1
|
Koopaie M, Karimi H, Sohrabi M, Norouzi H. Cytotoxic, anti-proliferative, and apoptotic evaluation of Ramalina sinensis (Ascomycota, Lecanoromycetes), lichenized fungus on oral squamous cell carcinoma cell line; in-vitro study. BMC Complement Med Ther 2023; 23:296. [PMID: 37608377 PMCID: PMC10463489 DOI: 10.1186/s12906-023-04118-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/05/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Scientists and medical professionals are actively striving to improve the efficacy of treatment methods for oral squamous cell carcinoma (OSCC), the most frequently occurring cancer within the oral cavity, by exploring the potential of natural products. The active pharmacological compounds found in lichenized fungi have shown potential for aiding in cancer treatment. Recent research aims to evaluate the impact of the lichenized fungus Ramalina sinensis (R. sinensis) on the cell viability and apoptosis of OSCC cell lines, considering the anti-inflammatory and anti-cancer capabilities of lichens. METHODS Ramalina sinensis (Ascomycota, Lecanoromycetes) was selected for investigation of its effects on a human oral squamous cell carcinoma cell line. Acetone and methanol extracts of R. sinensis on an OSCC cell line (KB cell line, NCBI Code: C152) were investigated. Viability was assessed by MTT assay analysis, and apoptotic cells were measured using flow cytometry analysis. Scratch assay was used to assess cell migration. The chemical composition and metabolic profiling of R. sinensis were investigated. RESULTS The growth and multiplication of KB cells were observed to undergo a gradual but remarkable inhibition when exposed to various concentrations. Specifically, concentrations of 6.25, 12.5, 25, 50, 100, and 200 μg/mL exhibited a significant suppressive effect on the proliferation of KB cells. The inhibition of cell proliferation exhibited a statistically significant difference between the extracts obtained from acetone and methanol. Flow cytometry results show an increase in apoptosis of OSCC cells by acetone extract. R. sinensis exerted a concentration-dependent inhibitory effect on the migration of OSCC cells. The chemical composition of R. sinensis was investigated using liquid chromatography positive ion electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS), and 33 compounds in the acetone and methanol extracts of R. sinensis were detected. CONCLUSION The findings provide evidence supporting the beneficial effects of R. sinensis extract on inducing apoptosis in OSCC cells and exerting anti-cancer properties.
Collapse
Affiliation(s)
- Maryam Koopaie
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, North Kargar St, P.O. Box: 14395 -433, Tehran, 14399-55991, Iran.
| | - Hanieh Karimi
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, North Kargar St, P.O. Box: 14395 -433, Tehran, 14399-55991, Iran
| | - Mohammad Sohrabi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Hooman Norouzi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
- Former graduate student of the Department of Horticultural Sciences, Faculty of Agriculture, Bu-Ali Sina University, Hamedan, Iran
| |
Collapse
|
2
|
Hashemi M, Paskeh MDA, Orouei S, Abbasi P, Khorrami R, Dehghanpour A, Esmaeili N, Ghahremanzade A, Zandieh MA, Peymani M, Salimimoghadam S, Rashidi M, Taheriazam A, Entezari M, Hushmandi K. Towards dual function of autophagy in breast cancer: A potent regulator of tumor progression and therapy response. Biomed Pharmacother 2023; 161:114546. [PMID: 36958191 DOI: 10.1016/j.biopha.2023.114546] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023] Open
Abstract
