1
|
Hao P, Zhang C, Bian H, Li Y. The mechanism of action of myricetin against lung adenocarcinoma based on bioinformatics, in silico and in vitro experiments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4089-4104. [PMID: 38015259 DOI: 10.1007/s00210-023-02859-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Myricetin is a natural flavonoid with anti-cancer and anti-inflammatory effects, but its mechanism for treating lung adenocarcinoma (LUAD) remains unclearly. Therefore, bioinformatics, in silico and in vitro experiments were employed to elucidate this issue in this study. The core targets of myricetin against LUAD were screened by PharmaMapper (v2017), Assistant for Clinical Bioinformatics, STRING (v11.5) and Cytoscape (v3.8.1). Using Kaplan-Meier Plotter (v2022.04.20), UALCAN (v2021.12.13) and GEPIA (v2.0) databases, the correlation between core genes and the prognosis of LUAD patients were analyzed, and the expression levels of core genes were verified. In silico studies were used to analyze the binding energies and sites of myricetin with core genes. The effects of myricetin on H1975 cells were explored through thiazolyl blue (MTT), cell migration, colony formation and western blot assays. A total of 72 potential targets of myricetin against LUAD were identified through bioinformatics. Among the four core targets obtained by multiple networks and in silico assays, the up-regulated MMP9 (HR = 1.14 (1-1.29), logrank P = 0.046) and down-regulated PIK3R1 (HR = 0.58 (0.51-0.66), logrank P < 1E-16) were positively correlated with poor survival outcomes in LUAD patients. In vitro experiments demonstrated that myricetin inhibited the proliferation and migration of H1975 cells, promoting their apoptosis. Myricetin inhibits the proliferation of H1975 cells and induces cell apoptosis through its influence on the expression levels of MMP1, MMP3, MMP9, and PIK3R1 and regulating the multiple pathways these genes participate in. Both MMP9 and PIK3R1 are potential biomarkers for LUAD.
Collapse
Affiliation(s)
- Pengfei Hao
- Nanyang Institute of Technology, Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang, 473000, China
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Chinese Materia Medica and Prepared Slices), Zhengzhou, 450000, China
| | - Chaoyun Zhang
- Nanyang Institute of Technology, Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang, 473000, China
| | - Hua Bian
- Nanyang Institute of Technology, Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang, 473000, China
| | - Yixian Li
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Chinese Materia Medica and Prepared Slices), Zhengzhou, 450000, China.
| |
Collapse
|
2
|
Yan CF, Xia J, Qun WS, Bing WY, Guo WJ, Yong HG, Sheng SJ, Lei ZG. Tumor-associated macrophages-derived exo-let-7a promotes osteosarcoma metastasis via targeting C15orf41 in osteosarcoma. ENVIRONMENTAL TOXICOLOGY 2023; 38:1318-1331. [PMID: 36919336 DOI: 10.1002/tox.23766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Osteosarcoma (OS) immune environment is complexed and the immune factors-related to OS progression need to be explored. Tumor-associated macrophages (TAMs) are regarded as immune suppressive and tumor-promoting cells. However, the underlying mechanisms through which TAMs function are still fragmentary. Here, we aim to explore the underlying mechanisms by which TAMs regulate OS progression. METHODS TAMs from OS tissues were isolated by flow cytometry. Exosomes derived from TAMs were separated using ultracentrifugation and western blotting. Transmission electron microscopy (TEM), and flow cytometry were constructed to characterize TAMs-derived exosomes. Additionally, the differential MicroRNAs (miRNAs) and genes were detected through RNA sequencing, and further validated using real-time PCR (RT-PCR). OS cell metastasis ability was assessed using transwell invasion and scratch wound healing assays. MiRNAs mimic and lentiviral vectors were utilized to explore the effects on OS progression. RESULTS Exosome secreted by TAMs accelerated the OS metastasis. Let-7a level was upregulated in TAMs derived exosomes, which downregulated C15orf41 by targeting 3'-untranslated region (UTR). Furthermore, overexpressing let-7a enhanced invasion and migration by blocking the transcription of C15orf41. In consistent, up-regulating let-7a promoted OS progression and made the prognosis to be worse, which can be reversed by C15orf41 overexpression. CONCLUSION This study highlighted the critical role of TAMs-derived exosomes in OS progression and explored the potential value of the let-7a/C15orf41 axis as an indicator or target for OS.
