1
|
Mueed A, Deng Z, Korma SA, Shibli S, Jahangir M. Anticancer potential of flaxseed lignans, their metabolites and synthetic counterparts in relation with molecular targets: current challenges and future perspectives. Food Funct 2023; 14:2286-2303. [PMID: 36820797 DOI: 10.1039/d2fo02208g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lignans are known dietary polyphenols found in cereals, plants and seeds. Flaxseed is one of the major sources of lignans mainly existing in the form of secoisolariciresinol diglucoside (SDG) which can be metabolised by the gut microbes into secoisolariciresinol (SECO) and mammalian lignan (enterodiol and enterolactone) that are easily absorbed through the intestines. Numerous studies reveal that flaxseed lignans (FLs) can be promising chemotherapeutics/chemopreventive agents. Their anticancer activity can occur through the induction of apoptosis, inhibition of cell proliferation, and the hindering of metastasis and angiogenesis. The anti-carcinogenesis of flaxseed lignans is achieved through multiple molecular mechanisms involving biochemical entities such as cellular kinases, cell cycle mediators, transcription factors, inflammatory cytokines, reactive oxygen species, and drug transporters. This review summarizes the bioavailability of FLs, their anticancer mechanisms in relevance to molecular targets, safety, and the scope of future research. Overall, FLs can be utilized in functional foods, dietary supplements, and pharmaceuticals for the management and prevention of cancers.
Collapse
Affiliation(s)
- Abdul Mueed
- State key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Zeyuan Deng
- State key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Sameh A Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, 44519 Zagazig, Egypt
| | - Sahar Shibli
- Food Science Research Institute, National Agriculture Research Center, Islamabad, Pakistan
| | - Muhammad Jahangir
- Department of Food Science & Technology, The University of Haripur, Khyber-Pakhtunkhwa, Pakistan
| |
Collapse
|
2
|
Shahrokhi SZ, Tehrani FSK, Salami S. Molecular mechanisms of bilirubin induced G1 cell cycle arrest and apoptosis in human breast cancer cell lines: involvement of the intrinsic pathway. Mol Biol Rep 2022; 49:10421-10429. [DOI: 10.1007/s11033-022-07757-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/24/2022]
|
3
|
Synthetic Secoisolariciresinol Diglucoside (LGM2605) Prevents Asbestos-Induced Inflammation and Genotoxic Cell Damage in Human Mesothelial Cells. Int J Mol Sci 2022; 23:ijms231710085. [PMID: 36077483 PMCID: PMC9456329 DOI: 10.3390/ijms231710085] [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: 07/28/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Although alveolar macrophages play a critical role in malignant transformation of mesothelial cells following asbestos exposure, inflammatory and oxidative processes continue to occur in the mesothelial cells lining the pleura that may contribute to the carcinogenic process. Malignant transformation of mesothelial cells following asbestos exposure occurs over several decades; however, amelioration of DNA damage, inflammation, and cell injury may impede the carcinogenic process. We have shown in an in vitro model of asbestos-induced macrophage activation that synthetic secoisolariciresinol diglucoside (LGM2605), given preventively, reduced inflammatory cascades and oxidative/nitrosative cell damage. Therefore, it was hypothesized that LGM2605 could also be effective in reducing asbestos-induced activation and the damage of pleural mesothelial cells. LGM2605 treatment (50 µM) of huma n pleural mesothelial cells was initiated 4 h prior to exposure to asbestos (crocidolite, 20 µg/cm2). Supernatant and cells were evaluated at 0, 2, 4, and 8 h post asbestos exposure for reactive oxygen species (ROS) generation, DNA damage (oxidized guanine), inflammasome activation (caspase-1 activity) and associated pro-inflammatory cytokine release (IL-1β, IL-18, IL-6, TNFα, and HMGB1), and markers of oxidative stress (malondialdehyde (MDA) and 8-iso-prostaglandin F2a (8-iso-PGF2α). Asbestos induced a time-dependent ROS increase that was significantly (p < 0.0001) reduced (29.4%) by LGM2605 treatment. LGM2605 pretreatment also reduced levels of asbestos-induced DNA damage by 73.6% ± 1.0%. Although levels of inflammasome-activated cytokines, IL-1β and IL-18, reached 29.2 pg/mL ± 0.7 pg/mL and 43.9 pg/mL ± 0.8 pg/mL, respectively, LGM2605 treatment significantly (p < 0.0001) reduced cytokine levels comparable to baseline (non-asbestos exposed) values (3.8 pg/mL ± 0.2 pg/mL and 5.4 pg/mL ± 0.2 pg/mL, respectively). Furthermore, levels of IL-6 and TNFα in asbestos-exposed mesothelial cells were high (289.1 pg/mL ± 2.9 pg/mL and 511.3 pg/mL ± 10.2 pg/mL, respectively), while remaining undetectable with LGM2605 pretreatment. HMGB1 (a key inflammatory mediator and initiator of malignant transformation) release was reduced 75.3% ± 0.4% by LGM2605. Levels of MDA and 8-iso-PGF2α, markers of oxidative cell injury, were significantly (p < 0.001) reduced by 80.5% ± 0.1% and 76.6% ± 0.3%, respectively. LGM2605, given preventively, reduced ROS generation, DNA damage, and inflammasome-activated cytokine release and key inflammatory mediators implicated in asbestos-induced malignant transformation of normal mesothelial cells.
Collapse
|
4
|
Chen T, Wang Z, Zhong J, Zhang L, Zhang H, Zhang D, Xu X, Zhong X, Wang J, Li H. Secoisolariciresinol diglucoside induces pyroptosis by activating caspase-1 to cleave GSDMD in colorectal cancer cells. Drug Dev Res 2022; 83:1152-1166. [PMID: 35472101 PMCID: PMC9543314 DOI: 10.1002/ddr.21939] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/05/2022]
Abstract
Secoisolariciresinol diglucoside (SDG) is the main component of lignans with various biological activities, including anticancer activity. However, whether SDG has obvious anticancer effects on colorectal cancer (CRC) is unclear. Pyroptosis, a form of programmed cell death, has received increasing attention in cancer‐related research. In this study, we aimed to test the anticancer properties and relatecd functional mechanisms of SDG. we found that SDG not only inhibited the cell viability of HCT116 cells, but also induced HCT116 cells to swell with apparent large bubbles, which are typical signs of pyroptosis. Furthermore, SDG induced cell pyroptosis by enhancing cleavage of the N‐terminal fragment of gasdermin D (GSDMD) in CRC cells, accompanied by increased caspase‐1 cleavage. Consistent with this, SDG‐induced GSDMD‐N‐terminal fragment cleavage and pyroptosis were reduced by siRNA‐mediated silencing of caspase‐1 or treatment with the specific caspase‐1 inhibitor VX‐765 treatment, suggesting that active caspase‐1 further induces pyroptosis. A mechanistic study showed that SDG induced reactive oxygen species (ROS) accumulation and inhibits phosphatidylinositol 3‐kinase (PI3K) phosphorylation and increases pyroptosis, while increasing GSDMD and caspase‐1 cleavage and enhancing expression of BCL2‐associated X (BAX), which could be rescued by the ROS scavenger (NAC), suggesting that SDG‐induced GSDME‐dependent pyroptosis is related to the ROS/PI3K/AKT/BAX‐mitochondrial apoptotic pathway. In vivo results showed that SDG significantly inhibited tumor growth and induced pyroptosis in the HCT116‐CRC nude mouse model. In conclusion, our findings suggest that the anticancer activity of SDG in CRC is associated with the induction of GSDMD‐dependent pyroptosis by SDG through the generation of ROS/P13K/AKT/BAK‐mitochondrail apoptosis pathway, providing insights into SDG in its potential new application in cancer treatment.