As a devastating disease, breast cancer has been responsible for decrease in life expectancy of females and its morbidity and mortality are high. Breast cancer is the most common tumor in females and its treatment has been based on employment of surgical resection, chemotherapy and radiotherapy. The changes in biological behavior of breast tumor relies on genomic and epigenetic mutations and depletions as well as dysregulation of molecular mechanisms that autophagy is among them. Autophagy function can be oncogenic in increasing tumorigenesis, and when it has pro-death function, it causes reduction in viability of tumor cells. The carcinogenic function of autophagy in breast tumor is an impediment towards effective therapy of patients, as it can cause drug resistance and radio-resistance. The important hallmarks of breast tumor such as glucose metabolism, proliferation, apoptosis and metastasis can be regulated by autophagy. Oncogenic autophagy can inhibit apoptosis, while it promotes stemness of breast tumor. Moreover, autophagy demonstrates interaction with tumor microenvironment components such as macrophages and its level can be regulated by anti-tumor compounds in breast tumor therapy. The reasons of considering autophagy in breast cancer therapy is its pleiotropic function, dual role (pro-survival and pro-death) and crosstalk with important molecular mechanisms such as apoptosis. Moreover, current review provides a pre-clinical and clinical evaluation of autophagy in breast tumor.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sima Orouei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pegah Abbasi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esmaeili
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azin Ghahremanzade
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari 4815733971, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
3
|
Spjut R, Simon A, Guissard M, Magain N, Sérusiaux E. The fruticose genera in the Ramalinaceae (Ascomycota, Lecanoromycetes): their diversity and evolutionary history. MycoKeys 2020; 73:1-68. [PMID: 32994702 PMCID: PMC7501315 DOI: 10.3897/mycokeys.73.47287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 07/19/2020] [Indexed: 02/08/2023] Open
Abstract
We present phylogenetic analyses of the fruticose Ramalinaceae based on extensive collections from many parts of the world, with a special focus on the Vizcaíno deserts in north-western Mexico and the coastal desert in Namibia. We generate a four-locus DNA sequence dataset for accessions of Ramalina and two additional loci for Niebla and Vermilacinia. Four genera are strongly supported: the subcosmopolitan Ramalina, the new genus Namibialina endemic to SW Africa, and a duo formed by Niebla and Vermilacinia, endemic to the New World except the sorediate V. zebrina that disjunctly occurs in Namibia. The latter three genera are restricted to coastal desert and chaparral where vegetation depends on moisture from ocean fog. Ramalina is subcosmopolitan and much more diverse in its ecology. We show that Ramalina and its sister genus Namibialina diverged from each other at c. 48 Myrs, whereas Vermilacinia and Niebla split at c. 30 Myrs. The phylogeny of the fruticose genera remains unresolved to their ancestral crustose genera. Species delimitation within Namibialina and Ramalina is rather straightforward. The phylogeny and taxonomy of Vermilacinia are fully resolved, except for the two youngest clades of corticolous taxa, and support current taxonomy, including four new taxa described here. Secondary metabolite variation in Niebla generally coincides with major clades which are comprised of species complexes with still unresolved phylogenetic relationships. A micro-endemism pattern of allopatric species is strongly suspected for both genera, except for the corticolous taxa within Vermilacinia. Both Niebla and saxicolous Vermilacinia have chemotypes unique to species clades that are largely endemic to the Vizcaíno deserts. The following new taxa are described: Namibialina gen. nov. with N. melanothrix (comb. nov.) as type species, a single new species of Ramalina (R. krogiae) and four new species of Vermilacinia (V. breviloba, V. lacunosa, V. pustulata and V. reticulata). The new combination V. granulans is introduced. Two epithets are re-introduced for European Ramalina species: R. crispans (= R. peruviana auct. eur.) and R. rosacea (= R. bourgeana auct. p.p). A lectotype is designated for Vermilacinia procera. A key to saxicolous species of Vermilacinia is presented.