Collapse
Affiliation(s)
- Chen-Fei Yan
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Wang-Si Qun
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei-Yi Bing
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Wu-Jian Guo
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Huang-Gang Yong
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Jing Sheng
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhao-Guang Lei
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Pourali G, Kazemi D, Pourali R, Rahmani N, Razzaghi E, Maftooh M, Fiuji H, Ghorbani E, Khazaei M, Ferns GA, Hassanian SM, Avan A. Bioactive Peptides: Potential Impact on the Treatment of Gastrointestinal Cancers. Curr Pharm Des 2023; 29:2450-2460. [PMID: 37877510 DOI: 10.2174/0113816128261378231019201709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 10/26/2023]
Abstract
We have reviewed the potential use of bioactive peptides in the treatment of gastrointestinal (GI) malignancies, which are a significant cause of morbidity and mortality globally. Conventional therapies, such as surgery, chemotherapy, and radiotherapy, are associated with numerous side effects that may lead to longterm complications. Bioactive peptides are short-chain amino acids that can be extracted from natural sources or synthesized, and they have various potential health benefits, including anti-inflammatory, anti-hypertensive, antioxidant, antimicrobial, and anti-cancer properties. Bioactive peptides can be acquired from animal or plant sources, and can be classified based on their function, such as ACE-inhibiting, antimicrobial, and electrolyte- regulating peptides. Recent studies have demonstrated the promising role of bioactive peptides in tumor suppression, especially when combined with conventional therapies. In this study, we have reviewed the beneficial properties of bioactive peptides and their role in suppressing tumor activity. The mechanisms of bioactive peptides in tumor suppression are discussed. We have further reviewed the findings of preclinical and clinical studies that have investigated the application of bioactive peptides in the treatment of GI cancers. This review highlights the potential use of bioactive peptides as a promising treatment method for GI malignancies to increase the quality of life of GI cancer patients.
Collapse
Affiliation(s)
- Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Danial Kazemi
- School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan, Iran
| | - Roozbeh Pourali
- Student Research Committee, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nafise Rahmani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Erfan Razzaghi
- School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib Street, Isfahan, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Fiuji
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Field EK, Hartzheim A, Terry J, Dawson G, Haydt N, Neuman-Lee LA. Reptilian Innate Immunology and Ecoimmunology: What Do We Know and Where Are We Going? Integr Comp Biol 2022; 62:1557-1571. [PMID: 35833292 DOI: 10.1093/icb/icac116] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/05/2023] Open
Abstract
Reptiles, the only ectothermic amniotes, employ a wide variety of physiological adaptations to adjust to their environments but remain vastly understudied in the field of immunology and ecoimmunology in comparison to other vertebrate taxa. To address this knowledge gap, we assessed the current state of research on reptilian innate immunology by conducting an extensive literature search of peer-reviewed articles published across the four orders of Reptilia (Crocodilia, Testudines, Squamata, and Rhynchocephalia). Using our compiled dataset, we investigated common techniques, characterization of immune components, differences in findings and type of research among the four orders, and immune responses to ecological and life-history variables. We found that there are differences in the types of questions asked and approaches used for each of these reptilian orders. The different conceptual frameworks applied to each group has led to a lack of unified understanding of reptilian immunological strategies, which, in turn, have resulted in large conceptual gaps in the field of ecoimmunology as a whole. To apply ecoimmunological concepts and techniques most effectively to reptiles, we must combine traditional immunological studies with ecoimmunological studies to continue to identify, characterize, and describe the reptilian immune components and responses. This review highlights the advances and gaps that remain to help identify targeted and cohesive approaches for future research in reptilian ecoimmunological studies.