Collapse
Affiliation(s)
- Tuo Chen
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong, Sichuan, China.,Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhen Wang
- Department of Gastroenterology, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Junbo Zhong
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Lijun Zhang
- Respiratory and Critical Care Medicine, Yanzhou Branch of Affiliated Hospital of Jining Medical University, Jining, China
| | - Haopeng Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Dayong Zhang
- Department of Urinary Surgery, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Xiaofeng Xu
- Department of Urinary Surgery, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Xianfei Zhong
- Department of Gastroenterology, The People's Hospital of Leshan, Leshan, Sichuan, China
| | - Jing Wang
- Xi'an Medical University School of Stomatolog, Xi'an, Shanxi, China
| | - Hai Li
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
5
|
Wang W, Zhang R, Wang J, Tang J, Wang M, Kuang Y. Antitumour Activity of Muricatacin Isomers and its Derivatives in Human Colorectal Carcinoma Cell HCT116. Anticancer Agents Med Chem 2021; 20:254-263. [PMID: 31729294 DOI: 10.2174/1871520619666191115111032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/15/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND PURPOSE Colorectal cancer is one of the leading causes of cancer-related death in elderly people. The natural product muricatacin is an important member of the γ-lactone family, and it has exhibited antitumour activity in multiple cancer cell lines; however, the antitumour activities of muricatacin stereoisomers and their derivatives in colorectal cancer cells have not yet been systematically explored. METHODS The colorectal carcinoma cell line HCT116 was investigated in this study. Cell proliferation was assessed by MTT assay or crystal violet staining. Cell cycle arrest and cell apoptosis were evaluated by flow cytometry assay. The expression levels of p53, p21, cyclin E, cyclin D1, caspase-3, cleaved caspase-3, caspase-9, cleaved caspase-9 and LC3B were measured using western blot analysis. Autophagy induced by M2 was monitored by immunofluorescence assay with an antibody against LC3B. RESULTS Cell proliferation assays showed that both naturally occurring muricatacin (M4) and its synthetic stereoisomer (M2) are potent cell growth inhibitors in HCT116 cells, with IC50 values of 79.43 and 83.17μM, respectively; these values are much lower than those of the other two isomers, M1 and M3, and those of the sixmembered lactone analogues. The flow cytometry analysis revealed that M2 and M4 induced significant cell cycle arrest during G0/G1 phase and caused relatively low apoptosis rates in HCT116 cells. Further analysis indicated that M2 caused p53-independent p21 induction and cyclin E/cyclin D1 downregulation. In addition, M2 also markedly induced autophagy in the early stage of administration. CONCLUSION Our results suggested that muricatacins possess potent antitumour activity against the colorectal carcinoma cell line HCT116 through inducing G0/G1 phase cell cycle arrest and autophagy in the early stage of administration.
Collapse
Affiliation(s)
- Wencong Wang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Rui Zhang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jinxing Wang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jun Tang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Mingan Wang
- Department of Applied Chemistry, College of Sciences, China Agricultural University, Beijing, 100193, China
| | - Yu Kuang
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
6
|
Li D, Liu Z, Ning G. [Expression of CDC25A in non-small cell lung cancer and its relationship with let-7 gene]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1622-1627. [PMID: 33243735 DOI: 10.12122/j.issn.1673-4254.2020.11.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the expression of CDC25A in non- small cell lung cancer (NSCLC) tissues and explore its correlation with the clinicpathological features of the patients and the expressions of let-7a1 and let-7c. METHODS We collected surgical specimens of pathologically confirmed NSCLC tissues and paired adjacent lung tissues from 44 patients and tissues of benign lung lesions from 9 patients. The expressions of CDC25A protein and mRNA in the tissues were detected by immunohistochemistry and fluorescence quantitative RT-PCR, respectively; the expressions of let-7a1 and let-7c mRNA were detected using tail-adding fluorescence quantitative RT-PCR. RESULTS The positivity rate of CDC25A protein expression was significantly higher in NSCLC tissues than in the adjacent tissues and benign pulmonary lesions (P < 0.05). CDC25A protein expression in NSCLC was not correlated with the patients' age, gender, pathological type, degree of tumor differentiation, or clinical stages (P > 0.05), and was significantly correlated with smoking and lymph node metastasis (P < 0.05). CDC25A mRNA expression was also significantly higher in NSCLC tissues than in the adjacent tissues and benign pulmonary lesions (F=6.33, P < 0.05), and was similar between the latter two tissues (P > 0.05). Pearson correlation analysis showed that CDC25A expression had a significant negative correlation with let-7c expression in both NSCLC tissues (r=-0.42) and adjacent lung tissues (r=-0.40) but was not correlated with let-7a1 expression. CONCLUSIONS The expression level of CDC25A is significantly increased in NSCLC with a negative correlation with Let-7c expression, which identifies CDC25A as a possible downstream target gene of Let-7c.
Collapse
Affiliation(s)
- Dianming Li
- Department of Respiratory and Critical Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Zhaofei Liu
- Department of Respiratory Medicine, Linquan County People's Hospital, Linquan 236400, China
| | - Guolan Ning
- Department of Respiratory and Critical Care Medicine, Fuyang Second People's Hospital, Fuyang 236000, China
| |
Collapse
|
7
|
Pei XD, Yao HL, Shen LQ, Yang Y, Lu L, Xiao JS, Wang XY, He ZL, Jiang LH. α-Cyperone inhibits the proliferation of human cervical cancer HeLa cells via ROS-mediated PI3K/Akt/mTOR signaling pathway. Eur J Pharmacol 2020; 883:173355. [PMID: 32687921 DOI: 10.1016/j.ejphar.2020.173355] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 01/06/2023]
Abstract
Cervical cancer is the fourth leading killer of female cancer patients worldwide. Each year more than half a million women are diagnosed with cervical cancer and the disease results in over 300, 000 deaths. α-Cyperone is known as the principal active ingredient in the Cyperus rotundus (Family: Cyperaceae). However, the effects of α-Cyperone on cancers, especially on cervical cancer, are yet to be explored. In the present study, the underlying mechanism of the anti-tumor activity of α-Cyperone against HeLa cells was investigated. The results showed that α-Cyperone inhibited proliferation and induced apoptosis in HeLa cells. Mechanistically, α-Cyperone promoted HeLa cells apoptosis via a mitochondrial apoptotic pathway, which was proved by increased level of intracellular reactive oxygen species (ROS) and upregulated expression of cytochrome c, cleaved caspase-3, PARP, and Bax. Further RNA-sequencing revealed α-Cyperone inhibited the activation of PI3K/Akt/mTOR signaling pathway in HeLa cells, which confirmed by PI3K inhibitor and agonist. The PI3K inhibitor (LY294002) synergized with α-Cyperone in arresting the growth of HeLa cells, whereas the PI3K agonist (IGF-1) abrogated such an effect. Interestingly, the expression of PD-L1 was attenuated by both α-Cyperone and LY294002, while the supplement of IGF-1 rescued the low expression of PD-L1. In conclusion, our results reveal that the inhibitory effect of α-Cyperone on HeLa cell growth is triggered via the ROS-mediated PI3K/Akt/mTOR signaling pathway and closely related to a decline in the PD-L1 expression.