Collapse
Affiliation(s)
- Richard Spjut
- World Botanical Associates, PO Box 81145, Bakersfield, California 93380, USA World Botanical Associates Bakersfield, CA United States of America
| | - Antoine Simon
- Evolution and Conservation Biology Unit, Sart Tilman B22, Quartier Vallée 1, chemin de la vallée 4, B-4000 Liège, Belgium Evolution and Conservation Biology Unit Liège Belgium
| | - Martin Guissard
- Evolution and Conservation Biology Unit, Sart Tilman B22, Quartier Vallée 1, chemin de la vallée 4, B-4000 Liège, Belgium Evolution and Conservation Biology Unit Liège Belgium
| | - Nicolas Magain
- Evolution and Conservation Biology Unit, Sart Tilman B22, Quartier Vallée 1, chemin de la vallée 4, B-4000 Liège, Belgium Evolution and Conservation Biology Unit Liège Belgium
| | - Emmanuël Sérusiaux
- Evolution and Conservation Biology Unit, Sart Tilman B22, Quartier Vallée 1, chemin de la vallée 4, B-4000 Liège, Belgium Evolution and Conservation Biology Unit Liège Belgium
| |
Collapse
|
4
|
Yu B, Yuan B, Li J, Kiyomi A, Kikuchi H, Hayashi H, Hu X, Okazaki M, Sugiura M, Hirano T, Fan Y, Pei X, Takagi N. JNK and Autophagy Independently Contributed to Cytotoxicity of Arsenite combined With Tetrandrine via Modulating Cell Cycle Progression in Human Breast Cancer Cells. Front Pharmacol 2020; 11:1087. [PMID: 32765280 PMCID: PMC7379898 DOI: 10.3389/fphar.2020.01087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Novel therapeutic strategies for breast cancer are urgently needed due to the sustained development of drug resistance and tumor recurrence. Trivalent arsenic derivative (arsenite, AsIII) has been reported to induce cytotoxicity in breast cancer cells. We recently demonstrated that AsIII plus tetrandrine (Tetra), a Chinese plant-derived alkaloid, exerted potent antitumor activity against human breast cancer cells, however, the underlying mechanisms for their action have not been well defined. In order to provide fundamental insights for understanding the action of AsIII plus Tetra, the effects of the combined regimen on two breast cancer cell lines T47D and MDA-MB-231 were evaluated. Compared to T47D cells, MDA-MB-231 cells were much more susceptible to the synergistic cytotoxic effects of AsIII and Tetra. Besides the induction of apoptotic/necrotic cell death, S-phase arrest and autophagic cell death were also observed in MDA-MB-231 cells. Exposure of MDA-MB-231 cells to AsIII and Tetra caused the activation of MAPKs. Cytotoxicity of the combined regimen in MDA-MB-231 cell was significantly abrogated by SP600125, a potent c-Jun N-terminal kinase (JNK) inhibitor. However, similar abrogation was not caused by p38 and ERK inhibitors. The addition of either autophagy inhibitors (3-methyladenine or wortmannin) or SP600125 corrected the combined regimen-triggered S-phase arrest, whereas had little effect on the apoptosis/necrosis induction in the cells. Surprisingly, SP600125NC, a negative control for SP600125, significantly strengthened S-phase arrest and the cytotoxicity induced by the combined regimen. The addition of SP600125 did not alter autophagy induction. In conclusion, the cytotoxicity of AsIII combined with Tetra was attributed to the induction of S-phase arrest, apoptotic/necrotic and autophagic cell death. The enhanced cytotoxicity of the two drugs by SP600125NC might be explained by its capability to strengthen S-phase arrest. Our results suggested that JNK and autophagy independently contributed to the cytotoxicity via modulating cell cycle progression. The study further provides fundamental insights for the development of AsIII in combination with Tetra for patients with different types of breast cancer.
