1
|
Rodak O, Mrozowska M, Rusak A, Gomułkiewicz A, Piotrowska A, Olbromski M, Podhorska-Okołów M, Ugorski M, Dzięgiel P. Targeting SOX18 Transcription Factor Activity by Small-Molecule Inhibitor Sm4 in Non-Small Lung Cancer Cell Lines. Int J Mol Sci 2023; 24:11316. [PMID: 37511076 PMCID: PMC10379584 DOI: 10.3390/ijms241411316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
The transcription factor SOX18 has been shown to play a crucial role in lung cancer progression and metastasis. In this study, we investigated the effect of Sm4, a SOX18 inhibitor, on cell cycle regulation in non-small cell lung cancer (NSCLC) cell lines LXF-289 and SK-MES-1, as well as normal human lung fibroblast cell line IMR-90. Our results demonstrated that Sm4 treatment induced cytotoxic effects on all three cell lines, with a greater effect observed in NSCLC adenocarcinoma cells. Sm4 treatment led to S-phase cell accumulation and upregulation of p21, a key regulator of the S-to-G2/M phase transition. While no significant changes in SOX7 or SOX17 protein expression were observed, Sm4 treatment resulted in a significant upregulation of SOX17 gene expression. Furthermore, our findings suggest a complex interplay between SOX18 and p21 in the context of lung cancer, with a positive correlation observed between SOX18 expression and p21 nuclear presence in clinical tissue samples obtained from lung cancer patients. These results suggest that Sm4 has the potential to disrupt the cell cycle and target cancer cell growth by modulating SOX18 activity and p21 expression. Further investigation is necessary to fully understand the relationship between SOX18 and p21 in lung cancer and to explore the therapeutic potential of SOX18 inhibition in lung cancer.
Collapse
Affiliation(s)
- Olga Rodak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Monika Mrozowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Gomułkiewicz
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Mateusz Olbromski
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
2
|
Zhu Z, Liu Z, Cui J, Huang Y, Chen H, Wu Y, Huang X, Gan C. Apoptosis inducing properties of 3-biotinylate-6-benzimidazole B-nor-cholesterol analogues. Steroids 2021; 169:108822. [PMID: 33722574 DOI: 10.1016/j.steroids.2021.108822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 01/14/2023]
Abstract
In this work, a series of Biotin-substituted B-nor-cholesteryl benzimidazole compounds were synthesized. The antiproliferativeactivities of these compounds were evaluated in vitro using a series of human cancer cell lines, including HeLa (cervical cancer), SKOV3 (ovarian cancer), T-47D (thymus gland cancer), MCF-7 (human breast cancer) and HEK293T (normal renal epithelial) cells. These compounds displayed distinct antiproliferative activities against the currently tested cancer cells. The apoptotic properties induced by compound 6d were further investigated. Our results showed that compound 6d could induce the apoptosis of SKOV3 cells, blocking the cell growth in S-phase. Western blotting analyses revealed that compound 6d can induce cell apoptosis via the mitochondria-dependent pathway.
Collapse
Affiliation(s)
- Zhiling Zhu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Zhiping Liu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Jianguo Cui
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Yanmin Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Hualong Chen
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Yulan Wu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Xiaotong Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Chunfang Gan
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China.
