1
|
Yang X, Tan J, Zhao R, Duan T, Yang J, Chen B, Yang X, Wang J, Jiang C, Ni G, Zhang Y, He Y. Arsenic-induced circASXL1 regulates 16HBE cell proliferation through the P65 signaling axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118298. [PMID: 40349468 DOI: 10.1016/j.ecoenv.2025.118298] [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: 12/05/2024] [Revised: 04/09/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Arsenic is a well-known environmental toxicant, strongly associated with severe toxicity across multiple organs, including the lungs. Circular RNAs (circRNAs) are critical in various cellular processes and linked to disease due to their highly stable covalent closed-loop structure, making them potential therapeutic targets. This study aimed to investigate the expression of circASXL1 under arsenic exposure and its role in 16HBE cell proliferation upon circASXL1 knockdown. Treatment with sodium arsenite increased circASXL1 expression in 16HBE cells, while metabolites had no effect. Notably, circASXL1 knockdown enhanced cell viability and proliferation, concomitant with coordinated activation of STAT3 and P65. Although the precise mechanism requires further validation, our Western blot analyses suggested that STAT3 activation may promote P65 transcriptional activity, as evidenced by upregulated expression of its downstream targets PCNA, BCL2, BCL-XL, and CIAP1. Intriguingly, combinatorial treatment with arsenic and circASXL1-specific siRNAs attenuated both cell proliferation and P65 activation. Our findings propose that arsenic modulates circASXL1 to engage STAT3-P65 crosstalk. This study establishes the arsenic-circASXL1 axis as a novel regulator of STAT3/P65 signaling networks, providing mechanistic insights for combating arsenic-induced pulmonary pathologies.
Collapse
Affiliation(s)
- Xuefei Yang
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Jingwen Tan
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Ruihuan Zhao
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Tingzhi Duan
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Jingchao Yang
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Bin Chen
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Xinda Yang
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Jinhua Wang
- Bijie Weining Autonomous County Maternal and Child Health Hospital, China
| | - Chenglan Jiang
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China
| | - Guanghui Ni
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, China
| | - Yanliang Zhang
- Department of Clinical Laboratory, The First Afliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Yuefeng He
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
2
|
Xu B, Zhou L, Zhang Q. Curcumin Inhibits the Progression of Non-small Cell Lung Cancer by Regulating DMRT3/SLC7A11 Axis. Mol Biotechnol 2025; 67:1880-1892. [PMID: 38744789 DOI: 10.1007/s12033-024-01166-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: 01/15/2024] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a fatal malignancy all over the world. Emerging studies have shown that curcumin might repress NSCLC progression by regulating ferroptosis, but the underlying mechanism remains unclear. 16HBE, LK-2, and H1650 cell viability was detected using Cell Counting Kit-8 assay. LK-2 and H1650 cell proliferation, apoptosis, and angiopoiesis were measured using 5-ethynyl-2'-deoxyuridine, flow cytometry, and tube formation assay. Superoxide dismutase, Malondialdehyde, Glutathione, and lactate dehydrogenase levels in LK-2 and H1650 cells were examined using special assay kits. Fe+ level was assessed using an iron assay kit. Doublesex and Mab-3 related Transcription Factor 3 (DMRT3) and solute carrier family 7 member 11 (SLC7A11) protein levels were detected using western in NSCLC tissues, adjacent matched normal tissues, 16HBE cells, LK-2 cells, H1650 cells, and xenograft tumor tissues. Glutathione peroxidase 4, Acyl-CoA Synthetase Long Chain Family Member 4, and transferrin receptor 1 protein levels in LK-2 and H1650 cells were examined by western blot assay. DMRT3 and SLC7A11 levels were determined using real-time quantitative polymerase chain reaction. After JASPAR prediction, binding between DMRT3 and SLC7A11 promoter was verified using Chromatin immunoprecipitation and dual-luciferase reporter assays in LK-2 and H1650 cells. Role of curcumin on NSCLC tumor growth was assessed using the xenograft tumor model in vivo. Curcumin blocked NSCLC cell proliferation and angiopoiesis, and induced apoptosis and ferroptosis. DMRT3 or SLC7A11 upregulation partly abolished the suppressive role of curcumin on NSCLC development. In mechanism, DMRT3 was a transcription factor of SLC7A11 and increased the transcription of SLC7A11 via binding to its promoter region. Curcumin inhibited NSCLC growth in vivo by modulating DMRT3. Curcumin might constrain NSCLC cell malignant phenotypes partly through the DMRT3/SLC7A11 axis, providing a promising therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Bin Xu
- Department of TCM, Changzhou Cancer Hospital, No.68, Honghe Road, Xinbei District, Changzhou City, 213000, Jiangsu, China
| | - Li Zhou
- Department of TCM, Changzhou Cancer Hospital, No.68, Honghe Road, Xinbei District, Changzhou City, 213000, Jiangsu, China
| | - Qian Zhang
- Department of TCM, Changzhou Cancer Hospital, No.68, Honghe Road, Xinbei District, Changzhou City, 213000, Jiangsu, China.
| |
Collapse
|
3
|
Zhang Y, Wang L, Zeng J, Shen W. Research advances in polyphenols from Chinese herbal medicine for the prevention and treatment of chronic obstructive pulmonary disease: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03945-y. [PMID: 40035820 DOI: 10.1007/s00210-025-03945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a global health problem due to its high death and morbidity worldwide, which is characterized by an incompletely reversible limitation in airflow that is not fully reversible. Unfortunately, Western medical treatments are unable to reverse the progressive decline in lung function. Importantly, polyphenolic compounds isolated from Chinese herbal medicine exhibited therapeutic/interventional effects on COPD in preclinical studies. This review systematically analyzed the pathogenesis of COPD, such as inflammation, oxidative stress, protease/antiprotease imbalance, aging, cell death, and dysbiosis of gut microbiota. Moreover, this review summarized the regulatory mechanisms of natural polyphenolic compounds for the treatment of COPD. Several studies have demonstrated that natural polyphenolic compounds have therapeutic effects on COPD by regulating various biological processes, such as anti-inflammatory, reduction of oxidative damage, anti-cell death, and inhibition of airway hyperglycemia. Mechanistically, this review found that the promising effects of natural polyphenolic compounds on COPD were mainly achieved through modulating the NF-κB and MAPK inflammatory pathways, the Nrf2 oxidative stress pathway, and the SIRT1/PGC-1α lung injury pathway. Furthermore, this review analyzed the efficacy and safety of natural polyphenolic compounds for the treatment of COPD in clinical trials, and discussed their challenges and future development directions. In conclusion, this review combined the latest literature to illustrate the various pathogenesis and interrelationships of COPD in the form of graphs, texts, and tables, and sorted out the functional role and mechanisms of natural polyphenols in treating COPD, with a view to providing new ideas and plans for the in-depth research on COPD and the systemic treatment of COPD with Chinese herbal medicine.
Collapse
Affiliation(s)
- Yang Zhang
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Lijuan Wang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Jinyi Zeng
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Wen Shen
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China.
| |
Collapse
|
4
|
Rajeeve AD, Yamuna R, Namboori PKK. Elucidating the potential of EGFR mutated NSCLC and identifying its multitargeted inhibitors. Sci Rep 2025; 15:3649. [PMID: 39880831 PMCID: PMC11779874 DOI: 10.1038/s41598-024-83868-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related fatalities globally, accounting for the highest mortality rate among both men and women. Mutations in the epidermal growth factor receptor (EGFR) gene are frequently found in non-small cell lung cancer (NSCLC). Since curcumin and CB[2]UN support various medicinal applications in drug delivery and design, we investigated the effect of curcumin and CB[2]UN-based drugs in controlling EGFR-mutant NSCLC through a dodecagonal computational approach. Molecular docking studies revealed that the ligands curcumin (-6.9 kcal/mol) and CB[2]UN (-8.1 kcal/mol) bound more strongly to the EGFR-mutant NSCLC proteins with 2ITX and 2ITV, respectively. Molecular dynamics simulation (50 ns) investigation of protein-ligand complexes using RMSD, RMSF, Rg, and SASA indicated that curcumin and CB[2]UN with EGFR-mutant proteins are kinetically stable. In addition, MMPBSA/MMGBSA analysis confirmed the thermodynamic stability of each curcumin and CB[2]UN protein-ligand complex. Finally, KDeep absolute binding affinity calculations show energies of -6.13 kcal/mol and - 5.26 kcal/mol for 2ITX-CUR and 2ITV-CB[2]UN protein-ligand complexes, respectively. Thus, our dodecagonal strategy reveals that 2ITX-CUR and 2ITV-CB[2]UN are more likely to form protein-ligand complexes with more significant binding affinities and excellent stability throughout the 50 ns simulation time.
Collapse
Affiliation(s)
- Anakha D Rajeeve
- Department of Chemistry, Amrita School of Physical Sciences Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India
- Bio-materials Chemistry Research Laboratory, Amrita School of Engineering Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India
| | - Ramasamy Yamuna
- Department of Chemistry, Amrita School of Physical Sciences Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India.
- Bio-materials Chemistry Research Laboratory, Amrita School of Engineering Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India.
| | - P K Krishnan Namboori
- Amrita School of Artificial Intelligences, Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India.
- Computational Chemistry Group (CCG), Amrita School of Engineering, Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India.
- Center for Computational Engineering and Networking (CEN), Amrita School of Engineering, Coimbatore, Amrita Vishwa Vidyapeetham, Coimbatore, India.
- Biopharma Solutions, Coimbatore, 641105, India.
| |
Collapse
|
5
|
Anghel AC, Țăranu I, Orțan A, Marcu Spinu S, Dragoi Cudalbeanu M, Rosu PM, Băbeanu NE. Polyphenols and Microbiota Modulation: Insights from Swine and Other Animal Models for Human Therapeutic Strategies. Molecules 2024; 29:6026. [PMID: 39770115 PMCID: PMC11678809 DOI: 10.3390/molecules29246026] [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: 11/07/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
High consumption of ultra-processed foods, rich in sugar and unhealthy fats, has been linked to the onset of numerous chronic diseases. Consequently, there has been a growing shift towards a fiber-rich diet, abundant in fruits, vegetables, seeds, and nuts, to enhance longevity and quality of life. The primary bioactive components in these plant-based foods are polyphenols, which exert significant effects on modulating the gastrointestinal microbiota through their antioxidant and anti-inflammatory activities. This modulation has preventive effects on neurodegenerative, metabolic, and cardiovascular diseases, and even cancer. The antimicrobial properties of polyphenols against pathogenic bacteria have significantly reduced the need for antibiotics, thereby lowering the risk of antibiotic resistance. This paper advances the field by offering novel insights into the beneficial effects of polyphenols, both directly through the metabolites produced during digestion and indirectly through changes in the host's gastrointestinal microbiota, uniquely emphasizing swine as a model highly relevant to human health, a topic that, to our knowledge, has not been thoroughly explored in previous reviews. This review also addresses aspects related to both other animal models (mice, rabbits, and rats), and humans, providing guidelines for future research into the benefits of polyphenol consumption. By linking agricultural and biomedical perspectives, it proposes strategies for utilizing these bioactive compounds as therapeutic agents in both veterinary and human health sciences.