Collapse
Affiliation(s)
- Bowen Yu
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan.,Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Bo Yuan
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan.,Laboratory of Pharmacology, School of Pharmacy, Josai University, Saitama, Japan
| | - JingZhe Li
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anna Kiyomi
- Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Xiaomei Hu
- Hematology Department, XiYuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mari Okazaki
- Laboratory of Pharmacology, School of Pharmacy, Josai University, Saitama, Japan
| | - Munetoshi Sugiura
- Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Toshihiko Hirano
- Clinical Pharmacology, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Yingyi Fan
- Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaohua Pei
- Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| |
Collapse
|
5
|
Vo QV, Tam NM, Bay MV, Mechler A. The radical scavenging activity of natural ramalin: A mechanistic and kinetic study. Chem Phys Lett 2020. [DOI: 10.1016/j.cplett.2019.137004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
6
|
Ozturk S, Erkisa M, Oran S, Ulukaya E, Celikler S, Ari F. Lichens exerts an anti-proliferative effect on human breast and lung cancer cells through induction of apoptosis. Drug Chem Toxicol 2019; 44:259-267. [PMID: 30835567 DOI: 10.1080/01480545.2019.1573825] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Successful cancer treatment still requires new complexes or compounds from natural sources. Therefore, we investigated anti-growth/apoptotic effects of methanol extracts of the lichen species (Xanthoparmelia somloensis (Gleyn.) Hale, Usnea intermedia (A. Massal.) Jatta, Bryoria capillaris (Ach.) Brodo & D. Hawksw and Lobaria pulmonaria (L.) Hoffm.) on human lung (A549, H1299) and breast (MCF-7, MDA-MB-231) cancer cell lines. Anti-growth effects were monitored by the MTT and ATP viability assays. Cell death mode was evaluated by employing the fluorescence staining of nucleus, caspase-cleaved cytokeratin 18 detection, caspase 3/7 activity assay, Anneksin V cytofluorimetric assay and mitochondria membrane potential assay. Among the lichen extracts, Usnea intermedia exhibited strong anti-growth activity in a dose-dependent manner (1.56-100 µg/ml) compared to the others. Usnea intermedia was especially cytotoxic against MDA-MB-231 and H1299 cells (IC50 value for was found 3.0 and 10.2 μg/ml respectively). The cytotoxicity was resulted from apoptosis as proved by the presence of pyknotic nuclei, caspase 3/7 activity, phosphatidylserine translocation and loss of mitochondria membrane potential. In conclusion, Usnea intermedia warrants for further in vivo evaluation as a new alternative in cancer treatment.
Collapse
Affiliation(s)
- Sule Ozturk
- Department of Biology, Faculty of Science and Arts, Bursa Uludag University, Bursa, Turkey
| | - Merve Erkisa
- Department of Medical Biochemistry, School of Medicine, Istinye University, Istanbul, Turkey
| | - Seyhan Oran
- Department of Biology, Faculty of Science and Arts, Bursa Uludag University, Bursa, Turkey
| | - Engin Ulukaya
- Department of Medical Biochemistry, School of Medicine, Istinye University, Istanbul, Turkey
| | - Serap Celikler
- Department of Biology, Faculty of Science and Arts, Bursa Uludag University, Bursa, Turkey
| | - Ferda Ari
- Department of Biology, Faculty of Science and Arts, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
7
|
Suh SS, Kim TK, Kim JE, Hong JM, Nguyen TTT, Han SJ, Youn UJ, Yim JH, Kim IC. Anticancer Activity of Ramalin, a Secondary Metabolite from the Antarctic Lichen Ramalina terebrata, against Colorectal Cancer Cells. Molecules 2017; 22:molecules22081361. [PMID: 28817102 PMCID: PMC6152360 DOI: 10.3390/molecules22081361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 01/06/2023] Open
Abstract
Colorectal cancer is a leading cause of death worldwide and occurs through the highly complex coordination of multiple cellular pathways, resulting in carcinogenesis. Recent studies have increasingly revealed that constituents of lichen extracts exhibit potent pharmaceutical activities, including anticancer activity against various cancer cells, making them promising candidates for new anticancer therapeutic drugs. The main objective of this study was to evaluate the anticancer capacities of ramalin, a secondary metabolite from the Antarctic lichen Ramalina terebrata, in the human colorectal cancer cell line HCT116. In this study, ramalin displayed concentration-dependent anticancer activity against HCT116 cells, significantly suppressing proliferation and inducing apoptosis. Furthermore, ramalin induced cell cycle arrest in the gap 2/mitosis (G2/M) phase through the modulation of hallmark genes involved in the G2/M phase transition, such as tumour protein p53 (TP53), cyclin-dependent kinase inhibitor 1A (CDKN1A), cyclin-dependent kinase 1 (CDK1) and cyclin B1 (CCNB1). At both the transcriptional and translational level, ramalin caused a gradual increase in the expression of TP53 and its downstream gene CDKN1A, while decreasing the expression of CDK1 and CCNB1 in a concentration-dependent manner. In addition, ramalin significantly inhibited the migration and invasion of colorectal cancer cells in a concentration-dependent manner. Taken together, these data suggest that ramalin may be a therapeutic candidate for the targeted therapy of colorectal cancer.