Collapse
Affiliation(s)
- Xiao-Dong Pei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563100, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Hong-Liang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Drug Synthesis and Evaluation Center, Guangdong Institute of Applied Biological Resources, Guangdong, PR China
| | - Li-Qun Shen
- Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi University for Nationalities, Nanning, 530006, PR China
| | - Yang Yang
- Department of Pharmacology, Guilin Medical University, Guilin, Guangxi, 541000, PR China
| | - Lan Lu
- Sichuan Industrial Institute of Antibiotics, Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Chengdu University, 610106, Chengdu, PR China
| | - Jun-Song Xiao
- Beijing Higher Institution Engineering Research Center of Food Additives and Ingredients, Beijing Technology and Business University-BTBU, Beijing, 100048, PR China
| | - Xin-Yu Wang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Zhi-Long He
- College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Li-He Jiang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563100, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China; Medical College, Guangxi University, Nanning, 530004, PR China; School of Basic Medical Science, YouJiang Medical University for Nationaties, No. 98 Chengxiang Road, Baise, Guangxi, 533000, PR China.
| |
Collapse
|
8
|
Arsenic trioxide and BIBR1532 synergistically inhibit breast cancer cell proliferation through attenuation of NF-κB signaling pathway. Life Sci 2020; 257:118060. [PMID: 32645343 DOI: 10.1016/j.lfs.2020.118060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/27/2020] [Accepted: 07/03/2020] [Indexed: 12/29/2022]
Abstract
AIMS Despite the remarkable anti-proliferative effects of Arsenic trioxide (ATO) in breast cancer cells, the requirement of high, toxic concentrations to induce apoptosis may cause serious side effects in patients. In the present study, we aimed to use BIBR1532, an hTERT inhibitor, in combination with ATO to sensitize MCF7 and MDA-231 cells to lower concentrations of ATO. MAIN METHODS Breast cancer cell lines MCF7 and MDA-231 were cultured and treated with different doses of ATO and BIBR1532 for 48 h and its effects on cell survival and proliferation were analyzed by MTT, crystal violet staining, colony formation assay, cell cycle, AnnexinV/PI and Real-time PCR tests. KEY FINDINGS ATO and BIBR1532 synergistically inhibited proliferation and colony-forming ability of breast cancer cells. Besides, BIBR1532 augmented ATO-induced cytotoxic effects via triggering G1 cell cycle arrest and induction of apoptosis coupled with the down-regulation of NF-κB target genes that were involved in cell cycle progression (e.g. CCND1 and CDK6) and prevention of apoptosis such as Bcl-2, Bcl-xl, c-IAP2, and Survivin Respectively. Moreover, ATO-BIBR1532 significantly reduced the mRNA expression level of RELA, NFKB1, and several validated target genes of the NF-κB signaling pathway including NFKBIA, VEGFC, c-Myc, and hTERT. SIGNIFICANCE The combination of ATO and BIBR1532 synergistically induced its anti-proliferative effect in breast cancer cells by targeting the two key cancer-related pathways, hTERT and NF-κB, and disrupting their feed-forward loop at the same time which result in the reduction of NF-κB transcriptional activity and subsequent down-regulation of its target genes.
Collapse
|
9
|
Umar S, Soni R, Durgapal SD, Soman S, Balakrishnan S. A synthetic coumarin derivative (4-flourophenylacetamide-acetyl coumarin) impedes cell cycle at G0/G1 stage, induces apoptosis, and inhibits metastasis via ROS-mediated p53 and AKT signaling pathways in A549 cells. J Biochem Mol Toxicol 2020; 34:e22553. [PMID: 32578917 DOI: 10.1002/jbt.22553] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/29/2020] [Accepted: 05/15/2020] [Indexed: 01/16/2023]
Abstract
New chemotherapeutic agents with minimum side effects are indispensable to treat non-small-cell lung cancer (NSCLC) since the mortality rate of patients suffering from NSCLC remains high despite receiving conventional medication. In our previous study, many coumarin derivatives were screened for their anticancer properties in A549, an in vitro NSCLC model. One of these, 4-flourophenylacetamide-acetyl coumarin (4-FPAC), induced cytotoxicity at a concentration as low as 0.16 nM. Herein, initially, the cytotoxic potential of 4-FPAC was tested on a noncancerous cell line NIH3T3 and was found safe at the selected dose of 0.16 nM. Further, we investigated the mechanism by which 4-FPAC induced cytotoxicity and arrested the progression of cell cycle as well as metastasis in A549. Results of ethidium bromide/acridine orange (EtBr/AO), 4,6-diamidino-2-phenylindole, comet, and lactate dehydrogenase assays revealed that 4-FPAC caused cytotoxicity via reactive oxygen species-induced p53-mediated mechanism, which involves both extrinsic and intrinsic pathways of apoptosis. Dichlorodihydrofluorescein diacetate, rhodamine 123, and AO staining confirmed the involvement of both mitochondria and lysosome in inducing apoptosis. However, flow cytometric analysis revealed that it causes cell cycle arrest at the G0/G1 phase by modulating p21, CDK2, and CDK4 expression. Aggregation, soft-agar, clonogenic, and scratch assays as well as gene expression analysis collectively confirmed that 4-FPAC minimizes the metastatic property of A549 by downregulating Snail, matrix metalloproteinase 9, and interleukin-8. Additional studies reaffirmed the above findings and substantiated the role of PI3K/AKT in achieving them. The cell-type-specific selective cytostatic and antimetastatic properties shown by 4-FPAC indicate its potential to emerge as a drug of choice against NSCLC in the future.
