşehir 50040, Türkiye;
| | - Madina Amangeldinova
- Department of Biodiversity and Biological Resources, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Al-Farabi Av., 71, Almaty 050040, Kazakhstan; (M.A.); (N.O.K.); (N.V.T.)
- Institute of Genetic and Physiology, Al-Farabi Av., 93, Almaty 050040, Kazakhstan
| | - Nataliya O. Kudrina
- Department of Biodiversity and Biological Resources, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Al-Farabi Av., 71, Almaty 050040, Kazakhstan; (M.A.); (N.O.K.); (N.V.T.)
- Institute of Genetic and Physiology, Al-Farabi Av., 93, Almaty 050040, Kazakhstan
| | - Nina V. Terletskaya
- Department of Biodiversity and Biological Resources, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Al-Farabi Av., 71, Almaty 050040, Kazakhstan; (M.A.); (N.O.K.); (N.V.T.)
- Institute of Genetic and Physiology, Al-Farabi Av., 93, Almaty 050040, Kazakhstan
| |
Collapse
|
50
|
Hadkar VM, Mohanty C, Selvaraj CI. Biopolymeric nanocarriers in cancer therapy: unleashing the potency of bioactive anticancer compounds for enhancing drug delivery. RSC Adv 2024; 14:25149-25173. [PMID: 39139249 PMCID: PMC11317881 DOI: 10.1039/d4ra03911d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Effective cancer treatment is becoming a global concern, and recent developments in nanomedicine are essential for its treatment. Cancer is a severe metabolic syndrome that affects the human population and is a significant contributing factor to deaths globally. In science, nanotechnology offers rapidly developing delivery methods for natural bioactive compounds that are becoming increasingly prominent and can be used to treat diseases in a site-specific way. Chemotherapy and radiotherapy are conventional approaches for preventing cancer progression and have adverse effects on the human body. Many chemically synthesized drugs are used as anticancer agents, but they have several side effects; hence, they are less preferred. Medicinal plants and marine microorganisms represent a vast, mostly untapped reservoir of bioactive compounds for cancer treatment. However, they have several limitations, including nonspecific targeting, weak water solubility and limited therapeutic potential. An alternative option is the use of biopolymeric nanocarriers, which can generate effective targeted treatment therapies when conjugated with natural anticancer compounds. The present review focuses on biopolymeric nanocarriers utilizing natural sources as anticancer drugs with improved tumor-targeting efficiency. This review also covers various natural anticancer compounds, the advantages and disadvantages of natural and synthetic anticancer compounds, the problems associated with natural anticancer drugs and the advantages of biopolymeric nanocarriers over synthetic nanocarriers as drug delivery agents. This review also discusses various biopolymeric nanocarriers for enhancing the controlled delivery of anticancer compounds and the future development of nanomedicines for treating cancer.
Collapse
Affiliation(s)
- Vrushali Manoj Hadkar
- School of Biosciences and Technology, Vellore Institute of Technology (VIT) Vellore 632014 Tamil Nadu India
| | - Chirasmita Mohanty
- School of Biosciences and Technology, Vellore Institute of Technology (VIT) Vellore 632014 Tamil Nadu India
| | - Chinnadurai Immanuel Selvaraj
- Department of Genetics and Plant Breeding, VIT School of Agricultural Sciences and Advanced Learning (VAIAL), VIT Vellore 632014 Tamil Nadu India
| |
Collapse
|