Collapse
Affiliation(s)
- Sung-Suk Suh
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
| | - Tai Kyoung Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
| | - Jung Eun Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
- Department of Pharmacy, Graduate School, Sungkyunkwan University, Suwon 16419, Korea.
| | - Ju-Mi Hong
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
| | - Trang Thu Thi Nguyen
- Department of Pharmacological Medical and Agronomical Biotechnology, University of Science and Technology of Hanoi, Hanoi 100000, Vietnam.
| | - Se Jong Han
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
- Department of Polar Life Science, University of Science and Technology, Incheon 21990, Korea.
| | - Ui Joung Youn
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
| | - Joung Han Yim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
- Department of Polar Life Science, University of Science and Technology, Incheon 21990, Korea.
| | - Il-Chan Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Korea.
- Department of Polar Life Science, University of Science and Technology, Incheon 21990, Korea.
| |
Collapse
|
8
|
Cheng MF, Lin CS, Chen YH, Sung PJ, Lin SR, Tong YW, Weng CF. Inhibitory Growth of Oral Squamous Cell Carcinoma Cancer via Bacterial Prodigiosin. Mar Drugs 2017; 15:md15070224. [PMID: 28714874 PMCID: PMC5532666 DOI: 10.3390/md15070224] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/02/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy drugs for oral cancers always cause side effects and adverse effects. Currently natural sources and herbs are being searched for treated human oral squamous carcinoma cells (OSCC) in an effort to alleviate the causations of agents in oral cancers chemotherapy. This study investigates the effect of prodigiosin (PG), an alkaloid and natural red pigment as a secondary metabolite of Serratia marcescens, to inhibit human oral squamous carcinoma cell growth; thereby, developing a new drug for the treatment of oral cancer. In vitro cultured human OSCC models (OECM1 and SAS cell lines) were used to test the inhibitory growth of PG via cell cytotoxic effects (MTT assay), cell cycle analysis, and Western blotting. PG under various concentrations and time courses were shown to effectively cause cell death and cell-cycle arrest in OECM1 and SAS cells. Additionally, PG induced autophagic cell death in OECM1 and SAS cells by LC3-mediated P62/LC3-I/LC3-II pathway at the in vitro level. These findings elucidate the role of PG, which may target the autophagic cell death pathways as a potential agent in cancer therapeutics.
Collapse
Affiliation(s)
- Ming-Fang Cheng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 10086, Taiwan.
- Division of Histology and Clinical Pathology, Hualian Army Forces General Hospital, Hualien 97144, Taiwan.
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei 10086, Taiwan.
| | - Yu-Hsin Chen
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 94450, Taiwan.
| | - Ping-Jyun Sung
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 94450, Taiwan.