| |
Collapse
|
3
|
Li SY, Wang CY, Zhao JJ, Long CY, Xiao YX, Tang XB, Yuan ZW, Bai YZ. Upregulation of PPPDE1 contributes to anorectal malformations via the mitochondrial apoptosis pathway during hindgut development in rats. Exp Cell Res 2021; 402:112574. [PMID: 33794264 DOI: 10.1016/j.yexcr.2021.112574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Congenital anorectal malformations (ARMs) are among the most prominent deformities of the gastrointestinal tract; however, their precise aetiology remains obscure. Immunohistochemistry demonstrated that, in the ARM group, the PPPDE1-positive cells were widely distributed in the hindgut epithelial tissue from GD13 to GD16. Immunofluorescence revealed that most TUNEL-, Bax-, and Cytochrome C (Cyt C)-positive cells overlapped with PPPDE1-positive cells in the urorectal septum (URS). Western blotting and quantitative real-time RT-PCR revealed that PPPDE1 levels were significantly higher in the ARM group from GD13 to GD14 (p < 0.05). IEC-6 cells were transfected with PPPDE1 overexpression plasmid/NC (negative control) or si-PPPDE1/si-NC. Flow cytometry analysis and CCK-8 assay (used to detect apoptosis and proliferation, respectively), as well as western blotting, showed that the levels of PPPDE1 were positively correlated with the pro-apoptotic molecules Bax and Cyt C. Accordingly, aberrantly high expression of PPPDE1 caused a spatiotemporal imbalance in foetal rats with ARMs during hindgut development. Therefore, the upregulation of PPPDE1 may promote epithelial apoptosis and reduce proliferation in the hindgut via the mitochondrial apoptotic pathway. This could affect the fusion of the URS and cloacal membrane, ultimately inhibiting the hindgut development and resulting in ARMs.
Collapse
Affiliation(s)
- Si Ying Li
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Chen Yi Wang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Jing Jing Zhao
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Cai Yun Long
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Yun Xia Xiao
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Xiao Bing Tang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Zheng Wei Yuan
- The Key Laboratory of Health Ministry for Congenital Malformation, Shenyang, 110004, China
| | - Yu Zuo Bai
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
4
|
Ou X, Zhang GT, Xu Z, Chen JS, Xie Y, Liu JK, Liu XP. Desumoylating Isopeptidase 2 (DESI2) Inhibits Proliferation and Promotes Apoptosis of Pancreatic Cancer Cells through Regulating PI3K/AKT/mTOR Signaling Pathway. Pathol Oncol Res 2018; 25:635-646. [DOI: 10.1007/s12253-018-0487-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
|
5
|
Yan H, Guo W, Li K, Tang M, Zhao X, Lei Y, Nie CL, Yuan Z. Combination of DESI2 and endostatin gene therapy significantly improves antitumor efficacy by accumulating DNA lesions, inducing apoptosis and inhibiting angiogenesis. Exp Cell Res 2018; 371:50-62. [DOI: 10.1016/j.yexcr.2018.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/11/2018] [Accepted: 07/24/2018] [Indexed: 10/28/2022]
|
6
|
Lin C, Yan H, Yang J, Li L, Tang M, Zhao X, Nie C, Luo N, Wei Y, Yuan Z. Combination of DESI2 and IP10 gene therapy significantly improves therapeutic efficacy against murine carcinoma. Oncotarget 2017; 8:56281-56295. [PMID: 28915590 PMCID: PMC5593561 DOI: 10.18632/oncotarget.17623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/20/2017] [Indexed: 02/05/2023] Open
Abstract
DESI2 (also known as PNAS-4) is a novel pro-apoptotic gene activated during the early response to DNA damage. We previously reported that overexpression of DESI2 induces S phase arrest and apoptosis by activating checkpoint kinases. The present study was designed to test whether combination of DESI2 and IP10 could improve the therapy efficacy in vitro and in vivo. The recombinant plasmid co-expressing DESI2 and IP10 was encapsulated with DOTAP/Cholesterol nanoparticle. Immunocompetent mice bearing CT26 colon carcinoma and LL2 lung cancer were treated with the complex. We found that, in vitro, the combination of DESI2 and IP10 more efficiently inhibited proliferation of CT26, LL2, SKOV3 and A549 cancer cells via apoptosis. In vivo, the combined gene therapy more significantly inhibited tumor growth and efficiently prolonged the survival of tumor bearing mice. Mechanistically, the augmented antitumor activity in vivo was associated with induction of apoptosis and inhibition of angiogenesis. The anti-angiogenesis was further mimicked by inhibiting proliferation of immortalized HUVEC cells in vitro. Meanwhile, the infiltration of lymphocytes also contributed to the enhanced antitumor effects. Depletion of CD8+ T lymphocytes significantly abrogated the antitumor activity, whereas depletion of CD4+ T cells or NK cells showed partial abrogation. Our data suggest that the combined gene therapy of DESI2 and IP10 can significantly enhance the antitumor activity as apoptosis inducer, angiogenesis inhibitor and immune response initiator. The present study may provide a novel and effective method for treating cancer.