Collapse
Affiliation(s)
- Emily K Field
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Alyssa Hartzheim
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Jennifer Terry
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Grant Dawson
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Natalie Haydt
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Lorin A Neuman-Lee
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| |
Collapse
|
5
|
Reptiles as Promising Sources of Medicinal Natural Products for Cancer Therapeutic Drugs. Pharmaceutics 2022; 14:pharmaceutics14040874. [PMID: 35456708 PMCID: PMC9025323 DOI: 10.3390/pharmaceutics14040874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/14/2022] Open
Abstract
Natural products have historically played an important role as a source of therapeutic drugs for various diseases, and the development of medicinal natural products is still a field with high potential. Although diverse drugs have been developed for incurable diseases for several decades, discovering safe and efficient anticancer drugs remains a formidable challenge. Reptiles, as one source of Asian traditional medicines, are known to possess anticancer properties and have been used for a long time without a clarified scientific background. Recently, it has been reported that extracts, crude peptides, sera, and venom isolated from reptiles could effectively inhibit the survival and proliferation of various cancer cells. In this article, we summarize recent studies applying ingredients derived from reptiles in cancer therapy and discuss the difficulties and prospective development of natural product research.
Collapse
|
6
|
Chook CYB, Chen FM, Leung FP, Chen ZY, Wong WT. Potential of crocodile blood as a medication and dietary supplement: A systemic review. Clin Exp Pharmacol Physiol 2021; 48:1043-1058. [PMID: 33987869 DOI: 10.1111/1440-1681.13524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
Crocodile blood has long been used as a traditional medicine in many Asian countries to treat diseases such as asthma, allergies, and many others. Yet, only recently has the safety and effectiveness of using crocodile blood as a medicine been examined using modern scientific methods; with both conserved and novel active components identified from crocodile blood. Further in vitro and in vivo investigations found that crocodile blood can have a wide range of beneficial effects, including antimicrobial, antiviral, anti-oxidative, anti-inflammatory, antitumour effects, anti-anaemia, and enhancement of wound healing. A systematic research of literature published in English-language journals up to April 2020 was conducted in PubMed, Google Scholar, and Web of Science. Based on the biological and chemical knowledge of crocodile immunity and crocodile blood, this article aims to: provide a critical review on the proposed properties of crocodile blood, identify the knowledge gap and offer some insights for future investigations regarding the use of crocodile blood as a medication or dietary supplement.
Collapse
Affiliation(s)
- Chui Yiu Bamboo Chook
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis M Chen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Fung Ping Leung
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhen-Yu Chen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Tak Wong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
7
|
Maraming P, Klaynongsruang S, Boonsiri P, Peng SF, Daduang S, Rungsa P, Tavichakorntrakool R, Chung JG, Daduang J. Anti-metastatic Effects of Cationic KT2 Peptide (a Lysine/Tryptophan-rich Peptide) on Human Melanoma A375.S2 Cells. In Vivo 2021; 35:215-227. [PMID: 33402468 DOI: 10.21873/invivo.12250] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM KT2 is a lysine/tryptophan-rich peptide modified from Crocodylus siamensis Leucrocin I. In this study, we examined the cell toxicity, cellular uptake, anti-migration and anti-invasion activities of KT2 in A375.S2 human melanoma cells. MATERIALS AND METHODS A375.S2 cells were treated with KT2 peptide and then we performed MTT assay, study of cellular uptake by a confocal microscope, wound healing assay, transwell migration/invasion assay, and evaluation of the expression of metastasis-associated proteins. RESULTS KT2 can be internalized through the plasma membrane and can slightly alter cell morphology, decrease the percentage of viable cells and inhibit cell migration and invasion of A375.S2 cells in a dose-dependent manner. This peptide suppressed MMP-2 activity, as measured by gelatine zymography assay. The protein level of MMP-2 was decreased by KT2. KT2 also down-regulated metastasis pathway-related molecules, including FAK, RhoA, ROCK1, GRB2, SOS-1, p-JNK, p-c-Jun, PI3K, p-AKT (Thr308), p-AKT (Ser473), p-p38, MMP-9, NF-kB, and uPA. CONCLUSION These results indicate that KT2 inhibits the migration and invasion of human melanoma cells by decreasing MMP-2 and MMP-9 expression through inhibition of FAK, uPA, MAPK, PI3K/AKT NF-kB, and RhoA-ROCK signalling pathways. These findings suggest that KT2 deserves further investigation as an anti-metastatic agent for human melanoma.