Collapse
Affiliation(s)
- Shweta Umar
- Department of Zoology, Faculty of Science, The M. S. University of Baroda, Vadodara, India
| | - Rina Soni
- Department of Chemistry, Faculty of Science, The M. S. University of Baroda, Vadodara, India
| | - Sunil D Durgapal
- Department of Chemistry, Faculty of Science, The M. S. University of Baroda, Vadodara, India
| | - Subhangi Soman
- Department of Chemistry, Faculty of Science, The M. S. University of Baroda, Vadodara, India
| | - Suresh Balakrishnan
- Department of Zoology, Faculty of Science, The M. S. University of Baroda, Vadodara, India
| |
Collapse
|
10
|
Kumar M, Martin A, Nirgude S, Chaudhary B, Mondal S, Sarkar A. Quinacrine inhibits GSTA1 activity and induces apoptosis through G 1/S arrest and generation of ROS in human non-small cell lung cancer cell lines. Oncotarget 2020; 11:1603-1617. [PMID: 32405336 PMCID: PMC7210017 DOI: 10.18632/oncotarget.27558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/02/2019] [Indexed: 01/09/2023] Open
Abstract
Background: Quinacrine (QC) is popular for its anti-malarial activity. It has been reported exhibiting anti-cancerous properties by suppressing nuclear factor-κB and activating p53 signaling; however, its effect on cellular pathways in human non-small cell lung cancer (NSCLC) has not been studied. Materials and Methods: Binding of QC with GSTA1 was studied computationally as well as through GST activity assay kit. Cell viability, cell cycle and mitochondrial membrane potential activity were studied using flow cytometry. RT-PCR and western blot were carried out to understand the involvement of various genes at their mRNA as well as protein level. Results: QC inhibited the activity of GSTA1 approximately by 40–45% which inhibits cell survival and promotes apoptosis. QC reduced viability of NSCLC cells in a dose-dependent manner. It also causes nuclear fragmentation, G1/S arrest of cell cycle and ROS generation; which along with disruption of mitochondrial membrane potential activity leads to apoptotic fate. Conclusions: Results revealed, QC has promising anti-cancer potential against NSCLC cells via inhibition of GSTA1, induction of G1/S arrest and ROS mediated apoptotic signaling.
Collapse
Affiliation(s)
- Makhan Kumar
- CMBL, Department of Biological Sciences, CMBL, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa 40372, India
| | - Ansie Martin
- CMBL, Department of Biological Sciences, CMBL, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa 40372, India.,Present Address: UMR 1236, Faculty of Medicine, Rennes 35043, France
| | - Snehal Nirgude
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bangalore, Electronics City Phase 1, Bengaluru, Karnataka 560100, India.,Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Bibha Chaudhary
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bangalore, Electronics City Phase 1, Bengaluru, Karnataka 560100, India
| | - Sukanta Mondal
- CMBL, Department of Biological Sciences, CMBL, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa 40372, India
| | - Angshuman Sarkar
- CMBL, Department of Biological Sciences, CMBL, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa 40372, India
| |
Collapse
|
11
|
Khan F, Singh VK, Saeed M, Kausar MA, Ansari IA. Carvacrol Induced Program Cell Death and Cell Cycle Arrest in Androgen-Independent Human Prostate Cancer Cells via Inhibition of Notch Signaling. Anticancer Agents Med Chem 2020; 19:1588-1608. [PMID: 31364516 DOI: 10.2174/1871520619666190731152942] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/09/2019] [Accepted: 07/01/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Several studies have revealed that abnormal activation of Notch signaling is closely related with the development and progression of prostate cancer. Although there are numerous therapeutic strategies, a more effective modality with least side effects is urgently required for the treatment of prostate cancer. Carvacrol is a monoterpenoid phenol and majorly present in the essential oils of Lamiaceae family plants. Many previous reports have shown various biological activities of carvacrol like antioxidant, antiinflammatory and anticancer properties. Recently, we have shown potent anticancer property of carvacrol against prostate cancer cell line DU145. In the current study, we report the chemopreventive and therapeutic potential of carvacrol against another prostate cancer cell line PC-3 with its detailed mechanism of action. METHODS To determine the effect of the carvacrol on prostate cancer cells, the cell viability was estimated by MTT assay and cell death was estimated by LDH release assay. The apoptotic assay was performed by DAPI staining and FITC-Annexin V assay. Reactive Oxygen Species (ROS) was estimated by DCFDA method. Cell cycle analysis was performed by flow cytometry. Gene expression analysis was performed by quantitative real time PCR. RESULTS Our results suggested that the carvacrol treatment significantly reduced the cell viability of PC-3 cells in a dose- and time-dependent manner. The antiproliferative action of carvacrol was correlated with apoptosis which was confirmed by nuclear condensation, FITC-Annexin V assay, modulation in expression of Bax, Bcl-2 and caspase activation. The mechanistic insight into carvacrol-induced apoptosis leads to finding of elevated level of Reactive Oxygen Species (ROS) and mitochondrial membrane potential disruption. Cell cycle analysis revealed that carvacrol prevented cell cycle in G0/G1 that was associated with decline in expression of cyclin D1 and Cyclin-Dependent Kinase 4 (CDK4) and augmented expression of CDK inhibitor p21. Having been said the role of hyperactivation of Notch signaling in prostate cancer, we also deciphered that carvacrol could inhibit Notch signaling in PC-3 cells via downregulation of Notch-1, and Jagged-1. CONCLUSION Thus, our previous and current findings have established the strong potential of carvacrol as a chemopreventive agent against androgen-independent human prostate cancer cells.
Collapse
Affiliation(s)
- Fahad Khan
- Department of Biosciences, Integral University, Dasauli, Kursi Road, Lucknow, 226026, India.,Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, 201306, India
| | - Vipendra K Singh
- Environmental Carcinogenesis Laboratory, Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Mohd A Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Hail, Saudi Arabia
| | - Irfan A Ansari
- Department of Biosciences, Integral University, Dasauli, Kursi Road, Lucknow, 226026, India
| |
Collapse
|
12
|
Piao L, Li H, Feng Y, Li X, Cui Y, Xuan Y. Leucine Zipper-EF-Hand Containing Transmembrane Protein 1 Is a Potential Prognostic Biomarker and Promotes Cell Progression in Prostate Cancer. Cancer Manag Res 2020; 12:1649-1660. [PMID: 32184668 PMCID: PMC7064284 DOI: 10.2147/cmar.s236482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/02/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose The leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) is a mitochondrial protein that has been associated with the occurrence and development of malignant tumors. Previous studies have shown that LETM1 expression is increased in several types of human cancer and is associated with a poor clinical outcome. However, the role of LETM1 in prostate cancer (PCa) has not yet been determined. In this study, we investigated the clinicopathological significance of LETM1 expression and its role in PCa progression. Methods We assessed the expression of LETM1 and genes related to cancer stemness, epithelial-mesenchymal transition (EMT), cell cycle, and PI3K/Akt signaling in 133 paraffin-embedded PCa tissue samples and cancer cells by using immunohistochemistry, immunofluorescence, and Western blotting. Results LETM1 expression was significantly increased in PCa, and it was positively correlated with Gleason score, pathologic tumor (pT) stage, clinical stage, and high microvessel density. Survival analysis showed that patients with PCa with a high level of LETM1 expression exhibited a low overall survival. Cox regression analysis indicated that LETM1 is an independent poor prognostic PCa factor. Additionally, the expression of LETM1 was correlated with cancer cell stemness-associated genes, EMT-related genes, cell cycle regulatory genes, and PI3K/Akt signaling gene expression in PCa. Furthermore, knocking down LETM1 expression down-regulated the expression of stemness-related proteins, while inhibiting tumor spheroid formation, EMT-like changes, cell proliferation, migration, and invasion in PCa cells. Importantly, the PI3K inhibitor LY294002 strongly inhibited the expression of LETM1, pPI3K-p85, and pAkt (Thr308, Ser473) in PCa cells. Conclusion These results indicate that LETM1 expression is associated with cancer cell stemness, promotes EMT-like changes and cell proliferation and is a potential prognostic biomarker for PCa.