Collapse
Affiliation(s)
- Chao Lin
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - HuaYing Yan
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
- Department of Functional Imaging, Sichuan Provincial Women's and Children's Hospital, Chengdu, 610031, China
| | - Jun Yang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Lei Li
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Mei Tang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Xinyu Zhao
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Chunlai Nie
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Na Luo
- Nankai University School of Medicine, Collaborative Innovation Center of Biotherapy, Tianjin, 300071, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Zhu Yuan
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Chen RJ, Wu PH, Ho CT, Way TD, Pan MH, Chen HM, Ho YS, Wang YJ. P53-dependent downregulation of hTERT protein expression and telomerase activity induces senescence in lung cancer cells as a result of pterostilbene treatment. Cell Death Dis 2017; 8:e2985. [PMID: 28796247 PMCID: PMC5596539 DOI: 10.1038/cddis.2017.333] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022]
Abstract
Cellular senescence is characterized by permanent cell cycle arrest, triggered by a variety of stresses, such as telomerase inhibition, and it is recognized as a tumor-suppressor mechanism. In recent years, telomerase has become an important therapeutic target in several cancers; inhibition of telomerase can induce senescence via the DNA damage response (DDR). Pterostilbene (PT), a dimethyl ether analog of resveratrol, possesses a variety of biological functions, including anticancer effects; however, the molecular mechanisms underlying these effects are not fully understood. In this study, we investigated the possible mechanisms of PT-induced senescence through telomerase inhibition in human non-small cell lung cancer cells and delineated the role of p53 in senescence. The results indicated that PT-induced senescence is characterized by a flattened morphology, positive staining for senescence-associated-β galactosidase activity, and the formation of senescence-associated heterochromatic foci. Telomerase activity and protein expression was significantly decreased in H460 (p53 wild type) cells compared with H1299 (p53 null) cells and p53 knockdown H460 cells (H460-p53-). A more detailed mechanistic study revealed that PT-induced senescence partially occurred via a p53-dependent mechanism, triggering inhibition of telomerase activity and protein expression, and leading to the DDR, S phase arrest and, finally, cellular senescence. This study is the first to explore the novel anticancer mechanism of PT senescence induction via the inhibition of telomerase in lung cancer cells.
Collapse
Affiliation(s)
- Rong-Jane Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Hsuan Wu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Tzong-Der Way
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan.,Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization; Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains; Huanggang Normal University, Huanggang, Hubei, China
| | - Hsiu-Min Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Soon Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Biomedical Informatics, Asia University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
8
|
Yang J, Zhao X, Tang M, Li L, Lei Y, Cheng P, Guo W, Zheng Y, Wang W, Luo N, Peng Y, Tong A, Wei Y, Nie C, Yuan Z. The role of ROS and subsequent DNA-damage response in PUMA-induced apoptosis of ovarian cancer cells. Oncotarget 2017; 8:23492-23506. [PMID: 28423586 PMCID: PMC5410321 DOI: 10.18632/oncotarget.15626] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/14/2017] [Indexed: 02/05/2023] Open
Abstract
PUMA is a member of the "BH3-only" branch of the BCL-2 family. Our previous study suggests a therapeutic potential of PUMA in treating ovarian cancer, however, the action mechanism of PUMA remains elusive. In this work, we found that in PUMA adenovirus-infected A2780s ovarian cancer cells, exogenous PUMA was partially accumulated in the cytosol and mainly located to the mitochondria. We further showed that PUMA induces mitochondrial dysfunction-mediated apoptosis and ROS generation through functional BAX in a ROS generating enzyme- and caspase-independent manner irrespective of their p53 status, and results in activation of Nrf2/HO-1 pathway. Furthermore, PUMA induces DNA breaks in γ-H2AX staining, and causes activation of DNA damage-related kinases including ATM, ATR, DNA-PKcs, Chk1 and Chk2, which are correlated with the apoptosis. PUMA also results in ROS-triggered JNK activation. Intriguingly, JNK plays a dual role in both DNA damage response and apoptosis, and has an additional contribution to apoptosis. Taken together, we have provided new insight into the action mechanism by which elevated PUMA first induces ROS generation then results in DNA damage response and JNK activation, ultimately contributing to apoptosis in ovarian cancer cells.