Collapse
Affiliation(s)
- Pornsuda Maraming
- Biomedical Science Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Sompong Klaynongsruang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen, Thailand
| | - Patcharee Boonsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Sakda Daduang
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Prapenpuksiri Rungsa
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Ratree Tavichakorntrakool
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.;
| | - Jureerut Daduang
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
8
|
Jeyamogan S, Khan NA, Sagathevan K, Siddiqui R. Crocodylus porosus: a potential source of anticancer molecules. BMJ OPEN SCIENCE 2020; 4:e100040. [PMID: 35047686 PMCID: PMC8749261 DOI: 10.1136/bmjos-2019-100040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/09/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Background Cancer remains a global threat resulting in significant morbidity and mortality despite advances in therapeutic interventions, suggesting urgency for identification of anticancer agents. Crocodiles thrive in polluted habitat, feed on germ-infested meat, are exposed to carcinogenic heavy metals, are the very few species to survive the catastrophic Cretaceous–Paleogene extinction event, yet have a prolonged lifespan and rarely been reported to develop cancer. Therefore, we hypothesised that animals living in polluted environments such as crocodiles possess anticancer molecules/mechanisms. Methods Crocodylus porosus was procured, blood collected, dissected and lysates prepared from internal organs. Organ lysates and sera were tested for growth inhibition, cytotoxic effects and cell survival against HeLa, PC3 and MCF7 cells and subjected to liquid chromatography mass spectrometry. RNA transcriptome analysis and differential gene analysis were performed using Galaxy Bioinformatics. Results Sera exhibited potent growth inhibition and cytotoxic effects against cancer cells. 80 molecules were detected from C. porosus and 19 molecules were putatively identified. Additionally, more than 100 potential anticancer peptides were identified from sera using bioinformatics based on peptide amino acid composition, binary profile, dipeptide composition and pseudo-amino acid composition. Following transcriptome analysis, 14 genes in treated HeLa cells, 51 genes in treated MCF7 cells and 2 genes in treated PC3 cells, were found to be expressed, compared with untreated controls. Conclusion Animals residing in polluted milieus are an unexploited source for prospective pharmaceutical drugs, and could lead to identification of novel antitumour compound(s) and/or further understanding of the mechanisms of cancer resistance.