Collapse
Affiliation(s)
- Lihua Piao
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China
| | - Haoyue Li
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China.,Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China
| | - Ying Feng
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China.,Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China
| | - Xiaogang Li
- Department of Urology, Yanbian University Affiliated Hospital, Yanji 133002, Jilin Province, People's Republic of China
| | - Yan Cui
- Department of Oncology, Yanbian University Affiliated Hospital, Yanji 133002, Jilin Province, People's Republic of China
| | - Yanhua Xuan
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China.,Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, People's Republic of China
| |
Collapse
|
13
|
Zhang Y, Zhang Y, Xiao Y, Zhong C, Xiao F. Expression of Clusterin suppresses Cr(VI)-induced premature senescence through activation of PI3K/AKT pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 183:109465. [PMID: 31376806 DOI: 10.1016/j.ecoenv.2019.109465] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/18/2019] [Accepted: 07/22/2019] [Indexed: 05/28/2023]
Abstract
Our group found that long-term low-dose exposure to hexavalent chromium [Cr(VI)] in L-02 hepatocytes resulted in premature senescence, which accompanied by the increased expression of Clusterin (CLU), but the functional role of CLU in premature senescence has never been explored. In the present study, the CLU overexpressed or silenced L-02 hepatocytes were established by lentiviral vector transfection. Cell viability assay, cell cycle analysis, western blotting, plate clone formation assay, and confocal microcopy were performed. The results indicated that Cr(VI)-induced premature senescence was associated with phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway inhibition, and high expression of CLU in the senescent cells exerted its functional role of promoting cell proliferation. CLU could complex with eukaryotic translation initiation factor 3 subunit I (EIF3I) and prevent its degradation, leading to the increase of AKT activity in Cr(VI)-exposed senescent hepatocytes. Blockage of the PI3K/AKT pathway with its inhibitor LY294002 eliminated the inhibitory effect of CLU on Cr(VI)-induced premature senescence. We concluded that high expression of CLU suppressed Cr(VI)-induced premature senescence through activation of PI3K/AKT pathway, which will provide the experimental basis for the study of Cr(VI)-induced liver cancer, especially for the elucidation of the mechanism of liver cancer cells escaping from senescence.
Collapse
Affiliation(s)
- Yujing Zhang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Yiyuan Zhang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Yuanyuan Xiao
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Caigao Zhong
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, PR China
| | - Fang Xiao
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, PR China.
| |
Collapse
|
14
|
Shen S, Dean DC, Yu Z, Duan Z. Role of cyclin-dependent kinases (CDKs) in hepatocellular carcinoma: Therapeutic potential of targeting the CDK signaling pathway. Hepatol Res 2019; 49:1097-1108. [PMID: 31009153 DOI: 10.1111/hepr.13353] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/23/2019] [Accepted: 03/28/2019] [Indexed: 12/12/2022]
Abstract
Liver cancer is the fourth leading cause of cancer related mortality in the world, with hepatocellular carcinoma (HCC) representing the most common primary subtype. Two-thirds of HCC patients have advanced disease when diagnosed, and for these patients, treatment strategies remain limited. In addition, there is a high incidence of tumor recurrence after surgical resection with the current treatment regimens. The development of novel and more effective agents is required. Cyclin-dependent kinases (CDKs) constitute a family of 21 different protein kinases involved in regulating cell proliferation, apoptosis, and drug resistance, and are evaluated in preclinical and clinical trials as chemotherapeutics. To summarize and discuss the therapeutic potential of targeting CDKs in HCC, recent published articles identified from PubMed were comprehensively reviewed. The key words included hepatocellular carcinoma, cyclin-dependent kinases, and CDK inhibitors. This review focuses on the emerging evidence from studies describing the genetic and functional aspects of CDKs in HCC. We also present an overview of CDK inhibitors that have shown efficacy in laboratory studies of HCC. Although many of the studies assessing CDK-targeting therapies in HCC are at the preclinical stage, there is significant evidence that CDK inhibitors used alone or in combination with established chemotherapy drugs could have significant applications in HCC.
Collapse
Affiliation(s)
- Shen Shen
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Sarcoma Biology Laboratory, Department of Orthopedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA, USA
| | - Dylan C Dean
- Sarcoma Biology Laboratory, Department of Orthopedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA, USA
| | - Zujiang Yu
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenfeng Duan
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Sarcoma Biology Laboratory, Department of Orthopedic Surgery, David Geffen School of Medicine at University of Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
15
|
Hsu SY, Yu HY, Lee WC, Hsiao CE, Wu CL, Cheng HT, Lin LJ, Li F, Chou YT, Cheng JW. A novel CXCL8 analog is effective in inhibiting the growth via cell cycle arrest and attenuating invasion of Lewis lung carcinoma. Onco Targets Ther 2019; 12:7611-7621. [PMID: 31571912 PMCID: PMC6754332 DOI: 10.2147/ott.s215824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/09/2019] [Indexed: 12/30/2022] Open
Abstract
Purpose Lung cancer and other solid tumors contain not only tumor cells but various types of stromal cells, such as fibroblasts and endothelial cells. In addition, tumors are infiltrated by inflammatory cells (neutrophils, macrophages, and lymphocytes). Tumor cells, stromal cells, and the tumor-associated leukocytes are responsible for the production of chemokines inside the tumor and the maintenance of systemic circulating chemokine levels. CXCL8 and its receptors, CXCR1 and CXCR2, were found to play important roles in tumor proliferation, migration, survival, and growth. We have developed a novel ELR-CXC chemokine antagonist CXCL8-IP10 based on the structure of CXCL8 and IP10. Patients and methods We assessed the anticancer efficacies of the blockade of CXCL8-CXCR1/2 axis in the Lewis lung carcinoma (LL/2) model using CXCL8-IP10. Results We found that CXCL8-IP10 markedly reduced LL/2 cell anchorage-independent growth and invasion. Moreover, we demonstrated that CXCL8-IP10 could significantly suppress tumor growth and improve survival rate as well as lifespan of C57BL/6 mice inoculated with LL/2 cells. Conclusion Our results suggest that ELR-CXC chemokine antagonism would potentially be a useful therapeutic approach in patients with lung cancer.
