1
|
Evyapan G, Senturk NC, Celik IS. Ornidazole Inhibits the Angiogenesis and Migration Abilities of Non-small Cell Lung Cancer (NSCLC) via Downregulation of VEGFA/VEGFR2/NRP-1 and PI3K/AKT/mTOR Pathways. Cell Biochem Biophys 2024; 82:3277-3285. [PMID: 38886281 DOI: 10.1007/s12013-024-01358-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] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Around the world, non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths among all cancers. Despite advancements in new therapeutic approaches over the past few decades, the five-year survival rate still remains disappointing. The lack of effective anti-angiogenic and anti-migration drugs is the biggest obstacle to the treatment of metastatic lung cancer. Therefore, there is a need to develop new and effective therapeutic compounds targeting anti-angiogenic and anti-migration pathways for the treatment of lung cancer. Ornidazole is a nitroimidazole agent widely used in the treatment of parasitic infections such as trichomonas vaginalis, amebiasis and giardiasis. This study aimed to investigate the anti-proliferative, anti-angiogenic and anti-mitotic activities of the anti-parasitic drug Ornidazole in two human lung cancer cell lines (A549, H1299). METHODS We determined the effects of Ornidazole, on cell viability, apoptosis, migration, angiogenesis and metastatic ability against NSCLC in lung cancer cell lines. Its action on the mRNA and protein expression levels of VEGFA, VEGFR2, NRP1, Casp9, Casp3, Bax, Bcl-2, PIK3CA, AKT, MTOR, PTEN and FOX3A was assessed. Furthermore, in this study the effects on cell migration, cell viability and proliferation was evaluated through wound healing, MTT and Crystal violet assays. RESULTS This study demonstrated that Ornidazole effectively reduces cell viability and migration ability, inhibits angiogenesis and metastatic abilities in NSCLC cells. CONCLUSIONS In conclusion, these results may shed light on the treatment of NSCLC, and we suggest the anti-parasitic drug Ornidazole as a new agent with potential anti-angiogenic and anti-mitotic activity by interfering with the molecular pathways that trigger tumor angiogenesis and migration.
Collapse
Affiliation(s)
- Gulsah Evyapan
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey.
| | - Nesrin Cetinel Senturk
- Department of Medical Biology, Faculty of Medicine, Cukurova University, Balcali- Adana, Turkey
| | - Ibrahim Seyfettin Celik
- Department Of Medical Services And Techniques, Kahramanmaraş Health Services Vocational School, Pathology Laboratory Techniques Pr., Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| |
Collapse
|
2
|
Akar S, Cakir M, Ozkol H, Akkoc S, Ozdem B. A benzimidazolium salt induces apoptosis and arrests cells at sub-G1 phase in epithelial ovarian cancer cells. Mol Biol Rep 2024; 51:66. [PMID: 38170294 DOI: 10.1007/s11033-023-08981-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/24/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Ovarian cancer, also known as a silent killer, is the deadliest gynecological cancer in women worldwide. Epithelial ovarian cancers constitute the majority of ovarian cancers, and diagnosis can be made in advanced stages, which greatly reduces the likelihood of treatment and lowers the survival rate. For the treatment of epithelial ovarian cancers, the search for synthetic agents as well as agents of natural origin continues. The effects of 1-(2-cyanobenzyl)-3-(4-vinylbenzyl)-1H-benzo[d]imidazole-3-ium chloride (BD), a benzimidazole derivative, were investigated on epithelial ovarian cancer cells. METHODS AND RESULTS In our study, the effects of BD on proliferation, colony formation, cell death by apoptosis and the cell cycle in A2780 and A2780 Adriamycin (ADR) ovarian cancer cell lines were investigated. Proliferation was examined with cell viability analysis, colony formation and apoptosis with Annexin V staining and cell cycle analyses with PI staining, respectively. As a result of the analyses, BD inhibited cell proliferation and colony formation, induced apoptosis and cell death at 48 h in A2780 and A2780 ADR cells at 10.10 and 10.36 µM concentrations, respectively. In addition, A2780 and A2780ADR cells were arrested in the Sub-G1 phase of the cell cycle. CONCLUSIONS BD suppresses cancer cell progression by showing antiproliferative effects on ovarian cancer cells. Further analyses are required to determine the mechanism of action of this agent and to demonstrate its potential as a suitable candidate for the treatment of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Sakine Akar
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, 65090, Turkey.
| | - Mustafa Cakir
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, 65090, Turkey
| | - Halil Ozkol
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, 65090, Turkey
| | - Senem Akkoc
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
- Faculty of Engineering and Natural Sciences, Bahcesehir University, Istanbul, Turkey
| | - Berna Ozdem
- Department of Medical Biology and Genetics, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
3
|
Li C, He Z, Yao F, Liao S, Sun K, Sun S, Li Z, Wang Z. Role of Escin in breast cancer therapy: potential mechanism for inducing ferroptosis and synergistic antitumor activity with cisplatin. Apoptosis 2023:10.1007/s10495-023-01849-x. [PMID: 37149513 DOI: 10.1007/s10495-023-01849-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
Breast cancer (BC) has threatened women worldwide for a long time, and novel treatments are needed. Ferroptosis is a new form of regulated cell death that is a potential therapeutic target for BC. In this study, we identified Escin, a traditional Chinese medicine, as a possible supplement for existing chemotherapy strategies. Escin inhibited BC cell growth in vitro and in vivo, and ferroptosis is probable to be the main cause for Escin-induced cell death. Mechanistically, Escin significantly downregulated the protein level of GPX4, while overexpression of GPX4 could reverse the ferroptosis triggered by Escin. Further study revealed that Escin could promote G6PD ubiquitination and degradation, thus inhibiting the expression of GPX4 and contributing to the ferroptosis. Moreover, proteasome inhibitor MG132 or G6PD overexpression could partially reverse Escin-induced ferroptosis, when G6PD knockdown aggravated that. In vivo study also supported that downregulation of G6PD exacerbated tumor growth inhibition by Escin. Finally, our data showed that cell apoptosis was dramatically elevated by Escin combined with cisplatin in BC cells. Taken together, these results suggest that Escin inhibits tumor growth in vivo and in vitro via regulating the ferroptosis mediated by G6PD/GPX4 axis. Our findings provide a promising therapeutic strategy for BC.