Collapse
Affiliation(s)
- Shareni Jeyamogan
- Department of Biological Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - Naveed Ahmed Khan
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| | - K Sagathevan
- Science and Technology, Sunway College, Bandar Sunway, Selangor, Malaysia
| | - Ruqaiyyah Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| |
Collapse
|
9
|
Maraming P, Klaynongsruang S, Boonsiri P, Maijaroen S, Daduang S, Chung JG, Daduang J. Antitumor activity of RT2 peptide derived from crocodile leukocyte peptide on human colon cancer xenografts in nude mice. ENVIRONMENTAL TOXICOLOGY 2018; 33:972-977. [PMID: 30019842 DOI: 10.1002/tox.22584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
RT2, derived from the leukocyte peptide of Crocodylus siamensis, can kill human cervical cancer cells via apoptosis induction, but no evidence has shown in vivo. In this study, we investigated the antitumor effect of RT2 on human colon cancer xenografts in nude mice. Twenty-four mice were injected subcutaneously with human colon cancer HCT 116 cells. Eleven days after cancer cell implantation, the mice were treated with intratumoral injections of phosphate buffered saline (PBS) or RT2 (0.01, 0.1, and 1 mg/mouse) once every 2 days for a total of 5 times. The effect of a 10-day intratumoral injection of RT2 on body weight, biochemical, and hematological parameters in BALB/c mice showed no significant difference between the groups. Tumor volume showed a significant decrease only in the treatment group with RT2 (1 mg/mouse) at day 6 (P < .05), day 8 (P < .01), and day 10 (P < .01) after the first treatment. The protein expression levels of cleaved poly (ADP-ribose) polymerase (PARP), apoptosis-inducing factor (AIF), and the p53 tumor suppressor protein (p53) in xenograft tumors increased after treatment with RT2 (1 mg/mouse) compared to those in the PBS-injected group. Moreover, RT2 increased the expression of Endo G and Bcl-2 family proteins. Therefore, the peptide RT2 can inhibit tumor growth via the induction of apoptosis in an in vivo xenograft model.
Collapse
Affiliation(s)
- Pornsuda Maraming
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Sompong Klaynongsruang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Patcharee Boonsiri
- Faculty of Medicine, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Surachai Maijaroen
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Sakda Daduang
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Science, Khon Kaen University, Khon Kaen, Thailand
| | - Jing-Gung Chung
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Jureerut Daduang
- Center for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
10
|
Huang RZ, Jin L, Wang CG, Xu XJ, Du Y, Liao N, Ji M, Liao ZX, Wang HS. A pentacyclic triterpene derivative possessing polyhydroxyl ring A suppresses growth of HeLa cells by reactive oxygen species-dependent NF-κB pathway. Eur J Pharmacol 2018; 838:157-169. [PMID: 30153443 DOI: 10.1016/j.ejphar.2018.08.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Pentacyclic triterpene derivatives possessing polyhydroxyl ring A exhibit many important pharmacological activities. (1β, 2α, 3β, 19β, 23)-1,2,3,19,23-pentahydroxyolean-12-en-28-oic acid (5), a new bioactive phytochemical with tetra-hydroxyl ring A isolated from Euphorbia sieboldiana in our laboratory, showed potential inhibition effects against several cancer cells previously. This study was performed to investigate the underlying mechanisms of action for its antitumor activity. The results showed that compound 5 inhibited dose-/time-dependently cell growth with low toxicity to normal cells and induced apoptosis in cervical cancer cells. Also, compound 5 inhibited the growth and proliferation of HeLa cells and resulted in G1 phase arrest. Furthermore, exposure of cells to compound 5 caused inactivation of the TNF-α-TAK1-IKK-NF-κB axis and inhibition of TNF-α-stimulated NF-κB activity, followed by down-regulation of NF-κB target genes involved in cell apoptosis (Bcl-2) and in the cell cycle and growth (Cyclin D, c-Myc). Additionally, compound 5 significantly suppressed the migration of HeLa cells. In addition, exposure of HeLa cells to compound 5 decreased the activity of NF-κB through the generation of reactive oxygen species (ROS). Collectively, these results suggested that compound 5 exerted potent anticancer effects on HeLa cells in vitro through targeting the ROS-dependent NF-κB signaling cascade and this compound may be a promising anticancer agent for cancer treatment.
Collapse
Affiliation(s)
- Ri-Zhen Huang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Le Jin
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Chun-Gu Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, PR China
| | - Xiao-Jing Xu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ying Du
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Na Liao
- Department of Pharmacy, College of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhi-Xin Liao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| | - Heng-Shan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
11
|
Animals living in polluted environments are a potential source of anti-tumor molecule(s). Cancer Chemother Pharmacol 2017; 80:919-924. [DOI: 10.1007/s00280-017-3410-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/29/2017] [Indexed: 12/20/2022]
|