Collapse
Affiliation(s)
- Jun Yang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Zhao
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mei Tang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Li
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Lei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Cheng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenhao Guo
- 2 Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu Zheng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Wang
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Luo
- 3 Nankai University, School of Medicine/Collaborative Innovation Center of Biotherapy, Tianjin 300071, China
| | - Yong Peng
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Aiping Tong
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chunlai Nie
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhu Yuan
- 1 State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Chuffa LGA, Alves MS, Martinez M, Camargo ICC, Pinheiro PFF, Domeniconi RF, Júnior LAL, Martinez FE. Apoptosis is triggered by melatonin in an in vivo model of ovarian carcinoma. Endocr Relat Cancer 2016; 23:65-76. [PMID: 26555801 DOI: 10.1530/erc-15-0463] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/09/2015] [Indexed: 12/20/2022]
Abstract
Apoptosis plays an important role in the treatment of cancer, and targeting apoptosis-related molecules in ovarian cancer (OC) is of great therapeutic value. Melatonin (Mel) is an indoleamine displaying several anti-cancer properties and has been reported to modulate apoptosis signaling in multiple tumor subtypes. We investigated OC and the role of Mel therapy on the pro-apoptotic (p53, BAX, caspase-3, and cleaved caspase-3) and anti-apoptotic (Bcl-2 and survivin) proteins in an ethanol (EtOH)-preferring rat model. To induce OC, the left ovary was injected directly with a single dose of 100 μg 7,12-dimethylbenz(a)anthracene dissolved in 10 μl of sesame oil under the bursa. Right ovaries were used as sham-surgery controls. After developing OC, half of the animals received i.p. injections of Mel (200 μg/100 g BW per day) for 60 days. Body weight gain, EtOH consumption, and energy intake were unaffected by the treatments. Interestingly, absolute and relative OC masses showed a significant reduction after Mel therapy, regardless of EtOH consumption. To accomplish OC-related apoptosis, we first observed that p53, BAX, caspase-3, and cleaved caspase-3 were downregulated in OC tissue while Bcl-2 and survivin were overexpressed. Notably, Mel therapy and EtOH intake promoted apoptosis along with the upregulation of p53, BAX, and cleaved caspase-3. Fragmentation of DNA observed by TUNEL-positive nuclei was also enhanced following Mel treatment. In addition, Bcl-2 was downregulated by the EtOH intake and lower survivin levels were observed after Mel therapy. Taken together, these results suggest that Mel induce apoptosis in OC cells of EtOH-preferring animals.
Collapse
Affiliation(s)
- Luiz Gustavo A Chuffa
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Michelly S Alves
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Marcelo Martinez
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Isabel Cristina C Camargo
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Patricia F F Pinheiro
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Raquel F Domeniconi
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Luiz Antonio L Júnior
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Francisco Eduardo Martinez
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| |
Collapse
|
10
|
Yuan Z, Guo W, Yang J, Li L, Wang M, Lei Y, Wan Y, Zhao X, Luo N, Cheng P, Liu X, Nie C, Peng Y, Tong A, Wei Y. PNAS-4, an Early DNA Damage Response Gene, Induces S Phase Arrest and Apoptosis by Activating Checkpoint Kinases in Lung Cancer Cells. J Biol Chem 2015; 290:14927-44. [PMID: 25918161 DOI: 10.1074/jbc.m115.658419] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Indexed: 02/05/2023] Open
Abstract
PNAS-4, a novel pro-apoptotic gene, was activated during the early response to DNA damage. Our previous study has shown that PNAS-4 induces S phase arrest and apoptosis when overexpressed in A549 lung cancer cells. However, the underlying action mechanism remains far from clear. In this work, we found that PNAS-4 expression in lung tumor tissues is significantly lower than that in adjacent lung tissues; its expression is significantly increased in A549 cells after exposure to cisplatin, methyl methane sulfonate, and mitomycin; and its overexpression induces S phase arrest and apoptosis in A549 (p53 WT), NCI-H460 (p53 WT), H526 (p53 mutation), and Calu-1 (p53(-/-)) lung cancer cells, leading to proliferation inhibition irrespective of their p53 status. The S phase arrest is associated with up-regulation of p21(Waf1/Cip1) and inhibition of the Cdc25A-CDK2-cyclin E/A pathway. Up-regulation of p21(Waf1/Cip1) is p53-independent and correlates with activation of ERK. We further showed that the intra-S phase checkpoint, which occurs via DNA-dependent protein kinase-mediated activation of Chk1 and Chk2, is involved in the S phase arrest and apoptosis. Gene silencing of Chk1/2 rescues, whereas that of ATM or ATR does not affect, S phase arrest and apoptosis. Furthermore, human PNAS-4 induces DNA breaks in comet assays and γ-H2AX staining. Intriguingly, caspase-dependent cleavage of Chk1 has an additional role in enhancing apoptosis. Taken together, our findings suggest a novel mechanism by which elevated PNAS-4 first causes DNA-dependent protein kinase-mediated Chk1/2 activation and then results in inhibition of the Cdc25A-CDK2-cyclin E/A pathway, ultimately causing S phase arrest and apoptosis in lung cancer cells.
