1
|
Histone methylation in pre-cancerous liver diseases and hepatocellular carcinoma: recent overview. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1594-1605. [PMID: 36650321 DOI: 10.1007/s12094-023-03078-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/07/2023] [Indexed: 01/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is the prevalent form of liver cancer in adults and the fourth most common cause of cancer-related death worldwide. HCC predominantly arises in the context of cirrhosis as a result of chronic liver disease, injury and inflammation. Full-blown HCC has poor prognosis because it is highly aggressive and resistant to therapy. Consequently, interventions that can prevent or restrain HCC emergence from pre-cancerous diseased liver are a desirable strategy. Histone methylation is a dynamic, reversible epigenetic modification involving the addition or removal of methyl groups from lysine, arginine or glutamine residues. Aberrant activity of histone methylation writers, erases and readers has been implicated in several cancer types, including HCC. In this review, we provide an overview of research on the role of histone methylation in pre-cancerous and cancerous HCC published over the last 5 years. In particular, we present the evidence linking environmental factors such as diet, viral infections and carcinogenic agents with dysregulation of histone methylation during liver cancer progression with the aim to highlight future therapeutic possibilities.
Collapse
|
2
|
Li J, Zhang S, Liao D, Zhang Q, Chen C, Yang X, Jiang D, Pang J. Overexpression of PFKFB3 promotes cell glycolysis and proliferation in renal cell carcinoma. BMC Cancer 2022; 22:83. [PMID: 35057732 PMCID: PMC8772232 DOI: 10.1186/s12885-022-09183-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
Cancer cells prefer utilizing aerobic glycolysis in order to exacerbate tumor mass and maintain un-regulated proliferative rates. As a key glycolytic activator, phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) has been implicated in multiple tumor type progression. However, the specific function and clinical significance of PFKFB3 in renal cell carcinoma (RCC) are yet not clarified. This investigation assessed PFKFB3 roles in RCC.
Methods
PFKFB3 expression levels were analyzed in clear cell renal cell carcinoma (ccRCC) tissues, together with its relationship with clinical characteristics of ccRCC. Real-time PCR and Western blot assays were employed for determining PFKFB3 expression in different RCC cell lines. Furthermore, we determined the glycolytic activity by glucose uptake, lactate secretion assay and ECAR analysis. CCK-8 assay, clone formation, flow cytometry and EdU assessments were performed for monitoring tumor proliferative capacity and cell-cycle distribution. Furthermore, a murine xenograft model was employed for investigating the effect of PFKFB3 on tumor growth in vivo.
Results
PFKFB3 was significantly up-regulated in RCC specimens and cell lines in comparison to normal control. Overexpression of PFKFB3 was directly correlated to later TNM stages, thus becoming a robust prognostic biomarker for ccRCC cases. Furthermore, PFKFB3 knockdown suppressed cell glycolysis, proliferative rate and cell-cycle G1/S conversion in RCC cells. Importantly, in vivo experiments confirmed that PFKFB3 knockdown delayed tumor growth derived from the ACHN cell line.
Conclusions
Such results suggest that PFKFB3 is a key molecular player in RCC progression via mediating glycolysis / proliferation and provides a potential therapeutic target against RCC.
Collapse
|
3
|
Tongmee B, Ontawong A, Lailerd N, Mengamphan K, Amornlerdpisan D. Anti-inflammatory effects and enhancing immune response of freshwater hybrid catfish oil in RAW264.7 cells. Exp Ther Med 2021; 22:1223. [PMID: 34603520 PMCID: PMC8453337 DOI: 10.3892/etm.2021.10657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
The present study assessed the effect of freshwater hybrid catfish oil (FFO) on the inflammatory status of lipopolysaccharide (LPS)-stimulated RAW264.7 cells and investigated the underlying mechanisms. RAW264.7 cells were supplemented with various concentrations [0.125-2% in 0.5% propylene glycol (v/v)] of FFO with or without LPS (1 µg/ml) for 24 h. Inflammatory cytokines and mediators were quantified using ELISA and reverse transcription-quantitative PCR. The results revealed that FFO treatment inhibited the secretion and mRNA expression of the pro-inflammatory cytokines IL-6, IL-1β, TNF-α. In line with this, FFO suppressed the expression and secretion of the inflammatory mediators cyclooxygenase-2 and prostaglandin E2. FFO also reduced apoptotic body formation and DNA damage. Correspondingly, FFO enhanced the immune response by modulating the cell cycle regulators p53, cyclin D2 and cyclin E2. Accordingly, FFO may be developed as a nutraceutical product to prevent inflammation.