Collapse
Affiliation(s)
- Su-Ya Hsu
- Department of Medical Science, Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hui-Yuan Yu
- Division of Cancer Research, Rise Biopharmaceuticals Inc., Zhongguancun Shangdi Bio-medical Park, Beijing 100085, People's Republic of China
| | - Wei-Chen Lee
- Department of Medical Science, Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chia-En Hsiao
- Department of Medical Science, Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chih-Lung Wu
- Department of Medical Science, Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hsi-Tsung Cheng
- Division of Cancer Research, Rise Biopharmaceuticals Inc., Zhongguancun Shangdi Bio-medical Park, Beijing 100085, People's Republic of China
| | - Li-Jin Lin
- Division of Cancer Research, Rise Biopharmaceuticals Inc., Zhongguancun Shangdi Bio-medical Park, Beijing 100085, People's Republic of China
| | - Fang Li
- Department of Immunology, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Yu-Ting Chou
- Department of Medical Science, Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jya-Wei Cheng
- Department of Medical Science, Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
16
|
Zálešák F, Bon DJYD, Pospíšil J. Lignans and Neolignans: Plant secondary metabolites as a reservoir of biologically active substances. Pharmacol Res 2019; 146:104284. [PMID: 31136813 DOI: 10.1016/j.phrs.2019.104284] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
Lignans and neolignans are plant secondary metabolites derived from the oxidative coupling of phenylpropanoids. Biological activity of these phenolic compounds ranges from antioxidant, antitumor (terminaloside P, IC50 = 10 nM), anti-inflammatory, anti-neurodegenerative (schibitubin B, IC50 = 3.2 nM) and antiviral (patentiflorin A, IC50 = 14-23 nM) to antimicrobial. In addition, it was observed that several members of this group, namely enterolactone and its biochemical precursors also known as phytoestrogens, possess important protective properties. Most of these lignans and neolignans are presented in reasonable amounts in one's diet and thus the protection they provide against the colon and breast cancer, to name a few, is even more important to note. Similarly, neuroprotective properties were observed (schisanwilsonin G, IC50 = 3.2 nM) These structural motives also serve as an important starting point in the development of anticancer drugs. Presumably the most famous members of this family, etoposide and teniposide, synthetic derivatives of podophyllotoxin, are used in the clinical treatment of lymphocytic leukemia, certain brain tumors, and lung tumors already for nearly 20 years. This review describes 413 lignans and neolignans which have been isolated between 2016 and mid-2018 being reported in more than 300 peer-reviewed articles. It covers their source, structure elucidation, and bioactivity. Within the review, the structure-based overview of compounds as well as the bioactivity-based overview of compounds are described.
Collapse
Affiliation(s)
- František Zálešák
- Department of Organic Chemistry, Faculty of Science, Palacky University, tř. 17. listopadu 1192/12, CZ-771 46 Olomouc, Czech Republic.
| | - David Jean-Yves Denis Bon
- Department of Organic Chemistry, Faculty of Science, Palacky University, tř. 17. listopadu 1192/12, CZ-771 46 Olomouc, Czech Republic.
| | - Jiří Pospíšil
- Department of Organic Chemistry, Faculty of Science, Palacky University, tř. 17. listopadu 1192/12, CZ-771 46 Olomouc, Czech Republic; Laboratory of Growth Regulators, The Czech Academy of Sciences, Institute of Experimental Botany & Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic.
| |
Collapse
|
17
|
Li H, Jiang M, Cui M, Feng G, Dong J, Li Y, Xiao H, Fan S. MiR-365 enhances the radiosensitivity of non-small cell lung cancer cells through targeting CDC25A. Biochem Biophys Res Commun 2019; 512:392-398. [PMID: 30902389 DOI: 10.1016/j.bbrc.2019.03.082] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 12/25/2022]
Abstract
Radioresistance is a major challenge in lung cancer radiotherapy (RT), and consequently, new radiosensitizers are urgently needed. MicroRNAs (miRNAs) have been demonstrated to participate in many important cellular processes including radiosensitization. MiR-365 is dysregulated in non-small cell lung cancer (NSCLC) and is able to restrain the development of NSCLC. However, the relationship between miR-365 and radiosensitivities of NSCLC cells remains largely unknown. Here we reveal that overexpression of miR-365 is able to enhance the radiosensitivity of NSCLC cells through targeting CDC25A. We found that the expression level of miR-365 was positively correlated with the radiosensitivity of NSCLC cell lines. Furthermore, our results showed that overexpression of miR-365 could sensitize A549 cells to the irradiation. However, knockdown of miR-365 in H460 cells could act the converse manner. Mechanically, miR-365 was able to directly target 3'UTR of cell division cycle 25A (CDC25A) mRNA and reduce the expression of CDC25A at the levels of mRNA and protein. And we confirmed that miR-365 could increase the radiosensitivity of NSCLC cells by targeting CDC25A using in vitro and in vivo assays. Taken together, restoration of miR-365 expression enhances the radiosensitivity of NSCLC cells by suppressing CDC25A, and miR-365 could be used as a radiosensitizer for NSCLC therapy.
Collapse
Affiliation(s)
- Hang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Mian Jiang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Guoxing Feng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Huiwen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
18
|
Mali AV, Padhye SB, Anant S, Hegde MV, Kadam SS. Anticancer and antimetastatic potential of enterolactone: Clinical, preclinical and mechanistic perspectives. Eur J Pharmacol 2019; 852:107-124. [PMID: 30771348 DOI: 10.1016/j.ejphar.2019.02.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
Currently cancer is the second leading cause of death globally and worldwide incidence and mortality rates of all cancers of males and females are rising tremendously. In spite of advances in chemotherapy and radiation, metastasis and recurrence are considered as the major causes of cancer related deaths. Hence there is a mounting need to develop new therapeutic modalities to treat metastasis and recurrence in cancers. A significant amount of substantiation from epidemiological, clinical and laboratory research highlights the importance of diet and nutrition in cancer chemoprevention. Enterolactone (EL) is a bioactive phenolic metabolite known as a mammalian lignan derived from dietary lignans. Here in we review the reported anti-cancer properties of EL at preclinical as well as clinical level. Several in-vivo and in-vitro studies have provided strong evidence that EL exhibits potent anti-cancer and/or protective properties against different cancers including breast, prostate, colo-rectal, lung, ovarian, endometrial, cervical cancers and hepatocellular carcinoma. Reported laboratory studies indicate a clear role for EL in preventing cancer progression at various stages including cancer cell proliferation, survival, angiogenesis, inflammation and metastasis. In clinical settings, EL has been reported to reduce risk, decrease mortality rate and improve overall survival particularly in breast, prostate, colon, gastric and lung cancer. Further, the in-vitro human cell culture studies provide strong evidence of the anticancer and antimetastatic mechanisms of EL in several cancers. This comprehensive review supports an idea of projecting EL as a promising candidate for developing anticancer drug or adjunct dietary supplements and nutraceuticals.
Collapse
Affiliation(s)
- Aniket V Mali
- Center for Innovation in Nutrition Health and Disease (CINHD), Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Dhankawadi, Pune, Maharashtra 411043, India; Pharmaceutical Sciences, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra 411038, India
| | - Subhash B Padhye
- Interdisciplinary Science and Technology Research Academy, Abeda Inamdar College, University of Pune, Pune 411001, India; Cancer Biology, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Cancer Biology, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Mahabaleshwar V Hegde
- Center for Innovation in Nutrition Health and Disease (CINHD), Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Dhankawadi, Pune, Maharashtra 411043, India.