Collapse
Affiliation(s)
- Andrei Cristian Anghel
- Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 Marasti Boulevard, 011464 Bucharest, Romania; (A.C.A.); (N.E.B.)
- National Research-Development Institute for Animal Biology and Nutrition (IBNA), 1 Calea Bucuresti, 077015 Balotesti, Romania;
| | - Ionelia Țăranu
- National Research-Development Institute for Animal Biology and Nutrition (IBNA), 1 Calea Bucuresti, 077015 Balotesti, Romania;
| | - Alina Orțan
- Faculty of Land Reclamation and Environmental Engineering, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 MarastiBoulevard, 011464 Bucharest, Romania; (S.M.S.); (M.D.C.)
| | - Simona Marcu Spinu
- Faculty of Land Reclamation and Environmental Engineering, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 MarastiBoulevard, 011464 Bucharest, Romania; (S.M.S.); (M.D.C.)
| | - Mihaela Dragoi Cudalbeanu
- Faculty of Land Reclamation and Environmental Engineering, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 MarastiBoulevard, 011464 Bucharest, Romania; (S.M.S.); (M.D.C.)
| | - Petronela Mihaela Rosu
- Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 Marasti Boulevard, 011464 Bucharest, Romania;
| | - Narcisa Elena Băbeanu
- Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 59 Marasti Boulevard, 011464 Bucharest, Romania; (A.C.A.); (N.E.B.)
| |
Collapse
|
6
|
Ameer SF, Mohamed MY, Elzubair QA, Sharif EAM, Ibrahim WN. Curcumin as a novel therapeutic candidate for cancer: can this natural compound revolutionize cancer treatment? Front Oncol 2024; 14:1438040. [PMID: 39507759 PMCID: PMC11537944 DOI: 10.3389/fonc.2024.1438040] [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: 05/24/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer remains one of the leading causes of death worldwide. Despite advances in medical treatments, current therapeutic strategies, including radiotherapy, chemotherapy, targeted therapy, and surgical resection, have not significantly reduced the global incidence and mortality rates of cancer. Oncologists face considerable challenges in devising effective treatment plans due to the adverse side effects associated with standard therapies. Therefore, there is an urgent need for more effective and well-tolerated cancer treatments. Curcumin, a naturally occurring compound, has garnered significant attention for its diverse biological properties. Both preclinical studies and clinical trials have highlighted curcumin's potential in cancer treatment, demonstrating its ability to inhibit the proliferation of various cancer cell types through multiple cellular and molecular pathways. This paper examines the antineoplastic properties, and the therapeutic mechanisms including cell signalling pathways targeted by curcumin that are implicated in cancer development and explores the challenges in advancing curcumin as a viable anticancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Qin X, Wang P, Liang H, Si W. Curcumin suppresses copper accumulation in non-small cell lung cancer by binding ATOX1. BMC Pharmacol Toxicol 2024; 25:54. [PMID: 39169392 PMCID: PMC11340132 DOI: 10.1186/s40360-024-00784-0] [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: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is associated with intracellular copper accumulation. Antioxidant 1 (ATOX1) is a copper chaperone. This study aimed to analyze the anti-cancer effects of curcumin on the ATOX1-mediated copper pathway in NSCLC. METHODS A binding activity between curcumin and ATOX1 was measured using molecular docking. NSCLC cells, A549 and H1299, were treated with different doses of curcumin (10, 20, 40 µM) or DC-AC50 (5, 10, 20 µM) for 24 h. The cell viability and levels of ATOX1, ATP7A and COX17 proteins were observed in cells. Overexpressing ATOX1 in cells was established by pcDNA3.1-ATOX1 transfection for 24 h. The ATOX1 overexpressing cells were treated with 40 µM curcumin or 20 µM DC-AC50 for 24 h to analyze the mechanism of curcumin in NSCLC treatment. Cell viability was measured by CCK-8, and levels of proteins were measured by western blotting. The copper level in cells was labeled by copper sensor-1. Moreover, nude mice models were induced by injection of A549 cells and treated with 20 mg/kg/d DC-AC50 or 40 mg/kg/d curcumin. Tumor growth was observed by measuring tumor volume and tumor weight. The levels of ATOX1, ATP7A and COX17 in tumors were measured by immunohistochemistry and western blotting. RESULTS Curcumin bound to ATOX1 (score = -6.1 kcal/mol) and decreased the levels of ATOX1, ATP7A and COX17 proteins in NSCLC cells. The curcumin or DC-AC50 treatment suppressed cell viability by inhibiting the ATOX1-mediated copper signaling in NSCLC cells. The ATOX1 overexpression in cells significantly weakened the effects of curcumin on suppressing copper accumulation and the ATOX1-mediated copper pathway (p < 0.05). In mice models, curcumin or DC-AC50 treatment also suppressed tumor growth by suppressing the ATOX1-mediated copper pathway in tumors. CONCLUSION This study demonstrated that curcumin bound ATOX1 to suppress copper accumulation in NSCLC cells, providing a new mechanism of curcumin for NSCLC treatment.
Collapse
Affiliation(s)
- Xiao Qin
- Pulmonary and Critical Care Medicine, Yantai Shan Hospital, Yantai, 264025, China
| | - Peng Wang
- Ministry of Scientific and Technological Innovation, Yantai Hi-tech Industrial Development Zone, Yantai, 264025, China
| | - Haiyue Liang
- Drug Business Management Department, Yantai Center for Food and Drug Control, Yantai, 264000, China
| | - Wentao Si
- Oncology Department, Yantai Traditional Chinese Medicine Hospital, No. 39, Xingfu Road, Yantai, 264001, China.
| |
Collapse
|
8
|
Zhang J, Ma Y. Luteolin as a potential therapeutic candidate for lung cancer: Emerging preclinical evidence. Biomed Pharmacother 2024; 176:116909. [PMID: 38852513 DOI: 10.1016/j.biopha.2024.116909] [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: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Lung cancer is a prevalent malignant tumor and a leading cause of cancer-related fatalities globally. However, current treatments all have limitations. Therefore, there is an urgent need to identify a readily available therapeutic agent to counteract lung cancer development and progression. Luteolin is a flavonoid derived from vegetables and herbs that possesses preventive and therapeutic effects on various cancers. With the goal of providing new directions for the treatment of lung cancer, we review here the recent findings on luteolin so as to provide new ideas for the development of new anti-lung cancer drugs. The search focused on studies published between January 1995 and January 2024 that explored the use of luteolin in lung cancer. A comprehensive literature search was conducted in the SCOPUS, Google Scholar, PubMed, and Web of Science databases using the keywords "luteolin" and "lung cancer." By collecting previous literature, we found that luteolin has multiple mechanisms of therapeutic effects, including promotion of apoptosis in lung cancer cells; inhibition of tumor cell proliferation, invasion and metastasis; and modulation of immune responses. In addition, it can be used as an adjuvant to radio-chemotherapy and helps to ameliorate cancer complications. This review summarizes the structure, natural sources, physicochemical properties and pharmacokinetics of luteolin, and focuses on the anti-lung cancer mechanism of luteolin, so as to provide new ideas for the development of new anti-lung cancer drugs.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China
| | - Yue Ma
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|
9
|
Karati D, Mukherjee S, Roy S. Deciphering the molecular mechanistic paths describing the chemotherapeutic potential and epigenetic regulation of curcumin in lung cancer: a mini review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2715-2725. [PMID: 37982888 DOI: 10.1007/s00210-023-02838-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/03/2023] [Indexed: 11/21/2023]
Abstract
In an uncontrolled inflammatory environment, the complex process of lung carcinogenesis occurs. Lung cancer remains the leading cause of cancer-related mortality worldwide. The average 5-year survival rate is still low despite significant advancements in our knowledge of lung carcinogenesis and the development of innovative therapies in recent decades. Research on adjuvant treatment, lung carcinogenesis pathways, and possible prognostic indicators has to be refocused using an innovative approach. The majority of lung cancers are discovered at an advanced stage when there is little chance of recovery. It has grown in popularity in recent years to supplement already available chemotherapeutic therapies with adjuvant herbal medications, which may lessen toxicity and adverse effects without sacrificing therapeutic efficiency. One such prospective contender is curcumin. In-depth research has been done on curcumin as a multi-target anti-tumor and anti-inflammatory molecule. A pharmacologically active polyphenol produced from turmeric is called curcumin. Over the past few decades, curcumin's therapeutic potential has been thoroughly studied, and data indicate that curcumin may play a part in a variety of biological processes, most notably its potent anticancer activity. Being a pleiotropic chemical, curcumin regulates a variety of molecules that are key players in many cell signaling pathways. It has been shown to stifle transformation, restrain proliferation, and trigger apoptosis. Curcumin can reduce the development of non-small cell LC by downregulating Circular RNA hsa_circ_0007580, which in turn controls the expression of integrin subunit beta 1 by adsorbing miR-384. Nevertheless, despite all these advantages, curcumin's effectiveness is still restricted because of its weak bioavailability, poor absorption within the systemic circulation, and quick removal from the body. In an effort to overcome these constraints, scientists from all around the world are working to develop a synthetic and improved curcuminoid by appropriately altering the parent skeleton structurally. These curcuminoids will simultaneously improve the physicochemical properties and efficacy. This review presents evidence from the most recent clinical trials coupled with the molecular mechanisms of curcumin in LC.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
10
|
Salehi M, Negahdari B, Mehryab F, Shekari F. Milk-Derived Extracellular Vesicles: Biomedical Applications, Current Challenges, and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8304-8331. [PMID: 38587896 DOI: 10.1021/acs.jafc.3c07899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Extracellular vesicles (EVs) are nano to-micrometer-sized sacs that are released by almost all animal and plant cells and act as intercellular communicators by transferring their cargos between the source and target cells. As a safe and scalable alternative to conditioned medium-derived EVs, milk-derived EVs (miEVs) have recently gained a great deal of popularity. Numerous studies have shown that miEVs have intrinsic therapeutic actions that can treat diseases and enhance human health. Additionally, they can be used as natural drug carriers and novel classes of biomarkers. However, due to the complexity of the milk, the successful translation of miEVs from benchtop to bedside still faces several unfilled gaps, especially a lack of standardized protocols for the isolation of high-purity miEVs. In this work, by comprehensively reviewing the bovine miEVs studies, we provide an overview of current knowledge and research on miEVs while highlighting their challenges and enormous promise as a novel class of theranostics. It is hoped that this study will pave the way for clinical applications of miEVs by addressing their challenges and opportunities.