Collapse
Affiliation(s)
- Bussarin Tongmee
- Agricultural Interdisciplinary Program, Faculty of Engineer and Agro-Industry, Maejo University, Chiang Mai 50290, Thailand
| | - Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Narissara Lailerd
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kriangsak Mengamphan
- Center of Excellence in Agricultural Innovation for Graduate Entrepreneur, Maejo University, Chiang Mai 50290, Thailand
| | - Doungporn Amornlerdpisan
- Center of Excellence in Agricultural Innovation for Graduate Entrepreneur, Maejo University, Chiang Mai 50290, Thailand
- Faculty of Fisheries Technology and Aquatic Resources, Maejo University, Chiang Mai 50290, Thailand
| |
Collapse
|
4
|
Zeng X, Zhong M, Yang Y, Wang Z, Zhu Y. Down-regulation of RCC1 sensitizes immunotherapy by up-regulating PD-L1 via p27 kip1 /CDK4 axis in non-small cell lung cancer. J Cell Mol Med 2021; 25:4136-4147. [PMID: 33630417 PMCID: PMC8051708 DOI: 10.1111/jcmm.16383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/29/2020] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, although Immune Checkpoint Inhibitors (ICIs) significantly improves survival both in local advanced stage and advanced stage of non-small cell lung cancer (NSCLC), the objective response rate of ICI monotherapy is still only about 20%. Thus, to identify the mechanisms of ICI resistance is critical to increase the efficacy of ICI treatments. By bioinformatics analysis, we found that the expression of regulator of chromosome condensation 1 (RCC1) in lung adenocarcinoma was significantly higher than that in normal lung tissue in TCGA and Oncomine databases. The survival analysis showed that high expression RCC1 was associated with the poor prognosis of NSCLC. And the expression of RCC1 was inversely related to the number of immune cell infiltration. In vitro, knockdown of RCC1 not only significantly inhibited the proliferation of lung adenocarcinoma cells but also increased the expression levels of p27kip1 and PD-L1, and decreased the expression level of CDK4 and p-Rb. In vivo, knockdown of RCC1 significantly slowed down the growth rate of tumour, and further reduced the volume and weight of tumour model after treated by PD-L1 monoclonal antibody. Therefore, RCC1 could up-regulate the expression level of PD-L1 by regulating p27kip1 /CDK4 pathway and decrease the resistance to ICIs. And this study might provide a new way to increase the efficacy of PD-L1 monoclonal antibody by inhibiting RCC1.
Collapse
Affiliation(s)
- Xiaozhu Zeng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Clinical Cancer Research Center, Chongqing, China
| | - Maoxi Zhong
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Clinical Cancer Research Center, Chongqing, China
| | - Yumeng Yang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Clinical Cancer Research Center, Chongqing, China
| | - Zhi Wang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Clinical Cancer Research Center, Chongqing, China
| | - Yuxi Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Clinical Cancer Research Center, Chongqing, China
| |
Collapse
|
5
|
Ng ZL, Siew J, Li J, Ji G, Huang M, Liao X, Yu S, Chew Y, Png CW, Zhang Y, Wen S, Yang H, Zhou Y, Long YC, Jiang ZH, Wu Q. PATZ1 (MAZR) Co-occupies Genomic Sites With p53 and Inhibits Liver Cancer Cell Proliferation via Regulating p27. Front Cell Dev Biol 2021; 9:586150. [PMID: 33598459 PMCID: PMC7882738 DOI: 10.3389/fcell.2021.586150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/13/2021] [Indexed: 01/05/2023] Open
Abstract
Liver cancer is the third most common cause of cancer death in the world. POZ/BTB and AT-hook-containing zinc finger protein 1 (PATZ1/MAZR) is a transcription factor associated with various cancers. However, the role of PATZ1 in cancer progression remains controversial largely due to lack of genome-wide studies. Here we report that PATZ1 regulates cell proliferation by directly regulating CDKN1B (p27) in hepatocellular carcinoma cells. Our PATZ1 ChIP-seq and gene expression microarray analyses revealed that PATZ1 is strongly related to cancer signatures and cellular proliferation. We further discovered that PATZ1 depletion led to an increased rate of colony formation, elevated Ki-67 expression and greater S phase entry. Importantly, the increased cancer cell proliferation was accompanied with suppressed expression of the cyclin-dependent kinase inhibitor CDKN1B. Consistently, we found that PATZ1 binds to the genomic loci flanking the transcriptional start site of CDKN1B and positively regulates its transcription. Notably, we demonstrated that PATZ1 is a p53 partner and p53 is essential for CDKN1B regulation. In conclusion, our study provides novel mechanistic insights into the inhibitory role of PATZ1 in liver cancer progression, thereby yielding a promising therapeutic intervention to alleviate tumor burden.