Collapse
Affiliation(s)
- Chenyuan Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Feng Yao
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Shichong Liao
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Kai Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Shengrong Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| | - Zhiyu Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| | - Zhong Wang
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
4
|
Fazliev S, Tursunov K, Razzokov J, Sharipov A. Escin's Multifaceted Therapeutic Profile in Treatment and Post-Treatment of Various Cancers: A Comprehensive Review. Biomolecules 2023; 13:biom13020315. [PMID: 36830684 PMCID: PMC9952945 DOI: 10.3390/biom13020315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Although modern medicine is advancing at an unprecedented rate, basic challenges in cancer treatment and drug resistance remain. Exploiting natural-product-based drugs is a strategy that has been proven over time to provide diverse and efficient approaches in patient care during treatment and post-treatment periods of various diseases, including cancer. Escin-a plant-derived triterpenoid saponin-is one example of natural products with a broad therapeutic scope. Initially, escin was proven to manifest potent anti-inflammatory and anti-oedematous effects. However, in the last two decades, other novel activities of escin relevant to cancer treatment have been reported. Recent studies demonstrated escin's efficacy in compositions with other approved drugs to accomplish synergy and increased bioavailability to broaden their apoptotic, anti-metastasis, and anti-angiogenetic effects. Here, we comprehensively discuss and present an overview of escin's chemistry and bioavailability, and highlight its biological activities against various cancer types. We conclude the review by presenting possible future directions of research involving escin for medical and pharmaceutical applications as well as for basic research.
Collapse
Affiliation(s)
- Sunnatullo Fazliev
- Max Planck School Matter to Life, Jahnstrasse 29, 69120 Heidelberg, Germany
- Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany
| | - Khurshid Tursunov
- Department of Inorganic, Physical and Colloidal Chemistry, Tashkent Pharmaceutical Institute, Oybek Street 45, Tashkent 100015, Uzbekistan
- State Center for Expertise and Standardization of Medicines, Medical Devices and Medical Equipment, Agency for the Development of the Pharmaceutical Industry under the Ministry of Health of the Republic of Uzbekistan, Ozod Street 16, Tashkent 100002, Uzbekistan
| | - Jamoliddin Razzokov
- Institute of Fundamental and Applied Research, National Research University TIIAME, Kori Niyoziy 39, Tashkent 100000, Uzbekistan
- College of Engineering, Akfa University, Milliy Bog Street 264, Tashkent 111221, Uzbekistan
- Department of Physics, National University of Uzbekistan, Universitet 4, Tashkent 100174, Uzbekistan
- Laboratory of Experimental Biophysics, Centre for Advanced Technologies, Universitet 7, Tashkent 100174, Uzbekistan
| | - Avez Sharipov
- Department of Inorganic, Physical and Colloidal Chemistry, Tashkent Pharmaceutical Institute, Oybek Street 45, Tashkent 100015, Uzbekistan
- Department of Analytical and Pharmaceutical Chemistry, Institute of Pharmaceutical Education and Research, Yunusota Street 46, Tashkent 100114, Uzbekistan
- Correspondence:
| |
Collapse
|
5
|
Park S, Park JM, Park M, Ko D, Kim S, Seo J, Nam KD, Jung E, Farrand L, Kim YJ, Kim JY, Seo JH. β-Escin overcomes trastuzumab resistance in HER2-positive breast cancer by targeting cancer stem-like features. Cancer Cell Int 2022; 22:289. [PMID: 36127671 PMCID: PMC9490928 DOI: 10.1186/s12935-022-02713-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background The emergence of de novo or intrinsic trastuzumab resistance is exceedingly high in breast cancer that is HER2 positive and correlates with an abundant cancer stem cell (CSC)-like population. We sought to examine the capacity of β-escin, an anti-inflammatory drug, to address trastuzumab resistance in HER2-positive breast cancer cells. Methods The effect of β-escin on trastuzumab-resistant and -sensitive cell lines in vitro was evaluated for apoptosis, expression of HER2 family members, and impact on CSC-like properties. An in vivo model of trastuzumab-resistant JIMT-1 was used to examine the efficacy and toxicity of β-escin. Results β-escin induced mitochondrial-mediated apoptosis accompanied by reactive oxygen species (ROS) production and increased active p18Bax fragmentation, leading to caspase-3/-7 activation. Attenuation of CSC-related features by β-escin challenge was accompanied by marked reductions in CD44high/CD24low stem-like cells and aldehyde dehydrogenase 1 (ALDH1) activity as well as hindrance of mammosphere formation. β-escin administration also significantly retarded tumor growth and angiogenesis in a trastuzumab-resistant JIMT-1 xenograft model via downregulation of CSC-associated markers and intracellular domain HER2. Importantly, β-escin selectively inhibited malignant cells and was less toxic to normal mammary cells, and no toxic effects were found in liver and kidney function in animals. Conclusions Taken together, our findings highlight β-escin as a promising candidate for the treatment of trastuzumab-resistant HER2-positive breast cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02713-9.
Collapse
Affiliation(s)
- Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Jung Min Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Minsu Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Dongmi Ko
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Seongjae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Juyeon Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Kee Dal Nam
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Lee Farrand
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea.
| |
Collapse
|