Collapse
Affiliation(s)
- Zhu Yuan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China,
| | - Wenhao Guo
- the Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 37, Guoxue Road, Chengdu 610041, Sichuan Province, China, and
| | - Jun Yang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Lei Li
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Meiliang Wang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Yi Lei
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Yang Wan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Xinyu Zhao
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Na Luo
- the Nankai University School of Medicine/Collaborative Innovation Center of Biotherapy, Tianjin 300071, China
| | - Ping Cheng
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Xinyu Liu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Chunlai Nie
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Yong Peng
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Aiping Tong
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China,
| | - Yuquan Wei
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| |
Collapse
|
11
|
Figueiredo CR, Matsuo AL, Massaoka MH, Girola N, Azevedo RA, Rabaça AN, Farias CF, Pereira FV, Matias NS, Silva LP, Rodrigues EG, Lago JHG, Travassos LR, Silva RMG. Antitumor activity of kielmeyera coriacea leaf constituents in experimental melanoma, tested in vitro and in vivo in syngeneic mice. Adv Pharm Bull 2014; 4:429-36. [PMID: 25364658 DOI: 10.5681/apb.2014.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 02/07/2014] [Accepted: 02/19/2014] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The antitumor activity of Kielmeyera coriacea (Clusiaceae), a medicinal plant used in the treatment of parasitic, as well as fungal and bacterial infections by the Brazilian Cerrado population, was investigated. METHODS A chloroform extract (CE) of K. coriacea was tested in the murine melanoma cell line (B16F10-Nex2) and a panel of human tumor cell lines. Tumor cell migration was determined by the wound-healing assay and the in vivo antitumor activity of CE was investigated in a melanoma cell metastatic model. 1H NMR and GC/MS were used to determine CE chemical composition. RESULTS We found that CE exhibited strong cytotoxic activity against murine melanoma cells and a panel of human tumor cell lines in vitro. CE also inhibited growth of B16F10-Nex2 cells at sub lethal concentrations, inducing cell cycle arrest at S phase, and inhibition of tumor cell migration. Most importantly, administration of CE significantly reduced the number of melanoma metastatic nodules in vivo. Chemical analysis of CE indicated the presence of the long chain fatty compounds, 1-eicosanol, 1-docosanol, and 2-nonadecanone as main constituents. CONCLUSION These results indicate that K. coriacea is a promising medicinal plant in cancer therapy exhibiting antitumor activity both in vitro and in vivo against different tumor cell lines.