Collapse
Affiliation(s)
- Mahsa Salehi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
| | - Fatemeh Mehryab
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 14155-6153, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| |
Collapse
|
11
|
Zhang W, Xiao P, Liu B, Zhang Y. Circ-10720 as a ceRNA adsorbs microRNA-1238 and modulates ZEB2 to boost NSCLC development by activating EMT. Eur J Med Res 2024; 29:226. [PMID: 38610009 PMCID: PMC11010388 DOI: 10.1186/s40001-024-01715-9] [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: 03/03/2023] [Accepted: 02/06/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are critical regulators in the progression of tumors. This experimental design aimed to explore the mechanism of circ-10720 in non-small cell lung cancer (NSCLC). METHODS We used RT-qPCR to measure circ-10720 expression in clinical samples and analyzed its relationship with the clinicopathological characteristics of NSCLC patients. The expression levels of microRNA-1238 (miR-1238) and Zinc Finger E-box-binding Homeobox 2 (ZEB2) in clinical samples were detected by RT-qPCR. NSCLC cells were transfected with relevant plasmids or sequences. Circ-10720, miR-1238, and ZEB2 expressions in cells were analyzed via RT-qPCR or western blot. Cell proliferation, apoptosis, migration, and invasion were assessed with CCK-8, flow cytometry, and transwell assay, respectively. The protein expression of ZEB2 and epithelial-mesenchymal transition (EMT)-related markers (E-cadherin, Vimentin, N-cadherin) were detected via western blot. Xenograft assay was used to determine the effect of circ-10720 on NSCLC in vivo. Circ-10720 and ZEB2 expressions in tumors were detected using RT-qPCR or Western blot. Immunohistochemistry was used to evaluate E-cadherin and N-cadherin expression in tumors. Finally, the binding relationship between miR-1238 with circ-10720 or ZEB2 was verified by the bioinformatics website, dual luciferase reporter assay, RNA pull-down assay, and RIP assay. RESULTS Circ-10720 was upregulated in NSCLC and correlated with TNM stage of NSCLC patients. MiR-1238 was lowly expressed but ZEB2 was highly expressed in NSCLC. Circ-10720 silencing suppressed the proliferation, metastasis, and EMT of NSCLC cells. Mechanically, circ-10720 was a competitive endogenous RNA (ceRNA) for miR-1238, and ZEB2 was a target of miR-1238. circ-10720-modulated ZEB2 via competitively binding with miR-1238 to control NSCLC progression. In addition, circ-10720 knockdown suppressed tumor growth in vivo. CONCLUSIONS Circ-10720 acts as a ceRNA to adsorb miR-1238 and modulate ZEB2 to facilitate the proliferation, migration, invasion, and EMT of NSCLC cells.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, People's South Road, Section 4, Number 55, Chengdu, 610041, Sichuan, China
| | - Ping Xiao
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, Sichuan, China
| | - Bin Liu
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, People's South Road, Section 4, Number 55, Chengdu, 610041, Sichuan, China.
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, 37 Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
12
|
Zeb F, Naqeeb H, Osaili T, Faris ME, Ismail LC, Obaid RS, Naja F, Radwan H, Hasan H, Hashim M, AlBlooshi S, Alam I. Molecular crosstalk between polyphenols and gut microbiota in cancer prevention. Nutr Res 2024; 124:21-42. [PMID: 38364552 DOI: 10.1016/j.nutres.2024.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/18/2024]
Abstract
A growing body of evidence suggests that cancer remains a significant global health challenge, necessitating the development of novel therapeutic approaches. In recent years, the molecular crosstalk between polyphenols and gut microbiota has emerged as a promising pathway for cancer prevention. Polyphenols, abundant in many plant-based foods, possess diverse bioactive properties, including antioxidant, anti-inflammatory, and anticancer activities. The gut microbiota, a complex microbial community residing in the gastrointestinal tract, plays a crucial role in a host's health and disease risks. This review highlights cancer suppressive and oncogenic mechanisms of gut microbiota, the intricate interplay between gut microbiota modulation and polyphenol biotransformation, and the potential therapeutic implications of this interplay in cancer prevention. Furthermore, this review explores the molecular mechanisms underpinning the synergistic effects of polyphenols and the gut microbiota, such as modulation of signaling pathways and immune response and epigenetic modifications in animal and human studies. The current review also summarizes the challenges and future directions in this field, including the development of personalized approaches that consider interindividual variations in gut microbiota composition and function. Understanding the molecular crosstalk could offer new perspectives for the development of personalized cancer therapies targeting the polyphenol-gut axis. Future clinical trials are needed to validate the potential role of polyphenols and gut microbiota as innovative therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Falak Zeb
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates.
| | - Huma Naqeeb
- Department of Clinical Nutrition, Shaukat Khanam Cancer Hospital and Research Center Peshawar, Pakistan; Department of Human Nutrition and Dietetics, Women University Mardan, Pakistan
| | - Tareq Osaili
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates; Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, Irbid, Jordan
| | - MoezAllslam Ezzat Faris
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates
| | - Leila Cheikh Ismail
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates; Department of Women's and Reproductive Health, University of Oxford, Nuffield, Oxford, United Kingdom
| | - Reyad Shakir Obaid
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates
| | - Farah Naja
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates; Nutrition and Food Sciences Department, American University of Beirut, Beirut, Lebanon
| | - Hadia Radwan
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates
| | - Hayder Hasan
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates
| | - Mona Hashim
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates; Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates
| | - Sharifa AlBlooshi
- College of Natural and Health Sciences, Zayed University, United Arab Emirates
| | - Iftikhar Alam
- Department of Human Nutrition and Dietetics, Bacha Khan University Charsadda, Pakistan
| |
Collapse
|
13
|
Ngema L, Adeyemi SA, Marimuthu T, Ubanako PN, Ngwa W, Choonara YE. Short Antiangiogenic MMP-2 Peptide-Decorated Conjugated Linoleic Acid-Coated SPIONs for Targeted Paclitaxel Delivery in an A549 Cell Xenograft Mouse Tumor Model. ACS OMEGA 2024; 9:700-713. [PMID: 38222506 PMCID: PMC10785664 DOI: 10.1021/acsomega.3c06489] [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: 08/30/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 01/16/2024]
Abstract
The design of targeted antiangiogenic nanovectors for the delivery of anticancer drugs presents a viable approach for effective management of nonsmall-cell lung carcinoma (NSCLC). Herein, we report on the fabrication of a targeted delivery nanosystem for paclitaxel (PTX) functionalized with a short antimatrix metalloproteinase 2 (MMP-2) CTT peptide for selective MMP-2 targeting and effective antitumor activity in NSCLC. The fabrication of the targeted nanosystem (CLA-coated PTX-SPIONs@CTT) involved coating of superparamagnetic iron-oxide nanoparticles (SPIONs) with conjugated linoleic acid (CLA) via chemisorption, onto which PTX was adsorbed, and subsequent surface functionalization with carboxylic acid groups for conjugation of the CTT peptide. CLA-coated PTX SPIONs@CTT had a mean particle size of 99.4 nm and a PTX loading efficiency of ∼98.5%. The nanosystem exhibited a site-specific in vitro PTX release and a marked antiproliferative action on lung adenocarcinoma cells. The CTT-functionalized nanosystem significantly inhibited MMP-2 secretion by almost 70% from endothelial cells, indicating specific anti-MMP-2 activity. Treatment of tumor-bearing mice with subcutaneous injection of the CTT-functionalized nanosystem resulted in 69.7% tumor inhibition rate, and the administration of the nanosystem subcutaneously prolonged the half-life of PTX and circulation time in vivo. As such, CLA-coated PTX-SPIONs@CTT presents with potential for application as a targeted nanomedicine in NSCLC management.
Collapse
Affiliation(s)
- Lindokuhle
M. Ngema
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Samson A. Adeyemi
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Thashree Marimuthu
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Philemon N. Ubanako
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Wilfred Ngwa
- Sidney
Kimmel Comprehensive Cancer Center, Johns
Hopkins Medicine, Baltimore, Maryland 21218, United States
| | - Yahya E. Choonara
- Wits
Advanced Drug Delivery Platform Research Unit, Department of Pharmacy
and Pharmacology, School of Therapeutic Sciences, Faculty of Health
Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
14
|
Gad HA, Diab AM, Elsaied BE, Tayel AA. Biopolymer-based formulations for curcumin delivery toward cancer management. CURCUMIN-BASED NANOMEDICINES AS CANCER THERAPEUTICS 2024:309-338. [DOI: 10.1016/b978-0-443-15412-6.00009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
15
|
Gupta P, Neupane YR, Aqil M, Kohli K, Sultana Y. Lipid-based nanoparticle-mediated combination therapy for breast cancer management: a comprehensive review. Drug Deliv Transl Res 2023; 13:2739-2766. [PMID: 37261602 DOI: 10.1007/s13346-023-01366-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
Breast cancer due to the unpredictable and complex etiopathology combined with the non-availability of any effective drug treatment has become the major root of concern for oncologists globally. The number of women affected by the said disease state is increasing at an alarming rate attributed to environmental and lifestyle changes indicating at the exploration of a novel treatment strategy that can eradicate this aggressive disease. So far, it is treated by promising nanomedicine monotherapy; however, according to the numerous studies conducted, the inadequacy of these nano monotherapies in terms of elevated toxicity and resistance has been reported. This review, therefore, puts forth a new multimodal strategic approach to lipid-based nanoparticle-mediated combination drug delivery in breast cancer, emphasizing the recent advancements. A basic overview about the combination therapy and its index is firstly given. Then, the various nano-based combinations of chemotherapeutics involving the combination delivery of synthetic and herbal agents are discussed along with their examples. Further, the recent exploration of chemotherapeutics co-delivery with small interfering RNA (siRNA) agents has also been explained herein. Finally, a section providing a brief description of the delivery of chemotherapeutic agents with monoclonal antibodies (mAbs) has been presented. From this review, we aim to provide the researchers with deep insight into the novel and much more effective combinational lipid-based nanoparticle-mediated nanomedicines tailored specifically for breast cancer treatment resulting in synergism, enhanced antitumor efficacy, and low toxic effects, subsequently overcoming the hurdles associated with conventional chemotherapy.
Collapse
Affiliation(s)
- Priya Gupta
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yub Raj Neupane
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA, 52242, USA
| | - Mohd Aqil
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
- Lloyd Institute of Management & Technology (Pharm.), Plot No. 11, Knowledge Park-II, Greater Noida, Uttar Pradesh, 201308, India.
| | - Yasmin Sultana
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
16
|
Moutabian H, Radi UK, Saleman AY, Adil M, Zabibah RS, Chaitanya MNL, Saadh MJ, Jawad MJ, Hazrati E, Bagheri H, Pal RS, Akhavan-Sigari R. MicroRNA-155 and cancer metastasis: Regulation of invasion, migration, and epithelial-to-mesenchymal transition. Pathol Res Pract 2023; 250:154789. [PMID: 37741138 DOI: 10.1016/j.prp.2023.154789] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/25/2023]
Abstract
Among the leading causes of death globally has been cancer. Nearly 90% of all cancer-related fatalities are attributed to metastasis, which is the growing of additional malignant growths out of the original cancer origin. Therefore, a significant clinical need for a deeper comprehension of metastasis exists. Beginning investigations are being made on the function of microRNAs (miRNAs) in the metastatic process. Tiny non-coding RNAs called miRNAs have a crucial part in controlling the spread of cancer. Some miRNAs regulate migration, invasion, colonization, cancer stem cells' properties, the epithelial-mesenchymal transition (EMT), and the microenvironment, among other processes, to either promote or prevent metastasis. One of the most well-conserved and versatile miRNAs, miR-155 is primarily distinguished by overexpression in a variety of illnesses, including malignant tumors. It has been discovered that altered miR-155 expression is connected to a number of physiological and pathological processes, including metastasis. As a result, miR-155-mediated signaling pathways were identified as possible cancer molecular therapy targets. The current research on miR-155, which is important in controlling cancer cells' invasion, and metastasis as well as migration, will be summarized in the current work. The crucial significance of the lncRNA/circRNA-miR-155-mRNA network as a crucial regulator of carcinogenesis and a player in the regulation of signaling pathways or related genes implicated in cancer metastasis will be covered in the final section. These might provide light on the creation of fresh treatment plans for controlling cancer metastasis.