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan.
| | - Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Yi-Wen Tong
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 94450, Taiwan.
| |
Collapse
|
9
|
Park HJ, Jang YJ, Yim JH, Lee HK, Pyo S. Ramalin Isolated from Ramalina Terebrata Attenuates Atopic Dermatitis-like Skin Lesions in Balb/c Mice and Cutaneous Immune Responses in Keratinocytes and Mast Cells. Phytother Res 2016; 30:1978-1987. [PMID: 27558640 DOI: 10.1002/ptr.5703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/18/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that involves eczematous skin lesions with pruritic erythematous papules. In this study, we investigated the mitigating effects of ramalin, a component of the Antarctic lichen Ramalina terebrata against AD in vivo and in vitro. Oral administration of ramalin lessened scratching behaviors and significantly reduced both serum immunoglobulin E and IL-4 levels, and mRNA levels of IL-4 and IL-10 in AD-induced Balb/c mice. In vitro, treatment with ramalin produced significantly less inflammatory chemokines and cytokines, including TARC, MCP-1, RANTES, and IL-8 in TNF-α-stimulated HaCaT cells. In addition, ramalin inhibited the activation of nuclear factor-kappa B as well as the phosphorylation of mitogen-activated protein kinases (MAPK). Furthermore, ramalin treatment resulted in decreased production of β-hexosaminidase and proinflammatory cytokines IL-4, IL-6, and TNF-α in 2,4 dinitrophenyl-human serum albumin-stimulated RBL-2H3 cells through blocking MAPK signaling pathways. The results suggest that ramalin modulates the production of immune mediators by inhibiting the nuclear factor-kappa B and MAPK signaling pathways. Taken together, ramalin effectively attenuated the development of AD and promoted the mitigating effects on Th2 cell-mediated immune responses and the production of inflammatory mediators in mast cells and keratinocytes. Thus, ramalin may be a potential therapeutic agent for AD. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hye-Jin Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
| | - Yeon Jeong Jang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
| | - Joung-Han Yim
- Polar BioCenter, Korea Polar Research Institute, Incheon, 21990, Korea
| | - Hong-Kum Lee
- Polar BioCenter, Korea Polar Research Institute, Incheon, 21990, Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
| |
Collapse
|
10
|
Biological Effect of a Hybrid Anticancer Agent Based on Kinase and Histone Deacetylase Inhibitors on Triple-Negative (MDA-MB231) Breast Cancer Cells. Int J Mol Sci 2016; 17:ijms17081235. [PMID: 27483253 PMCID: PMC5000633 DOI: 10.3390/ijms17081235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/20/2016] [Accepted: 07/26/2016] [Indexed: 01/27/2023] Open
Abstract
We examined the effects of the histone deacetylase inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA) combined with the vascular endothelial growth factor receptor-1/2 inhibitor (3Z)-5-hydroxy-3-(1H-pyrrol-2-ylmethylidene)-2,3-dihydro-1H-indol-2-one on MDA-MB-231 breast cancer cells (triple-negative) in the form of both a cocktail of the separate compounds and a chemically synthesized hybrid (N-hydroxy-N'-[(3Z)-2-oxo-3-(1H-pyrrol-2-ylmethylidene)-2,3-dihydro-1H-indol-5-yl]octanediamide). Comparative flow cytometric and Western blot analyses were performed on cocktail- and hybrid-treated cells to evaluate cell cycle distribution, autophagy/apoptosis modulation, and mitochondrial metabolic state in order to understand the cellular basis of the cytotoxic effect. Cell cycle analysis showed a perturbation of the rate of progression through the cycle, with aspects of redistribution of cells over different cycle phases for the two treatments. In addition, the results suggest that the two distinct classes of compounds under investigation could induce cell death by different preferential pathways, i.e., autophagy inhibition (the cocktail) or apoptosis promotion (the hybrid), thus confirming the enhanced potential of the hybrid approach vs. the combination approach in finely tuning the biological activities of target cells and also showing the hybrid compound as an additional promising drug-like molecule for the prevention or therapy of “aggressive” breast carcinoma.
Collapse
|