| | - Shivajirao S Kadam
- Pharmaceutical Sciences, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra 411038, India
| |
Collapse
|
19
|
Singhal J, Chikara S, Horne D, Salgia R, Awasthi S, Singhal SS. RLIP inhibition suppresses breast-to-lung metastasis. Cancer Lett 2019; 447:24-32. [PMID: 30684594 DOI: 10.1016/j.canlet.2019.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/08/2019] [Accepted: 01/19/2019] [Indexed: 11/26/2022]
Abstract
Breast tumor metastasis is a leading cause of cancer-related deaths worldwide. Breast cancer (BC) cells frequently metastasize to the lungs, where they pose a formidable therapeutic challenge. In the current study, we evaluated the anti-proliferative and anti-metastatic effects of 2'-hydroxyflavanone (2HF) and RLIP inhibition in an array of triple-negative BC cell lines and an orthotopic mouse model of breast-to-lung metastasis. Compared to control treatment, RLIP inhibition reduced in-vitro cell viability and suppressed the migratory and invasive potential of BC cells. In-vitro studies showed that 2HF treatment reduced the expression of RLIP, KRAS, pERK, pSTAT3, and pP70S6K. Further, mice orthotopically implanted with lung-seeking luciferase-expressing TMD231 cells were treated with 2HF (50 mg/kg, b.w.), RLIP antisense (RAS; 5 mg/kg, b.w.), RLIP antibody (Rab; 5 mg/kg, b.w.) or a combination of 2HF + RAS + Rab. 2HF-, RAS-, and Rab-treated mice exhibited significantly lower primary tumor weight and reduced lung metastasis compared to control mice. Mice treated with a combination of 2HF + RAS + Rab exhibited no metastasis and significantly lower tumor weight than the single agent-treated mice. Collectively, our results suggest that 2HF has potential to be combined with RLIP inhibition/depletion to more effectively suppress primary breast tumor growth and metastasis to the lungs.
Collapse
Affiliation(s)
- Jyotsana Singhal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA; Department of Molecular Medicine, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Shireen Chikara
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - David Horne
- Department of Molecular Medicine, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Sanjay Awasthi
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Sharad S Singhal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
20
|
Chatterjee S, Pietrofesa RA, Park K, Tao JQ, Carabe-Fernandez A, Berman AT, Koumenis C, Sielecki T, Christofidou-Solomidou M. LGM2605 Reduces Space Radiation-Induced NLRP3 Inflammasome Activation and Damage in In Vitro Lung Vascular Networks. Int J Mol Sci 2019; 20:ijms20010176. [PMID: 30621290 PMCID: PMC6337675 DOI: 10.3390/ijms20010176] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/24/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022] Open
Abstract
Updated measurements of charged particle fluxes during the transit from Earth to Mars as well as on site measurements by Curiosity of Martian surface radiation fluxes identified potential health hazards associated with radiation exposure for human space missions. Designing mitigation strategies of radiation risks to astronauts is critical. We investigated radiation-induced endothelial cell damage and its mitigation by LGM2605, a radioprotector with antioxidant and free radical scavenging properties. We used an in vitro model of lung vascular networks (flow-adapted endothelial cells; FAECs), exposed to gamma rays, low/higher linear energy transfer (LET) protons (3⁻4 or 8⁻10 keV/µm, respectively), and mixed field radiation sources (gamma and protons), given at mission-relevant doses (0.25 gray (Gy)⁻1 Gy). We evaluated endothelial inflammatory phenotype, NLRP3 inflammasome activation, and oxidative cell injury. LGM2605 (100 µM) was added 30 min post radiation exposure and gene expression changes evaluated 24 h later. Radiation induced a robust increase in mRNA levels of antioxidant enzymes post 0.25 Gy and 0.5 Gy gamma radiation, which was significantly decreased by LGM2605. Intercellular cell adhesion molecule-1 (ICAM-1) and NOD-like receptor protein 3 (NLRP3) induction by individual or mixed-field exposures were also significantly blunted by LGM2605. We conclude that LGM2605 is a likely candidate to reduce tissue damage from space-relevant radiation exposure.
Collapse
Affiliation(s)
- Shampa Chatterjee
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Ralph A Pietrofesa
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Kyewon Park
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Jian-Qin Tao
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Alejandro Carabe-Fernandez
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Abigail T Berman
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | | | - Melpo Christofidou-Solomidou
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
21
|
Francoz E, Lepiniec L, North HM. Seed coats as an alternative molecular factory: thinking outside the box. PLANT REPRODUCTION 2018; 31:327-342. [PMID: 30056618 DOI: 10.1007/s00497-018-0345-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/13/2018] [Indexed: 05/15/2023]
Abstract
Seed coats as commodities. Seed coats play important roles in the protection of the embryo from biological attack and physical damage by the environment as well as dispersion strategies. A significant part of the energy devoted by the mother plant to seed production is channeled into the production of the cell layers and metabolites that surround the embryo. Nevertheless, in crop species these are often discarded post-harvest and are a wasted resource that could be processed to yield co-products. The production of novel compounds from existing metabolites is also a possibility. A number of macromolecules are already accumulated in these maternal layers that could be exploited in industrial applications either directly or via green chemistry, notably flavonoids, lignin, lignan, polysaccharides, lipid polyesters and waxes. Here, we summarize our knowledge of the in planta biosynthesis pathways of these macromolecules and their molecular regulation as well as potential applications. We also outline recent work aimed at providing further tools for increasing yields of existing molecules or the development of novel biotech approaches, as well as trial studies aimed at exploiting this underused resource.
Collapse
Affiliation(s)
- Edith Francoz
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000, Versailles, France
| | - Loïc Lepiniec
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000, Versailles, France
| | - Helen M North
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000, Versailles, France.