Collapse
Affiliation(s)
- Hossein Moutabian
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Mv N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | | | - Ebrahi Hazrati
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rashmi Saxena Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
17
|
Wang Y, Wei M, Su M, Du Z, Dong J, Zhang Y, Wu Y, Li X, Su L, Liu X. DIRAS3 enhances RNF19B-mediated RAC1 ubiquitination and degradation in non-small-cell lung cancer cells. iScience 2023; 26:107157. [PMID: 37485351 PMCID: PMC10362343 DOI: 10.1016/j.isci.2023.107157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Distant metastasis remains the leading cause of high mortality in patients with non-small-cell lung cancer (NSCLC). DIRAS3 is a candidate tumor suppressor protein that is decreased in various tumors. However, the regulatory mechanism of DIRAS3 on metastasis of NSCLC remains unclear. Here, we found that DIRAS3 suppressed the migration of NSCLC cells. Besides, DIRAS3 stimulated the polyubiquitination of RAC1 and suppressed its protein expression. Furthermore, RNF19B, a member of the RBR E3 ubiquitin ligase family, was observed to be the E3 ligase involved in the DIRAS3-induced polyubiquitination of RAC1. DIRAS3 could promote the binding of RAC1 and RNF19B, thus enhancing the degradation of RAC1 by the ubiquitin-proteasome pathway. Finally, the DIRAS3-RNF19B-RAC1 axis was confirmed to be associated with the malignant progression of NSCLC. These findings may be beneficial for developing potential prognostic markers of NSCLC and may provide an effective treatment strategy.
Collapse
Affiliation(s)
- Yingying Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Minli Wei
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Min Su
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhiyuan Du
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Jiaxi Dong
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yu Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yingdi Wu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiaopeng Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ling Su
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiangguo Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| |
Collapse
|
18
|
Ngema LM, Adeyemi SA, Marimuthu T, Ubanako PN, Ngwa W, Choonara YE. Surface Immobilization of Anti-VEGF Peptide on SPIONs for Antiangiogenic and Targeted Delivery of Paclitaxel in Non-Small-Cell Lung Carcinoma. ACS APPLIED BIO MATERIALS 2023. [PMID: 37384895 PMCID: PMC10354746 DOI: 10.1021/acsabm.3c00224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
A design has been established for the surface decoration of superparamagnetic iron oxide nanoparticles (SPIONs) with anti-vascular endothelial growth factor peptide, HRH, to formulate a targeted paclitaxel (PTX) delivery nanosystem with notable tumor targetability and antiangiogenic activity. The design methodology included (i) tandem surface functionalization via coupling reactions, (ii) pertinent physicochemical characterization, (iii) in vitro assessment of drug release, anti-proliferative activity, and quantification of vascular endothelial growth factor A (VEGF-A) levels, and (iv) in vivo testing using a lung tumor xenograft mouse model. Formulated CLA-coated PTX-SPIONs@HRH depicted a size and surface charge of 108.5 ± 3.5 nm and -30.4 ± 2.3 mV, respectively, and a quasi-spherical shape relative to pristine SPIONs. Fourier transform infrared (FTIR) analysis and estimation of free carboxylic groups supported the preparation of the CLA-coated PTX-SPIONs@HRH. CLA-coated PTX-SPIONs@HRH exhibited high PTX loading efficiency (98.5%) and sustained release in vitro, with a marked dose dependent anti-proliferative activity in A549 lung adenocarcinoma cells, complimented by an enhanced cellular uptake. CLA-coated PTX-SPIONs@HRH significantly reduced secretion levels of VEGF-A in human dermal microvascular endothelial cells from 46.9 to 35.6 pg/mL compared to untreated control. A 76.6% tumor regression was recorded in a lung tumor xenograft mouse model following intervention with CLA-coated PTX-SPIONs@HRH, demonstrating tumor targetability and angiogenesis inhibition. CLA-coated PTX-SPIONs@HRH enhanced the half-life of PTX by almost 2-folds and demonstrated a prolonged PTX plasma circulation time from a subcutaneous injection (SC). Thus, it is suggested that CLA-coated PTX-SPIONs@HRH could provide a potential effective treatment modality for non-small-cell lung carcinoma as a nanomedicine.
Collapse
Affiliation(s)
- Lindokuhle M Ngema
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Samson A Adeyemi
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Thashree Marimuthu
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Philemon N Ubanako
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Wilfred Ngwa
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
19
|
Niu H, Wang D, Wen T, Liu H, Jie J, Song L, Li D. Anwuligan inhibits the progression of non-small cell lung cancer via let-7c-3p/PI3K/AKT/mTOR axis. Cancer Med 2023; 12:5908-5925. [PMID: 36412203 PMCID: PMC10028152 DOI: 10.1002/cam4.5382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/25/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Anwuligan (ANW) isolated from nutmeg, also known as myristyl lignan, has attracted attention due to its therapeutic potential in diseases. However, its effect on lung cancer is still unclear. METHODS In this study, the cytotoxicity of ANW on non-small cell lung cancer (NSCLC) cells was detected using a Cell Counting Kit-8 (CCK-8) assay. In vitro, clone formation, wound healing, and Transwell assays were used to investigate the migratory and invasive abilities of NSCLC cells after ANW treatment. The expression levels of the associated proteins were detected using Western blotting. A luciferase assay was used to validate the binding of let-7c-3p to the 3'-untranslated region (UTR) of PIK3CA. In vivo, an A549 cell xenograft mouse model was used to verify the effect of ANW on tumor growth. RESULTS The results showed that ANW treatment (20 or 50 μM) had obvious cytotoxicity against A549 and H460 cells, suppressing cell proliferation and migration in vitro. In vivo, the growth of the implanted tumor was inhibited by ANW in a nude mouse model. The growth of NSCLC cells was also inhibited by let-7c-3p overexpression in vitro and in vivo. The inhibitory effect of ANW on the proliferation and metastasis of NSCLC cells was weakened by the downregulation of let-7c-3p, whereas it was enhanced by the overexpression of let-7c-3p; PIK3CA was the main target of let-7c-3p. CONCLUSIONS In summary, ANW inhibits the growth and metastasis of NSCLC cells in vivo and in vitro by upregulating the expression of let-7c-3p, which can regulate the PI3K/AKT/mTOR signaling pathway. PIK3CA is the main target of let-7c-3p.
Collapse
Affiliation(s)
- Huikun Niu
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Dexiang Wang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingting Wen
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Han Liu
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Jing Jie
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Lei Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Najafi M, Tavakol S, Zarrabi A, Ashrafizadeh M. Dual role of quercetin in enhancing the efficacy of cisplatin in chemotherapy and protection against its side effects: a review. Arch Physiol Biochem 2022; 128:1438-1452. [PMID: 32521182 DOI: 10.1080/13813455.2020.1773864] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemotherapy has opened a new window in cancer therapy. However, the resistance of cancer cells has dramatically reduced the efficacy of chemotherapy. Cisplatin is a chemotherapeutic agent and its potential in cancer therapy has been restricted by resistance of cancer cells. As a consequence, the scientists have attempted to find new strategies in elevating chemotherapy efficacy. Due to great anti-tumour activity, naturally occurring compounds are of interest in polychemotherapy. Quercetin is a flavonoid with high anti-tumour activity against different cancers that can be used with cisplatin to enhance its efficacy and also are seen to sensitise cancer cells into chemotherapy. Furthermore, cisplatin has side effects such as nephrotoxicity and ototoxicity. Administration of quercetin is advantageous in reducing the adverse effects of cisplatin without compromising its anti-tumour activity. In this review, we investigate the dual role of quercetin in enhancing anti-tumour activity of cisplatin and simultaneous reduction in its adverse effects.
Collapse
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Turkey
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
21
|
Irshad R, Raj N, Gabr GA, Manzoor N, Husain M. Integrated network pharmacology and experimental analysis unveil multi-targeted effect of 18α- glycyrrhetinic acid against non-small cell lung cancer. Front Pharmacol 2022; 13:1018974. [PMID: 36313358 PMCID: PMC9596789 DOI: 10.3389/fphar.2022.1018974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/28/2022] [Indexed: 11/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most malignant types of cancer with soaring incidence rates worldwide, attributed to its heterogeneity and complex etiology. Evidently, alternative anti-cancer therapies comprising traditional medicines and natural products have gained attention for their ability to act as chemopreventive agents with minimal toxicities, either alone or in combination. Accumulating studies have substantiated the inevitability of network pharmacology studies for effectively mapping molecular targets of natural products against multifaceted diseases, including cancer. The 18α-Glycyrrhetinic acid (18α-GA), a triterpenoid found in licorice plants, has shown promising medicinal properties, although, its mechanism of action against NSCLC yet remains elusive. The present study was conducted to explore the anti- NSCLC potential of 18α-GA, employing integrative network pharmacology, molecular docking, and experimental research. Initially, network analysis revealed 181 common targets of 18α-GA in NSCLC as shown in the “compound-target- disease” network employing Cytoscape 3.8.2. Further analyses identified EGFR, AKT1, PI3KR1, MAPK1, IGF1, and SRC as the most crucial hub targets of 18α-GA against NSCLC. Moreover, molecular docking simulations and functional enrichment analyses indicated the involvement of multiple signaling pathways in suppressing NSCLC. Subsequent in-vitro studies verified the antiproliferative effect of 18α-GA on two NSCLC cancer cell lines, H1299 and A549. Mechanistically, 18α-GA arrested cell cycle at the G1 phase, induced apoptosis, decreased migratory potential, and protein expression levels of EGFR-PI3K/AKT, as examined by flow cytometry, morphological assessment, RT-PCR, and western blot. In conclusion, this study delineates the therapeutic potential and underlying mechanism(s) of 18α-GA as a putative novel drug against NSCLC. However, further studies are warranted to elucidate the complete molecular mechanism(s) using animal models of NSCLC.