Collapse
Affiliation(s)
- Carlos Rogério Figueiredo
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Alisson Leonardo Matsuo
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Mariana Hiromi Massaoka
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Natalia Girola
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ricardo Alexandre Azevedo
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Aline Nogueira Rabaça
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Camyla Fernandes Farias
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Felipe Valença Pereira
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Natalia Silva Matias
- Department of Biological Sciences - Laboratory of Herbal Medicines, Universidade Estadual Paulista (UNESP- FLC/Assis), São Paulo, Brazil
| | - Luciana Pereira Silva
- Department of Biological Sciences - Laboratory of Herbal Medicines, Universidade Estadual Paulista (UNESP- FLC/Assis), São Paulo, Brazil
| | - Elaine Guadelupe Rodrigues
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - João Henrique Guilardi Lago
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo (UNIFESP), Diadema, São Paulo, SP, Brazil
| | - Luiz Rodolpho Travassos
- Department of Microbiology, Immunology and Parasitology, Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Regildo Márcio Gonçalves Silva
- Department of Biological Sciences - Laboratory of Herbal Medicines, Universidade Estadual Paulista (UNESP- FLC/Assis), São Paulo, Brazil
| |
Collapse
|
12
|
Lee YS, Choi KM, Kim W, Jeon YS, Lee YM, Hong JT, Yun YP, Yoo HS. Hinokitiol inhibits cell growth through induction of S-phase arrest and apoptosis in human colon cancer cells and suppresses tumor growth in a mouse xenograft experiment. JOURNAL OF NATURAL PRODUCTS 2013; 76:2195-202. [PMID: 24308647 DOI: 10.1021/np4005135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Hinokitiol (1), a tropolone-related natural compound, induces apoptosis and has anti-inflammatory, antioxidant, and antitumor activities. In this study, the inhibitory effects of 1 were investigated on human colon cancer cell growth and tumor formation of xenograft mice. HCT-116 and SW-620 cells derived from human colon cancers were found to be similarly susceptible to 1, with IC50 values of 4.5 and 4.4 μM, respectively. Compound 1 induced S-phase arrest in the cell cycle progression and decreased the expression levels of cyclin A, cyclin E, and Cdk2. Conversely, 1 increased the expression of p21, a Cdk inhibitor. Compound 1 decreased Bcl-2 expression and increased the expression of Bax, and cleaved caspase-9 and -3. The effect of 1 on tumor formation when administered orally was evaluated in male BALB/c-nude mice implanted intradermally separately with HCT-116 and SW-620 cells. Tumor volumes and tumor weights in the mice treated with 1 (100 mg/kg) were decreased in both cases. These results suggest that the suppression of tumor formation by compound 1 in human colon cancer may occur through cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Youn-Sun Lee
- Department of Biology Education, College of Education, Chungbuk National University , Cheongju 361-763, Korea
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Qian C, Yao J, Wang J, Wang L, Xue M, Zhou T, Liu W, Si J. ERK1/2 inhibition enhances apoptosis induced by JAK2 silencing in human gastric cancer SGC7901 cells. Mol Cell Biochem 2013; 387:159-70. [PMID: 24178240 DOI: 10.1007/s11010-013-1881-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 10/18/2013] [Indexed: 12/11/2022]
Abstract
Recent studies suggest JAK2 signaling may be a therapeutic target for treatment of gastric cancer (GC). However, the exact roles of JAK2 in gastric carcinogenesis are not very clear. Here, we have targeted JAK2 to be silenced by shRNA and investigated the biological functions and related mechanisms of JAK2 in GC cell SGC7901. In this study, JAK2 is commonly highly expressed in GC tissues as compared to their adjacent normal tissues (n = 75, p < 0.01). Specific down-regulation of JAK2 suppressed cell proliferation and colony-forming units, induced G2/M arrest in SGC7901 cells, but had no significant effect on cell apoptosis in vitro or tumor growth inhibition in vivo. Interestingly, JAK2 silencing-induced activation of ERK1/2, and inactivation of ERK1/2 using the specific ERK inhibitor PD98059 markedly enhanced JAK2 shRNA-induced cell proliferation inhibition, cell cycle arrest and apoptosis. Ultimately, combination of PD98059 and JAK2 shRNA significantly inhibited tumor growth in nude mice. Our results implicate JAK2 silencing-induced cell proliferation inhibition, cell cycle arrest, and ERK1/2 inhibition could enhance apoptosis induced by JAK2 silencing in SGC7901 cells.
Collapse
Affiliation(s)
- Cuijuan Qian
- Institute of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, Zhejiang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|