| |
Collapse
|
22
|
Yao WF, Liu JW, Huang DS. MiR-200a inhibits cell proliferation and EMT by down-regulating the ASPH expression levels and affecting ERK and PI3K/Akt pathways in human hepatoma cells. Am J Transl Res 2018; 10:1117-1130. [PMID: 29736205 PMCID: PMC5934571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 11/06/2017] [Indexed: 06/08/2023]
Abstract
The primary objective of this study was to investigate the role of miR-200a in cell proliferation and epithelial-mesenchymal transition (EMT) through regulating targeting aspartate-β-hydroxylase (ASPH), which may further affect the activation of ERK/PI3K/Akt pathway. Liver cancer and adjacent tissues were collected from 72 cases of liver cancer patients with surgery in our hospital. In this study, the mRNA expression level of miR-200a was significantly decreased by real-time PCR (RT-PCR) detection. ASPH expressions, however, had an opposite tendency compared to that of miR-200a. We found a significantly negative correlation between miR-200a expressions and ASPH expressions. The survival rate of liver cancer patients with the low expressed ASPH was significantly higher than those with the high expressed ASPH. RT-PCR and Western blot results showed that low expressed miR-200a and highexpressed ASPH were found in liver cancer cell lines. Further research discovered that miR-200a transfection could significantly decrease the relative luciferase activity when it was integrated with ASPH 3'-untranslated region (3'-UTR) in HepG2 cells. Cell Counting Kit (CCK-8) detection showed that treatment with miR-200a mimics reduced cell viability, while the over-expressed ASPH increased cell viability by regulating the c-mycmrna (c-Myc) and Cyclin-D1 expressions. The EMT-related genes including E-Cadherin, N-Cadherin and Vimentin expressions were significantly increased, whereas the over-expressed ASPH exerted the opposite effects. In addition, extracellular signal regulated kinase (ERK), phosphoinositide-3-kinase (PI3K) and serine threonine kinase (AKT) were suppressed by miR-200a mimics. In conclusion, miR-200a inhibits cell proliferation and EMT in human hepatoma cells by targeting ASPH and affecting ERK and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Wei-Feng Yao
- Department of Hepato-Biliary-Pancreatic Surgery, People's Hospital of Hangzhou Medical College Hangzhou 310014, P.R. China
| | - Jun-Wei Liu
- Department of Hepato-Biliary-Pancreatic Surgery, People's Hospital of Hangzhou Medical College Hangzhou 310014, P.R. China
| | - Dong-Sheng Huang
- Department of Hepato-Biliary-Pancreatic Surgery, People's Hospital of Hangzhou Medical College Hangzhou 310014, P.R. China
| |
Collapse
|
23
|
Zhao C, Li Y, Zhang W, Zhao D, Ma L, Ma P, Yang F, Wang Y, Shu Y, Qiu W. IL‑17 induces NSCLC A549 cell proliferation via the upregulation of HMGA1, resulting in an increased cyclin D1 expression. Int J Oncol 2018; 52:1579-1592. [PMID: 29512693 DOI: 10.3892/ijo.2018.4307] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/16/2018] [Indexed: 11/06/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is considered to be an inflammation-associated carcinoma. Although interleukin‑17 (IL‑17) production contributes to the proliferation and growth of NSCLC, the mechanisms underlying IL‑17-induced NSCLC cell proliferation have not been fully elucidated. In the present study, by using ELISA and immunohistochemical analyses, we first found that the expression levels of IL‑17, IL‑17 receptor (IL‑17R), high-mobility group A1 (HMGA1) and cyclin D1 were elevated in the samples of patients with NSCLC. Subsequently, by RT-qPCR, western blot analysis and cell proliferation assay in vitro, we revealed that stimulation with recombinant human IL‑17 (namely IL‑17A) markedly induced the expression of HMGA1 and cyclin D1 in the A549 cells (a human lung adenocarcinoma cell line) and promoted cell proliferation. Furthermore, luciferase reporter and ChIP assays confirmed that upregulated HMGA1 directly bound to the cyclin D1 gene promoter and activated its transcription. Notably, the response element of HMGA1 binding to the cyclin D1 promoter was disclosed for the first time, at least to the best of our knowledge. Taken together, our findings indicate that the IL‑17/HMGA1/cyclin D1 axis plays an important role in NSCLC cell proliferation and may provide new insight into NSCLC pathogenesis and may thus aid in the development of novel therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Chenhui Zhao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Yongting Li
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Weiming Zhang
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Dan Zhao
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Ling Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Fengming Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Yingwei Wang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Wen Qiu
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
24
|
Chikara S, Mamidi S, Sreedasyam A, Chittem K, Pietrofesa R, Zuppa A, Moorthy G, Dyer N, Christofidou-Solomidou M, Reindl KM. Flaxseed Consumption Inhibits Chemically Induced Lung Tumorigenesis and Modulates Expression of Phase II Enzymes and Inflammatory Cytokines in A/J Mice. Cancer Prev Res (Phila) 2017; 11:27-37. [PMID: 29074535 DOI: 10.1158/1940-6207.capr-17-0119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/26/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022]
Abstract
Flaxseed consumption is associated with reduced oxidative stress and inflammation in lung injury models and has shown anticancer effects for breast and prostate tissues. However, the chemopreventive potential of flaxseed remains unexplored for lung cancer. In this study, we investigated the effect of flaxseed on tobacco smoke carcinogen (NNK)-induced lung tumorigenesis in an A/J mouse model. Mice exposed to NNK were fed a control diet or a 10% flaxseed-supplemented diet for 26 weeks. Flaxseed-fed mice showed reduced lung tumor incidence (78%) and multiplicity, with an average of 2.7 ± 2.3 surface lung tumor nodules and 1.0 ± 0.9 H&E cross-section nodules per lung compared with the control group, which had 100% tumor incidence and an average of 10.2 ± 5.7 surface lung tumor nodules and 3.9 ± 2.6 H&E cross-section nodules per lung. Furthermore, flaxseed-fed mice had a lower incidence of adenocarcinomas compared with control-fed mice. Western blotting performed on normal lung tissues showed flaxseed suppressed phosphorylation (activation) of p-AKT, p-ERK, and p-JNK kinases. RNA-Seq data obtained from normal lung and lung tumors of control and flaxseed-fed mice suggested that flaxseed intake resulted in differential expression of genes involved in inflammation-mediated cytokine signaling (IL1, 6, 8, 9, and 12α), xenobiotic metabolism (several CYPs, GSTs, and UGTs), and signaling pathways (AKT and MAPK) involved in tumor cell proliferation. Together, our results indicate that dietary flaxseed supplementation may be an effective chemoprevention strategy for chemically induced lung carcinogenesis by altering signaling pathways, inflammation, and oxidative stress. Cancer Prev Res; 11(1); 27-37. ©2017 AACR.
Collapse
Affiliation(s)
- Shireen Chikara
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota
| | - Sujan Mamidi
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | | | - Kishore Chittem
- Department of Plant Pathology, North Dakota State University, Fargo, North Dakota
| | - Ralph Pietrofesa
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Athena Zuppa
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ganesh Moorthy
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Neil Dyer
- Department of Animal Sciences, North Dakota State University, Fargo, North Dakota
| | | | - Katie M Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota.
| |
Collapse
|
25
|
Phytoestrogen metabolism by lactic acid bacteria: Enterolignan production by Lactobacillus salivarius and Lactobacillus gasseri strains. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.08.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
26
|
The Synthetic Lignan Secoisolariciresinol Diglucoside Prevents Asbestos-Induced NLRP3 Inflammasome Activation in Murine Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7395238. [PMID: 29075366 PMCID: PMC5615985 DOI: 10.1155/2017/7395238] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The interaction of asbestos with macrophages drives two key processes that are linked to malignancy: (1) the generation of reactive oxygen species (ROS)/reactive nitrogen species (RNS) and (2) the activation of an inflammation cascade that drives acute and chronic inflammation, with the NLRP3 inflammasome playing a key role. Synthetic secoisolariciresinol diglucoside (SDG), LGM2605, is a nontoxic lignan with anti-inflammatory and antioxidant properties and was evaluated for protection from asbestos in murine peritoneal macrophages (MF). METHODS MFs were exposed to crocidolite asbestos ± LGM2605 given 4 hours prior to exposure and evaluated at various times for NLRP3 expression, secretion of inflammasome-activated cytokines (IL-1β and IL-18), proinflammatory cytokines (IL-6, TNFα, and HMGB1), NF-κB activation, and levels of total nitrates/nitrites. RESULTS Asbestos induces a significant (p < 0.0001) increase in the NLRP3 subunit, release of proinflammatory cytokines, NLRP3-activated cytokines, NF-κB, and levels of nitrates/nitrites. LGM2605 significantly reduced NLRP3 ranging from 40 to 81%, IL-1β by 89-96%, and TNFα by 67-78%, as well as activated NF-κB by 48-49% while decreasing levels of nitrates/nitrites by 85-93%. CONCLUSIONS LGM2605 reduced asbestos-induced NLRP3 expression, proinflammatory cytokine release, NF-κB activation, and nitrosative stress in MFs supporting its possible use in preventing the asbestos-induced inflammatory cascade leading to malignancy.
Collapse
|