Collapse
Affiliation(s)
- Rasha Irshad
- Virology and Oncology Lab, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Nafis Raj
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Gamal A. Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al- Kharj, Saudi Arabia
| | - Nikhat Manzoor
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Husain
- Virology and Oncology Lab, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
- *Correspondence: Mohammad Husain,
| |
Collapse
|
22
|
Gholinia Sarpoli LM, Zare-Karizi S, Heidari E, Hasanzadeh A, Bayandori M, Azedi F, Hamblin MR, Karimi M. Co-delivery of curcumin and Bcl-2 siRNA to enhance therapeutic effect against breast cancer cells using PEI-functionalized PLGA nanoparticles. Pharm Dev Technol 2022; 27:785-793. [PMID: 36043390 DOI: 10.1080/10837450.2022.2120003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
PURPOSE Breast cancer is the second major cause of death worldwide among women. Co-delivery of anticancer drugs and nucleic acids targeting the apoptosis pathway could be a promising new approach. METHODS In the present study, we synthesized a novel nanostructure for co-delivery of curcumin and siRNA to breast cancer cells. Curcumin-loaded polylactic-co-glycolic acid (PLGA) was synthesized using an O/W emulsion-solvent diffusion method. It was coated with polyethylenimine (PEI) and subsequently complexed with Bcl-2 siRNA. Also nanoparticles were characterized such as zeta potential, size distribution and drug encapsulation. Finally the cytotoxicity of NP and Bcl2 experession was evaluated. RESULTS The curcumin loaded PLGA nanoparticles were 70nm in size, and increased to 84 nm after incorporation of PEI plus Bcl-2 siRNA. The encapsulation ratio of drug in our nanoparticle was 78%. Cellular internalization PLGA-CUR-PEI/Bcl-2 siRNA NPs was confirmed by fluorescence microscopy with broadcasting of the fluorescence in the cytoplasm and into the nucleus. The results of the cell viability assay revealed that curcumin-loaded PLGA coated with PEI and Bcl-2 siRNA exhibited the highest cytotoxicity against the T47D cell line, while the siRNA decreased the Bcl-2 expression by 90.7%. CONCLUSION The co-delivery of curcumin plus Bcl-2 siRNA with the PLGA-PEI nanosystem could be a synergistic drug carrier against breast cancer cells.
Collapse
Affiliation(s)
| | - Shohreh Zare-Karizi
- Department of Genetics, School of Biological Sciences, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Erfan Heidari
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akbar Hasanzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Bayandori
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Azedi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.,Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Karimi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Advances Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Gitogenin suppresses lung cancer progression by inducing apoptosis and autophagy initiation through the activation of AMPK signaling. Int Immunopharmacol 2022; 111:108806. [PMID: 35914447 DOI: 10.1016/j.intimp.2022.108806] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/18/2022] [Accepted: 04/24/2022] [Indexed: 11/22/2022]
Abstract
Lung cancer is a leading cause of tumor-associated death worldwide. Autophagy plays a key role in regulating lung cancer progression, and is a promising option for lung cancer treatment. Saponins are a group of naturally occurring plant glycosides, characterized by their strong foam-forming properties in aqueous solution, and exert various biological properties, such as anti-inflammation and anti-cancer. In the present study, we for the first time explored the effects of gitogenin (GIT), an important saponin derived from Tribulus longipetalus, on lung cancer progression both in vitro and in vivo. We found that GIT markedly reduced the proliferation and induced apoptosis in lung cancer cells through increasing the cleavage of Caspase-3 and poly (ADP-ribose) polymerases (PARPs). In addition, GIT-incubated lung cancer cells exhibited clear accumulation of autophagosome, which was essential for GIT-suppressed lung cancer. Mechanistically, GIT-induced autophagy initiation was mainly through activating AMP-activated protein kinase (AMPK) and blocking protein kinase B (AKT) signaling pathways, respectively. Moreover, the autophagic flux was disrupted in GIT-treated lung cancer cells, contributing to the accumulation of impaired autophagolysosomes. Importantly, we found that suppressing autophagy initiation could abolish GIT-induced cell death; however, autophagosomes accumulation sensitized lung cancer cells to cell death upon GIT treatment. More in vitro experiments showed that GIT led to reactive oxygen species (ROS) production in lung cancer cells, which was also involved in the modulation of apoptosis. The in vivo findings confirmed the effects of GIT against lung cancer progression with undetectable toxicity to organs. In conclusion, we provided new insights into the treatment of lung cancer, and GIT might be an effective strategy for future clinical application.
Collapse
|
24
|
Ngema LM, Adeyemi SA, Marimuthu T, Ubanako P, Wamwangi D, Choonara YE. Synthesis of Novel Conjugated Linoleic Acid (CLA)-Coated Superparamagnetic Iron Oxide Nanoparticles (SPIONs) for the Delivery of Paclitaxel with Enhanced In Vitro Anti-Proliferative Activity on A549 Lung Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14040829. [PMID: 35456663 PMCID: PMC9031641 DOI: 10.3390/pharmaceutics14040829] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 01/06/2023] Open
Abstract
The application of Superparamagnetic Iron Oxide Nanoparticles (SPIONs) as a nanomedicine for Non-Small Cell Lung Carcinoma (NSCLC) can provide effective delivery of anticancer drugs with minimal side-effects. SPIONs have the flexibility to be modified to achieve enhanced oading of hydrophobic anticancer drugs such as paclitaxel (PTX). The purpose of this study was to synthesize novel trans-10, cis-12 conjugated linoleic acid (CLA)-coated SPIONs loaded with PTX to enhance the anti-proliferative activity of PTX. CLA-coated PTX-SPIONs with a particle size and zeta potential of 96.5 ± 0.6 nm and −27.3 ± 1.9 mV, respectively, were synthesized. The superparamagnetism of the CLA-coated PTX-SPIONs was confirmed, with saturation magnetization of 60 emu/g and 29 Oe coercivity. CLA-coated PTX-SPIONs had a drug loading efficiency of 98.5% and demonstrated sustained site-specific in vitro release of PTX over 24 h (i.e., 94% at pH 6.8 mimicking the tumor microenvironment). Enhanced anti-proliferative activity was also observed with the CLA-coated PTX-SPIONs against a lung adenocarcinoma (A549) cell line after 72 h, with a recorded cell viability of 17.1%. The CLA-coated PTX-SPIONs demonstrated enhanced suppression of A549 cell proliferation compared to pristine PTX, thus suggesting potential application of the nanomedicine as an effective site-specific delivery system for enhanced therapeutic activity in NSCLC therapy.
Collapse
Affiliation(s)
- Lindokuhle M. Ngema
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (L.M.N.); (S.A.A.); (T.M.); (P.U.)
| | - Samson A. Adeyemi
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (L.M.N.); (S.A.A.); (T.M.); (P.U.)
| | - Thashree Marimuthu
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (L.M.N.); (S.A.A.); (T.M.); (P.U.)
| | - Philemon Ubanako
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (L.M.N.); (S.A.A.); (T.M.); (P.U.)
| | - Daniel Wamwangi
- School of Physics, Materials Physics Research Institute, University of the Witwatersrand, Private Bag 3, WITS, Johannesburg 2050, South Africa;
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (L.M.N.); (S.A.A.); (T.M.); (P.U.)
- Correspondence: ; Tel.: +27-11-717-2052
| |
Collapse
|
25
|
Study on the Mechanism of Action of Paclitaxel-Loaded Polylactic-co-glycolic Acid Nanoparticles in Non-Small-Cell Lung Carcinoma Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8524951. [PMID: 35432585 PMCID: PMC9007685 DOI: 10.1155/2022/8524951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 02/05/2022] [Indexed: 11/17/2022]
Abstract
Objective. To study effective carriers that can enhance the antitumor effect of paclitaxel (PTX). Methods. PTX-loaded polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) (PTX-PLGA NPs), constructed using the emulsification solvent evaporation method, were characterized by scanning electron microscopy and dynamic light scattering. Non-small-cell lung carcinoma (NSCLC) cells were divided into the dimethyl sulfoxide (DMSO) group, PLGA NPs group, PTX group, and PTX-PLGA NPs group. Cell viability was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), cell apoptosis was determined by flow cytometry, and cell migration and invasion were assessed using Transwell assay. Results. PTX-PLGA NPs were smooth in the surface and spherical in shape, with a particle size of
nm. Both PTX and PTX-PLGA NPs could effectively inhibit the activity of A549 and H1650 cells. At 12 and 24 h, PTX-PLGA NPs presented weaker inhibition on the activity of NSCLC cells than PTX, but at 48 and 72 h, PTX-PLGA NPs presented stronger inhibition. Compared with PTX, PTX-PLGA NPs were more effective in enhancing apoptosis and inhibiting migration and invasion of NSCLC cells. Conclusion. With good sustained release and the ability to promote cellular uptake, PTX-PLGA NPs can strongly inhibit the malignant activities of NSCLC cells, which can be used as a promising drug carrier.
Collapse
|
26
|
Circ_0078767 Inhibits the Progression of Non-Small-Cell Lung Cancer by Regulating the GPX3 Expression by Adsorbing miR-665. Int J Genomics 2022; 2022:6361256. [PMID: 35340708 PMCID: PMC8948607 DOI: 10.1155/2022/6361256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most serious cancers. The circular RNA_0078767 (circ_0078767) expression was decreased in NSCLC tissues. However, the molecular mechanism of circ_0078767 remains unknown. The expression of circ_0078767, microRNA-665 (miR-665), and glutathione peroxidase 3 (GPX3) was detected by quantitative real-time fluorescence polymerase chain reaction (qRT-PCR). Cell proliferation, migration, and invasion were detected by colony formation assay and transwell assay, respectively. The lactate production and glucose consumption were tested by glycolysis. Western blot examined the protein levels of hexokinase-2 (HK2), matrix metalloproteinase-9 (MMP9), and GPX3 cells. Circinteractome predicted the relationship between miR-665 and circ_0078767 or GPX3 and was verified by dual luciferase reporter assays. The xenotransplantation model was established to study the role of circ_0078767 in vivo. The expression of circ_0078767 and GPX3 was decreased in NSCLC tissues, while the expression of miR-665 was increased. Circ_0078767 can sponge miR-665, and GPX3 is the target of miR-665. In vitro complement experiments showed that knockdown of circ_0078767 significantly promoted malignant behavior of NSCLC, while cotransfection of miR-665 inhibitor partially reduced this change. In addition, the GPX3 overexpression decreased the promoting effects of miR-665 upregulation on proliferation, migration, and invasion of NSCLC cells. Mechanically, circ_0078767 regulates the GPX3 expression in NSCLC cells by spongy miR-665. In addition, in vivo studies have shown that downregulation of circ_0078767 promotes tumor growth. Circ_0078767 silencing promotes proliferation, migration, invasion, and glycolysis of NSCLC cells by regulating the miR-665/GPX3 axis, suggesting that circ_0078767/miR-665/GPX3 axis may be a potential regulatory mechanism for the treatment of NSCLC.
Collapse
|
27
|
Kuang P, Xie A, Deng J, Tang J, Wang P, Yu F. GTP-binding protein Di-RAS3 diminishes the migration and invasion of non-small cell lung cancer by inhibiting the RAS/extracellular-regulated kinase pathway. Bioengineered 2022; 13:5663-5674. [PMID: 35170376 PMCID: PMC8973588 DOI: 10.1080/21655979.2022.2031671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The GTP-binding protein Di-Ras3 (DIRAS3) has been established as a maternally imprinted tumor suppressor gene. Growing evidence has correlated the DIRAS3 gene with tumor progression, but its role in non-small cell lung cancer (NSCLC) is rarely reported. Accordingly, the current study sought to evaluate the role and mechanism of DIRAS3 in NSCLC cell progression. First, we uncovered that DIRAS3 was poorly expressed in NSCLC tissues and cells. Subsequently, we examined the effect of DIRAS3 over-expression or knockdown in different lung cancer cells on their malignant phenotypes, with the help of transwell cell migration and invasion assays, and Western blot analyses. It was found that the over-expression of DIRAS3 inhibited the migration and invasion of A549 cells or H520 cells, whereas knockdown of DIRAS3 led to opposing trends. In addition, over-expression of DIRAS3 attenuated the tumor growth and reduced the number of lung tumor nodules. Mechanistically, DIRAS3 may inhibit the migration and invasion of NSCLC cells by inhibiting the RAS/extracellular-regulated kinase (ERK) signaling pathway. Collectively, our findings indicate that DIRAS3 could serve as a potential therapeutic target biomarker for NSCLC.
Collapse
Affiliation(s)
- Peng Kuang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - An Xie
- Jiangxi Institute of Urology, The First Affiliated Hospital of Nanchang University, China
| | - Jianxiong Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiaming Tang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peijun Wang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Yu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
28
|
Maleki Dizaj S, Alipour M, Dalir Abdolahinia E, Ahmadian E, Eftekhari A, Forouhandeh H, Rahbar Saadat Y, Sharifi S, Zununi Vahed S. Curcumin nanoformulations: Beneficial nanomedicine against cancer. Phytother Res 2022; 36:1156-1181. [PMID: 35129230 DOI: 10.1002/ptr.7389] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022]
Abstract
Curcumin is a phytochemical achieved from the plant turmeric. It is extensively utilized for the treatment of several types of diseases such as cancers. Nevertheless, its efficiency has been limited because of rapid metabolism, low bioavailability, poor water solubility, and systemic elimination. Scientists have tried to solve these problems by exploring novel drug delivery systems such as lipid-based nanoparticles (NPs) (e.g., solid lipid NPs, nanostructured lipid carriers, and liposomes), polymeric NPs, micelles, nanogels, cyclodextrin, gold, and mesoporous silica NPs. Among these, liposomes have been the most expansively studied. This review mainly focuses on the different curcumin nanoformulations and their use in cancer therapy in vitro, in vivo, and clinical studies. Despite the development of curcumin-containing NPs for the treatment of cancer, potentially serious side effects, including interactions with other drugs, some toxicity aspects of NPs may occur that require more high-quality investigations to firmly establish the clinical efficacy.
Collapse
Affiliation(s)
- Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Dental Biomaterials, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Health innovation and acceleration center, Tabriz University of Medical Sciences, Tabriz, Iran.,Russian Institute for Advanced Study, Moscow State Pedagogical University, Moscow, Russian Federation
| | - Haleh Forouhandeh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
29
|
Wan L, Cheng Z, Sun Q, Jiang K. LncRNA HOXC-AS3 increases non-small cell lung cancer cell migration and invasion by sponging premature miR-96. Expert Rev Respir Med 2022; 16:587-593. [PMID: 35034519 DOI: 10.1080/17476348.2022.2030223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND HOXC cluster antisense RNA 3 (HOXC-AS3) has been characterized as a critical long noncoding RNA (lncRNA) player in breast cancer and gastric cancer, while its role in non-small cell lung cancer (NSCLC) is not clear. We hypothesized that HOXC-AS3 could interact with premature microRNA (miR)-96. This study was therefore carried out to explore the crosstalk between HOXC-AS3 and miR-96 in NSCLC. METHODS The expression of HOXC-AS3 and miR-96 (both mature and premature) were detected using RT-qPCR. Nuclear fractionation assay and RNA pull-down assay were performed to detect the subcellular location of HOXC-AS3 and potential interaction with premature miR-96, respectively. Overexpression assays were performed to determine the role of HOXC-AS3 in the maturation of miR-96. Transwell assays were performed to explore the role of HOXC-AS3 and miR-96 in NSCLC cell invasion and migration. RESULTS NSCLC tissues exhibited significantly increased expression levels of HOXC-AS3 and premature miR-96. HOXC-AS3 was localized to both nucleus and cytoplasm, and a direct interaction between HOXC-AS3 and premature miR-96 was observed. In NSCLC cells, HOXC-AS3 upregulated the expression of premature miR-96 but downregulated the expression of mature miR-96. Moreover, HOXC-AS3 suppressed the role of miR-96 in inhibiting NSCLC cell invasion and migration. CONCLUSION HOXC-AS3 may increase NSCLC cell growth and invasion by sponging premature miR-96 to suppress its maturation.
Collapse
Affiliation(s)
- Li Wan
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, 43000, People's Republic of China
| | - Zaixing Cheng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, 43000, People's Republic of China
| | - Quanchao Sun
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, 43000, People's Republic of China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, 43000, People's Republic of China
| |
Collapse
|
30
|
Zhao X, He X, Wang M, Zhang H, Cheng J, Wang H. LncRNA PLAC2 upregulates CDK6 by directly targeting miR-29C to promote cell proliferation in lung squamous cell carcinoma. Crit Rev Eukaryot Gene Expr 2022; 32:55-67. [DOI: 10.1615/critreveukaryotgeneexpr.2022044134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Jiang S, Fu Y, Zhang X, Yu T, Lu B, Du J. Research Progress of Carrier-Free Antitumor Nanoparticles Based on Phytochemicals. Front Bioeng Biotechnol 2021; 9:799806. [PMID: 34957085 PMCID: PMC8692885 DOI: 10.3389/fbioe.2021.799806] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/29/2021] [Indexed: 11/21/2022] Open
Abstract
Cancer is a major worldwide public health issue, responsible for millions of deaths every year. Cancer cases and deaths are expected to increase rapidly with population growth, age, and lifestyle behaviors that increase cancer risk. Long-term chemotherapy results in acquired drug resistance. Traditional treatment methods have limitations and cannot effectively treat distal metastatic cancers. Application of nanocarriers in multi-chemotherapy must be promoted. With research progress, the shortcomings of traditional nanocarriers have gradually become evident. Carrier-free nanodrugs with desirable bioactivity have attracted considerable attention. In this review, we provide an overview of recent reports on several carrier-free nanodrug delivery systems based on phytochemicals. This review focuses on the advantages of carrier-free nanodrugs, and provides new insights for establishment of ideal cancer treatment nanosystems.
Collapse
Affiliation(s)
- Siliang Jiang
- Department of Pharmacognosy, College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Yu Fu
- Department of Pharmacognosy, College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Xinyang Zhang
- School of Clinical Medicine, Jiamusi University, Jiamusi, China
| | - Tong Yu
- Department of Pharmacognosy, College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Bowen Lu
- Department of Pharmacognosy, College of Pharmacy, Jiamusi University, Jiamusi, China
| | - Juan Du
- Department of Pharmacognosy, College of Pharmacy, Jiamusi University, Jiamusi, China
| |
Collapse
|
32
|
Abd El-Hameed NM, Abd El-Aleem SA, Khattab MA, Ali AH, Mohammed HH. Curcumin activation of nuclear factor E2-related factor 2 gene (Nrf2): Prophylactic and therapeutic effect in nonalcoholic steatohepatitis (NASH). Life Sci 2021; 285:119983. [PMID: 34599938 DOI: 10.1016/j.lfs.2021.119983] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/11/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUNDS Modern dietary habits have been associated with Nonalcoholic Steatohepatitis (NASH). Curcumin is a natural herbal found to suppress cellular oxidative states and could be beneficial in NASH. This study investigates the effect of curcumin in an animal model of NASH. MATERIALS AND METHODS Fifty rats were allocated into five groups. Control, High Fat Diet (HFD), curcumin prophylactic (CP) and therapeutic (CT) groups. HFD regimen was given for 16 weeks. Curcumin was given along with HFD (prophylactic) or after establishment of the model for two weeks (therapeutic). Livers and blood samples were harvested for histological, biochemical, and molecular studies. KEY FINDINGS Livers from HFD groups showed vascular, inflammatory, cellular degenerative and fibrotic changes. The hepatic damage was reflected by the increased serum liver enzymes. HFD groups showed excessive fibrotic change. Interestingly, curcumin administration as prophylactic or therapeutic significantly preserved and/or restored liver structure. This was evidenced by the normalization of the liver enzymes, preservation and/or reversibility of cellular changes and the decrease of the stage of fibrosis. Nuclear factor E2-related factor 2 gene (Nrf2) expression showed no changes in the HFD groups, however it showed upregulation in curcumin treated groups. Thus, the protective and therapeutic effect of curcumin could be induced through upregulation of the Nrf2 gene. Curcumin has a beneficial prophylactic and therapeutic effect that could hinder the development and/or treat NASH in susceptible livers. SIGNIFICANCE Curcumin has a beneficial prophylactic and therapeutic effect that could hinder the development and/or treat NASH in susceptible livers.
Collapse
|
33
|
Circulating Tumour Cells (CTCs) in NSCLC: From Prognosis to Therapy Design. Pharmaceutics 2021; 13:pharmaceutics13111879. [PMID: 34834295 PMCID: PMC8619417 DOI: 10.3390/pharmaceutics13111879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023] Open
Abstract
Designing optimal (neo)adjuvant therapy is a crucial aspect of the treatment of non-small-cell lung carcinoma (NSCLC). Standard methods of chemotherapy, radiotherapy, and immunotherapy represent effective strategies for treatment. However, in some cases with high metastatic activity and high levels of circulating tumour cells (CTCs), the efficacy of standard treatment methods is insufficient and results in treatment failure and reduced patient survival. CTCs are seen not only as an isolated phenomenon but also a key inherent part of the formation of metastasis and a key factor in cancer death. This review discusses the impact of NSCLC therapy strategies based on a meta-analysis of clinical studies. In addition, possible therapeutic strategies for repression when standard methods fail, such as the administration of low-toxicity natural anticancer agents targeting these phenomena (curcumin and flavonoids), are also discussed. These strategies are presented in the context of key mechanisms of tumour biology with a strong influence on CTC spread and metastasis (mechanisms related to tumour-associated and -infiltrating cells, epithelial–mesenchymal transition, and migration of cancer cells).
Collapse
|
34
|
Feng N, Guo Z, Wu X, Tian Y, Li Y, Geng Y, Yu Y. Circ_PIP5K1A regulates cisplatin resistance and malignant progression in non-small cell lung cancer cells and xenograft murine model via depending on miR-493-5p/ROCK1 axis. Respir Res 2021; 22:248. [PMID: 34537072 PMCID: PMC8449484 DOI: 10.1186/s12931-021-01840-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023] Open
Abstract
Background Chemoresistance limits the therapeutic effect of cisplatin (DDP) on non-small cell lung cancer (NSCLC). Circular RNAs (circRNAs) function as important regulators in chemoresistance. This study aimed to explore the regulation of circRNA Phosphatidylinositol-4-Phosphate 5-Kinase Type 1 Alpha (circ_PIP5K1A) in DDP resistance. Methods The expression analysis of circ_PIP5K1A, micoRNA-493-5p (miR-493-5p) and Rho Associated Coiled-Coil Containing Protein Kinase 1 (ROCK1) was conducted through reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Cell sensitivity was determined using 3-(4,5-dimethylthiazol-2-y1)-2,5-diphenyl tetrazolium bromide (MTT) assay. Cell proliferation and cell viability were evaluated by colony formation assay and MTT assay, respectively. Cell cycle and apoptosis detection was performed via flow cytometry. Cell motility was examined by transwell migration or invasion assay. Dual-luciferase reporter assay was applied to confirm the target binding. ROCK1 protein level was assayed via Western blot. In vivo assay was carried out using xenograft model in mice. Results Circ_PIP5K1A level was abnormally increased in DDP-resistant NSCLC tissues and cells. Silencing circ_PIP5K1A reduced DDP resistance, proliferation, cell cycle progression and cell motility in DDP-resistant NSCLC cells. Circ_PIP5K1A directly interacted with miR-493-5p in NSCLC cells. The function of circ_PIP5K1A was dependent on the negative regulation of miR-493-5p. MiR-493-5p directly targeted ROCK1 and circ_PIP5K1A regulated the ROCK1 level via acting as a sponge of miR-493-5p. Overexpression of miR-493-5p inhibited chemoresistance and cancer progression by downregulating ROCK1 expression in DDP-resistant NSCLC cells. Circ_PIP5K1A regulated DDP sensitivity in vivo via the miR-493-5p/ROCK1 axis. Conclusion These findings suggested that circ_PIP5K1A upregulated the ROCK1 expression to promote DDP resistance and cancer progression in NSCLC by sponging miR-493-5p.
Collapse
Affiliation(s)
- Nan Feng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 West Fifth Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Zhi Guo
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 West Fifth Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Xiaokang Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 West Fifth Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Ying Tian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 West Fifth Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Yue Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 West Fifth Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Yan Geng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157 West Fifth Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China.
| | - Yan Yu
- The Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
35
|
Identification of Compound CB-2 as a Novel Late-Stage Autophagy Inhibitor Exhibits Inhibitory Potency against A549 Cells. Life (Basel) 2021; 11:life11080865. [PMID: 34440609 PMCID: PMC8401303 DOI: 10.3390/life11080865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/12/2021] [Accepted: 08/20/2021] [Indexed: 01/07/2023] Open
Abstract
Autophagy has been recognized as a stress tolerance mechanism that maintains cell viability, which contributes to tumor progression, dormancy, and treatment resistance. The inhibition of autophagy in cancer has the potential to improve the therapeutic efficacy. It is therefore of great significance to search for new autophagy inhibitors. In the present study, after screening a series of curcumin derivatives synthesized in our laboratory, (E)-3-((E)-4-chlorobenzylidene)-5-((5-methoxy-1H-indol-3-yl)methylene)-1-methylpiperidin-4-one (CB-2) was selected as a candidate for further study. We found that CB-2 increased the LC3B-II and SQSTM1 levels associated with the accumulation of autophagosomes in non-small cell lung cancer (NSCLC) A549 cells. The increased level of LC3B-II induced by CB-2 was neither eliminated when autophagy initiation was suppressed by wortmannin nor further increased when autophagosome degradation was inhibited by chloroquine (CQ). CB-2 enhanced the accumulation of LC3B-II under starvation conditions. Further studies revealed that CB-2 did not affect the levels of the key proteins involved in autophagy induction but significantly blocked the fusion of autophagosomes with lysosomes. High-dose CB-2 induced the apoptosis and necrosis of A549 cells, while a lower dose of CB-2 mainly impaired the migrative capacity of A549 cells, which only slightly induced cell apoptosis. CB-2 increased the levels of mitochondrial-derived reactive oxygen species (ROS) while decreasing the mitochondrial membrane potential (MMP). Scavenging ROS via N-acetylcysteine (NAC) reversed CB-2-induced autophagy inhibition and its inhibitory effect against A549 cells. In conclusion, CB-2 serves as a new late-stage autophagy inhibitor, which has a strong inhibitory potency against A549 cells.
Collapse
|
36
|
Wang Y, Guo M, Lin D, Liang D, Zhao L, Zhao R, Wang Y. Docetaxel-loaded exosomes for targeting non-small cell lung cancer: preparation and evaluation in vitro and in vivo. Drug Deliv 2021; 28:1510-1523. [PMID: 34263685 PMCID: PMC8284156 DOI: 10.1080/10717544.2021.1951894] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a highly lethal disease and the majority of NSCLC patients are desperate for therapies that can effectively target their cancer and ultimately improve their overall survival. Docetaxel (DTX) represents the first-line of the antitumor agent that is used to treat NSCLC; however, it has poor solubility in water and unsatisfactory encapsulation efficiency. In our study, exosomes were isolated from A549 cancer cells by ultracentrifugation and then characterized using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot (WB). The particle size changes of EXO and EXO-DTX were measured daily for seven days to test the stability. DTX was selected payload by electroporation (EXO-DTX). For the in vitro evaluation, cell proliferation, cell cycle, cell apoptosis, reactive oxygen species (ROS) assay and cellular uptake were evaluated in the A549 cells. Also, this study evaluated the target and therapeutic effect of DTX as an antitumor agent in vivo. As a result, EXO-DTX with a particle size of 149.5 nm were successfully prepared and the cytotoxicity of the EXO-DTX was much greater than that of DTX monomers. Exosomes significantly increased the cellular uptake in vitro evaluation and showed better targeting to tumor tissue compared to the free DTX in the mice. We also explored the potential of tumor cell-derived exosomes as a drug delivery agent to target the parent cancer. Hence, we conclude that exosomes might be used as a potential antitumor drug delivery system (DDS).
Collapse
Affiliation(s)
- Ying Wang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Mimi Guo
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dingmei Lin
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dajun Liang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ling Zhao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ruizhi Zhao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
37
|
Morshedi K, Borran S, Ebrahimi MS, Masoud Khooy MJ, Seyedi ZS, Amiri A, Abbasi-Kolli M, Fallah M, Khan H, Sahebkar A, Mirzaei H. Therapeutic effect of curcumin in gastrointestinal cancers: A comprehensive review. Phytother Res 2021; 35:4834-4897. [PMID: 34173992 DOI: 10.1002/ptr.7119] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/18/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Gastrointestinal (GI) cancers with a high global prevalence are a leading cause of morbidity and mortality. Accordingly, there is a great need to develop efficient therapeutic approaches. Curcumin, a naturally occurring agent, is a promising compound with documented safety and anticancer activities. Recent studies have demonstrated the activity of curcumin in the prevention and treatment of different cancers. According to systematic studies on curcumin use in various diseases, it can be particularly effective in GI cancers because of its high bioavailability in the gastrointestinal tract. Nevertheless, the clinical applications of curcumin are largely limited because of its low solubility and low chemical stability in water. These limitations may be addressed by the use of relevant analogues or novel delivery systems. Herein, we summarize the pharmacological effects of curcumin against GI cancers. Moreover, we highlight the application of curcumin's analogues and novel delivery systems in the treatment of GI cancers.
Collapse
Affiliation(s)
- Korosh Morshedi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sarina Borran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Zeynab Sadat Seyedi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Fallah
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
38
|
Wu Q, Ou H, Shang Y, Zhang X, Wu J, Fan F. Nanoscale Formulations: Incorporating Curcumin into Combination Strategies for the Treatment of Lung Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2695-2709. [PMID: 34188448 PMCID: PMC8232383 DOI: 10.2147/dddt.s311107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer remains the most common cancer worldwide. Although significant advances in screening have been made and early diagnosis strategies and therapeutic regimens have been developed, the overall survival rate remains bleak. Curcumin is extracted from the rhizomes of turmeric and exhibits a wide range of biological activities. In lung cancer, evidence has shown that curcumin can markedly inhibit tumor growth, invasion and metastasis, overcome resistance to therapy, and even eliminate cancer stem cells (CSCs). Herein, the underlying molecular mechanisms of curcumin were summarized by distinct biological processes. To solve the limiting factors that curtail the clinical applications of curcumin, nanoformulations encapsulating curcumin were surveyed in detail. Nanoparticles, including liposomes, micelles, carbon nanotubes (CNTs), solid lipid nanoparticles (SLNs), nanosuspensions, and nanoemulsions, were explored as proper carriers of curcumin. Moreover, it was firmly verified that curcumin has the ability to sensitize lung cancer cells to chemotherapeutic drugs, such as cisplatin and docetaxel, and to various targeted therapies. Regarding the advantages and drawbacks of curcumin, we concluded that combination therapy based on nanoparticles would be the optimal approach to broaden the application of curcumin in the clinic in the near future.
Collapse
Affiliation(s)
- Quhui Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Huiping Ou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Yan Shang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Xi Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Junyong Wu
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Fuyuan Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| |
Collapse
|
39
|
Malik P, Hoidal JR, Mukherjee TK. Recent Advances in Curcumin Treated Non-Small Cell Lung Cancers: An Impetus of Pleiotropic Traits and Nanocarrier Aided Delive ry. Curr Med Chem 2021; 28:3061-3106. [PMID: 32838707 DOI: 10.2174/0929867327666200824110332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023]
Abstract
Characterized by the abysmal 18% five year survival chances, non-small cell lung cancers (NSCLCs) claim more than half of their sufferers within the first year of being diagnosed. Advances in biomedical engineering and molecular characterization have reduced the NSCLC diagnosis via timid screening of altered gene expressions and impaired cellular responses. While targeted chemotherapy remains a major option for NSCLCs complications, delayed diagnosis, and concurrent multi-drug resistance remain potent hurdles in regaining normalcy, ultimately resulting in relapse. Curcumin administration presents a benign resolve herein, via simultaneous interception of distinctly expressed pathological markers through its pleiotropic attributes and enhanced tumor cell internalization of chemotherapeutic drugs. Studies on NSCLC cell lines and related xenograft models have revealed a consistent decline in tumor progression owing to enhanced chemotherapeutics cellular internalization via co-delivery with curcumin. This presents an optimum readiness for screening the corresponding effectiveness in clinical subjects. Curcumin is delivered to NSCLC cells either (i) alone, (ii) in stoichiometrically optimal combination with chemotherapeutic drugs, (iii) through nanocarriers, and (iv) nanocarrier co-delivered curcumin and chemotherapeutic drugs. Nanocarriers protect the encapsulated drug from accidental and non-specific spillage. A unanimous trait of all nanocarriers is their moderate drug-interactions, whereby native structural expressions are not tampered. With such insights, this article focuses on the implicit NSCLC curative mechanisms viz-a-viz, free curcumin, nanocarrier delivered curcumin, curcumin + chemotherapeutic drug and nanocarrier assisted curcumin + chemotherapeutic drug delivery.
Collapse
Affiliation(s)
- Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, India
| | - John R Hoidal
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Tapan K Mukherjee
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
40
|
Liang T, Sun X, Li W, Hou G, Gao F. 1,2,3-Triazole-Containing Compounds as Anti-Lung Cancer Agents: Current Developments, Mechanisms of Action, and Structure-Activity Relationship. Front Pharmacol 2021; 12:661173. [PMID: 34177578 PMCID: PMC8226129 DOI: 10.3389/fphar.2021.661173] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the most common malignancy and leads to around one-quarter of all cancer deaths. Great advances have been achieved in the treatment of lung cancer with novel anticancer agents and improved technology. However, morbidity and mortality rates remain extremely high, calling for an urgent need to develop novel anti-lung cancer agents. 1,2,3-Triazole could be readily interact with diverse enzymes and receptors in organisms through weak interaction. 1,2,3-Triazole can not only be acted as a linker to tether different pharmacophores but also serve as a pharmacophore. This review aims to summarize the recent advances in 1,2,3-triazole-containing compounds with anti-lung cancer potential, and their structure-activity relationship (SAR) together with mechanisms of action is also discussed to pave the way for the further rational development of novel anti-lung cancer candidates.
Collapse
Affiliation(s)
- Ting Liang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiangyang Sun
- Department of Interventional Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhong Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guihua Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
41
|
Tavakoli F, Khatami SS, Momeni F, Azadbakht J, Ghasemi F. Cervical Cancer Diagnosis: Insights into Biochemical Biomarkers and Imaging Techniques. Comb Chem High Throughput Screen 2021; 24:605-623. [PMID: 32875976 DOI: 10.2174/1386207323666200901101955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/14/2020] [Accepted: 08/06/2020] [Indexed: 11/22/2022]
Abstract
Cervical malignancy is known as one of the important cancers which is originated from cervix. This malignancy has been observed in women infected with papillomavirus who had regular oral contraceptives, multiple pregnancies, and sexual relations. Early and fast cervical cancer diagnosis is known as two important aspects of cervical cancer therapy. Several investigations indicated that early and fast detection of cervical cancer could be associated with better treatment process and increasing survival rate of patients with this malignancy. Imaging techniques are very important diagnosis tools that could be employed for diagnosis and following responses to therapy in various cervical cancer stages. Multiple lines of evidence indicated that utilization of imaging techniques is related to some limitations (i.e. high cost, and invasive effects). Hence, it seems that along with using imaging techniques, finding and developing new biomarkers could be useful in the diagnosis and treatment of subjects with cervical cancer. Taken together, many studies showed that a variety of biomarkers including, several proteins, mRNAs, microRNAs, exosomes and polymorphisms might be introduced as prognostic, diagnostic and therapeutic biomarkers in cervical cancer therapy. In this review article, we highlighted imaging techniques as well as novel biomarkers for the diagnosis of cervical cancer.
Collapse
Affiliation(s)
- Fatemeh Tavakoli
- Department of Biotechnology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Sadat Khatami
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Fatemeh Momeni
- Isfahan Research Committee of Multiple Sclerosis, Alzahra Research Institute, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Javid Azadbakht
- Department of Radiology and Imaging, Kashan University of Medical Science, Kashan, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
42
|
Tian S, Liao L, Zhou Q, Huang X, Zheng P, Guo Y, Deng T, Tian X. Curcumin inhibits the growth of liver cancer by impairing myeloid-derived suppressor cells in murine tumor tissues. Oncol Lett 2021; 21:286. [PMID: 33732362 PMCID: PMC7905673 DOI: 10.3892/ol.2021.12547] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Curcumin, one of the active ingredients of Curcuma longa (Jianghuang), has been reported to exert multiple bioactivities, including pro-apoptotic and anti-inflammatory activities. In recent years, curcumin has been extensively studied, and it has been revealed that curcumin inhibits the growth of numerous types of cancer. However, to the best of our knowledge, the inhibitory effects of curcumin on the activation or expansion of myeloid-derived suppressor cells (MDSCs) in liver cancer and the underlying mechanism have not yet been determined. Therefore, the present study aimed to investigate the inhibitory effect of curcumin on MDSC activity and the associated anti-neoplastic mechanism in a HepG2 ×enograft mouse model. The effect of curcumin on the viability of Huh-7, MHCC-97H and HepG2 cells in vitro was analyzed using a Cell Counting Kit-8 assay. The effects of curcumin on tumor growth, numbers of MDSCs, expression levels of proteins involved in the toll-like receptor 4 (TLR4)/NF-κB signaling pathway, levels of related inflammatory factors and angiogenesis were determined in HepG2 ×enograft model mice, which were given different doses of curcumin via intragastrical administration. The results of the present study revealed that curcumin inhibited the viability of Huh-7, MHCC-97H and HepG2 cells and the growth of HepG2 ×enograft tumors in mice. Flow cytometric analysis indicated that curcumin reduced the number of MDSCs in mouse xenograft tumors. In addition, the results demonstrated that curcumin inhibited the TLR4/NF-κB signaling pathway and the expression of inflammatory factors, including IL-6, IL-1β, prostaglandin E2 and cyclooxygenase-2, in mouse xenograft tumors. Furthermore, curcumin suppressed the secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte-colony stimulating factor (G-CSF), which are essential factors for MDSCs modulation, in tumor tissues. Additionally, curcumin was revealed to inhibit angiogenesis, which was demonstrated by the downregulation of the expression levels of vascular endothelial growth factor, CD31 and α-smooth muscle actin in western blotting, immunohistochemistry and immunofluorescence experiments. In conclusion, the findings of the present study identified a novel mechanism via which curcumin may suppress the growth of liver cancer by reducing the numbers of MDSCs and subsequently disrupting the process of angiogenesis. These conclusions were supported by the observed inactivation of the TLR4/NF-κB signaling pathway-mediated inflammatory response and the downregulation of GM-CSF and G-CSF secretion in xenograft tissues.
Collapse
Affiliation(s)
- Sha Tian
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Liu Liao
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xiaodi Huang
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Piao Zheng
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yinmei Guo
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Tianhao Deng
- Department of Oncology, The Affiliated Hospital of Hunan Institute of Chinese Medicine, Changsha, Hunan 410006, P.R. China
| | - Xuefei Tian
- Department of Internal Medicine, College of Integrated Chinese and Western Medicine of Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
43
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Rahmani Moghadam E, Raei M, Kalantari M, Tavakol S, Mohammadinejad R, Najafi M, Tay FR, Makvandi P. Progress in Natural Compounds/siRNA Co-delivery Employing Nanovehicles for Cancer Therapy. ACS COMBINATORIAL SCIENCE 2020; 22:669-700. [PMID: 33095554 PMCID: PMC8015217 DOI: 10.1021/acscombsci.0c00099] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Chemotherapy using natural compounds, such as resveratrol, curcumin, paclitaxel, docetaxel, etoposide, doxorubicin, and camptothecin, is of importance in cancer therapy because of the outstanding therapeutic activity and multitargeting capability of these compounds. However, poor solubility and bioavailability of natural compounds have limited their efficacy in cancer therapy. To circumvent this hurdle, nanocarriers have been designed to improve the antitumor activity of the aforementioned compounds. Nevertheless, cancer treatment is still a challenge, demanding novel strategies. It is well-known that a combination of natural products and gene therapy is advantageous over monotherapy. Delivery of multiple therapeutic agents/small interfering RNA (siRNA) as a potent gene-editing tool in cancer therapy can maximize the synergistic effects against tumor cells. In the present review, co-delivery of natural compounds/siRNA using nanovehicles are highlighted to provide a backdrop for future research.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty
of Engineering and Natural Sciences, Sabanci
University, Orta Mahalle,
Üniversite Caddesi No. 27, Orhanlı,
Tuzla, 34956 Istanbul, Turkey
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Ali Zarrabi
- Sabanci
University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul Turkey
| | - Kiavash Hushmandi
- Department
of Food Hygiene and Quality Control, Division of Epidemiology &
Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran
| | - Farid Hashemi
- Department
of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department
of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Mehdi Raei
- Health Research
Center, Life Style Institute, Baqiyatallah
University of Medical Sciences, Tehran 1435916471, Iran
| | - Mahshad Kalantari
- Department
of Genetics, Tehran Medical Sciences Branch, Azad University, Tehran 19168931813, Iran
| | - Shima Tavakol
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 1449614525, Iran
| | - Reza Mohammadinejad
- Pharmaceutics
Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Masoud Najafi
- Medical
Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Radiology
and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Franklin R. Tay
- College
of Graduate Studies, Augusta University, Augusta, Georgia 30912, United States
| | - Pooyan Makvandi
- Istituto
Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa Italy
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 14496-14535 Tehran, Iran
| |
Collapse
|
44
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Zarrin V, Moghadam ER, Hashemi F, Makvandi P, Samarghandian S, Khan H, Hashemi F, Najafi M, Mirzaei H. Toward Regulatory Effects of Curcumin on Transforming Growth Factor-Beta Across Different Diseases: A Review. Front Pharmacol 2020; 11:585413. [PMID: 33381035 PMCID: PMC7767860 DOI: 10.3389/fphar.2020.585413] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Immune response, proliferation, migration and angiogenesis are juts a few of cellular events that are regulated by transforming growth factor-β (TGF-β) in cells. A number of studies have documented that TGF-β undergoes abnormal expression in different diseases, e.g., diabetes, cancer, fibrosis, asthma, arthritis, among others. This has led to great fascination into this signaling pathway and developing agents with modulatory impact on TGF-β. Curcumin, a natural-based compound, is obtained from rhizome and roots of turmeric plant. It has a number of pharmacological activities including antioxidant, anti-inflammatory, anti-tumor, anti-diabetes and so on. Noteworthy, it has been demonstrated that curcumin affects different molecular signaling pathways such as Wnt/β-catenin, Nrf2, AMPK, mitogen-activated protein kinase and so on. In the present review, we evaluate the potential of curcumin in regulation of TGF-β signaling pathway to corelate it with therapeutic impacts of curcumin. By modulation of TGF-β (both upregulation and down-regulation), curcumin ameliorates fibrosis, neurological disorders, liver disease, diabetes and asthma. Besides, curcumin targets TGF-β signaling pathway which is capable of suppressing proliferation of tumor cells and invading cancer cells.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Pooyan Makvandi
- Centre for Micro-BioRobotics, Istituto Italiano di Tecnologia, Pisa, Italy
| | | | - Haroon Khan
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fardin Hashemi
- Medical Technology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
45
|
Ashrafizadeh M, Najafi M, Makvandi P, Zarrabi A, Farkhondeh T, Samarghandian S. Versatile role of curcumin and its derivatives in lung cancer therapy. J Cell Physiol 2020; 235:9241-9268. [PMID: 32519340 DOI: 10.1002/jcp.29819] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/24/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022]
Abstract
Lung cancer is a main cause of death all over the world with a high incidence rate. Metastasis into neighboring and distant tissues as well as resistance of cancer cells to chemotherapy demand novel strategies in lung cancer therapy. Curcumin is a naturally occurring nutraceutical compound derived from Curcuma longa (turmeric) that has great pharmacological effects, such as anti-inflammatory, neuroprotective, and antidiabetic. The excellent antitumor activity of curcumin has led to its extensive application in the treatment of various cancers. In the present review, we describe the antitumor activity of curcumin against lung cancer. Curcumin affects different molecular pathways such as vascular endothelial growth factors, nuclear factor-κB (NF-κB), mammalian target of rapamycin, PI3/Akt, microRNAs, and long noncoding RNAs in treatment of lung cancer. Curcumin also can induce autophagy, apoptosis, and cell cycle arrest to reduce the viability and proliferation of lung cancer cells. Notably, curcumin supplementation sensitizes cancer cells to chemotherapy and enhances chemotherapy-mediated apoptosis. Curcumin can elevate the efficacy of radiotherapy in lung cancer therapy by targeting various signaling pathways, such as epidermal growth factor receptor and NF-κB. Curcumin-loaded nanocarriers enhance the bioavailability, cellular uptake, and antitumor activity of curcumin. The aforementioned effects are comprehensively discussed in the current review to further direct studies for applying curcumin in lung cancer therapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pooyan Makvandi
- Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council (CNR), Naples, Italy
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|