Collapse
Affiliation(s)
- Zhen Long Ng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jiamin Siew
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jia Li
- Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore, Singapore
| | - Guanxu Ji
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| | - Min Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| | - Xiaohua Liao
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| | - Sue Yu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuanyuan Chew
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chin Wen Png
- Department of Microbiology, Immunology Programme, Life Sciences Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yongliang Zhang
- Department of Microbiology, Immunology Programme, Life Sciences Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shijun Wen
- Medicinal Chemistry and Molecular Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Henry Yang
- Cancer Science Institute of Singapore, Centre for Translational Medicine, Singapore, Singapore
| | - Yiting Zhou
- The Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Chau Long
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| | - Qiang Wu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| |
Collapse
|
6
|
Policing Cancer: Vitamin D Arrests the Cell Cycle. Int J Mol Sci 2020; 21:ijms21239296. [PMID: 33291213 PMCID: PMC7731034 DOI: 10.3390/ijms21239296] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022] Open
Abstract
Vitamin D is a steroid hormone crucial for bone mineral metabolism. In addition, vitamin D has pleiotropic actions in the body, including anti-cancer actions. These anti-cancer properties observed within in vitro studies frequently report the reduction of cell proliferation by interruption of the cell cycle by the direct alteration of cell cycle regulators which induce cell cycle arrest. The most recurrent reported mode of cell cycle arrest by vitamin D is at the G1/G0 phase of the cell cycle. This arrest is mediated by p21 and p27 upregulation, which results in suppression of cyclin D and E activity which leads to G1/G0 arrest. In addition, vitamin D treatments within in vitro cell lines have observed a reduced C-MYC expression and increased retinoblastoma protein levels that also result in G1/G0 arrest. In contrast, G2/M arrest is reported rarely within in vitro studies, and the mechanisms of this arrest are poorly described. Although the relationship of epigenetics on vitamin D metabolism is acknowledged, studies exploring a direct relationship to cell cycle perturbation is limited. In this review, we examine in vitro evidence of vitamin D and vitamin D metabolites directly influencing cell cycle regulators and inducing cell cycle arrest in cancer cell lines.
Collapse
|
7
|
He L, Guo S, Zhu T, Chen C, Xu K. Down-Regulation of the Mammalian Target of Rapamycin (mTOR) Pathway Mediates the Effects of the Paeonol-Platinum(II) Complex in Human Thyroid Carcinoma Cells and Mouse SW1736 Tumor Xenografts. Med Sci Monit 2020; 26:e922561. [PMID: 32594094 PMCID: PMC7341900 DOI: 10.12659/msm.922561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background This study aimed to investigate the effects of the paeonol-platinum(II) (PL-Pt[II]) complex on SW1736 human anaplastic thyroid carcinoma cell line and the BHP7-13 human thyroid papillary carcinoma cell line in vitro and on mouse SW1736 tumor xenografts in vivo. Material/Methods The cytotoxic effects of the PL-Pt(II) complex on SW1736 cells and BHP7-13 cells was measured using the MTT assay. Western blot measured the expression levels of cyclins, cell apoptotic proteins, and signaling proteins. DNA content and apoptosis were detected by flow cytometry. SW1736 cell thyroid tumor xenografts were established in mice followed by treatment with the PL-Pt(II) complex. Results Treatment of the SW1736 and BHP7-13 cells with the PL-Pt(II) complex reduced cell proliferation in a dose-dependent manner, with an IC50 of 1.25 μM and 1.0 μM, respectively, and increased the cell fraction in G0/G1phase, inhibited p53, cyclin D1, promoted p27 and p21 expression, and significantly increased the sub-G1 fraction. Treatment with the PL-Pt(II) complex increased caspase-3 degradation, reduced the expression of p-4EBP1, p-4E-BP1 and p-S6, and reduced the expression of p-ERK1/2 and p-AKT. Treatment with the PL-Pt(II) complex reduced the volume of the SW1736 mouse tumor xenografts on day 14 and day 21, and reduced AKT phosphorylation and S6 protein expression and increased degradation of caspase-3. Conclusions The cytotoxic effects of the PL-Pt(II) complex in human thyroid carcinoma cells, including activation of apoptosis and an increased sub-G1 cell fraction of the cell cycle, were mediated by down-regulation of the mTOR pathway.
Collapse
Affiliation(s)
- Ling He
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Song Guo
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Taiyang Zhu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Chen Chen
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Kun Xu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
8
|
Application of co-culture technology of epithelial type cells and mesenchymal type cells using nanopatterned structures. PLoS One 2020; 15:e0232899. [PMID: 32392240 PMCID: PMC7213697 DOI: 10.1371/journal.pone.0232899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/23/2020] [Indexed: 01/23/2023] Open
Abstract
Various nanopatterning techniques have been developed to improve cell proliferation and differentiation efficiency. As we previously reported, nanopillars and pores are able to sustain human pluripotent stem cells and differentiate pancreatic cells. From this, the nanoscale patterns would be effective environment for the co-culturing of epithelial and mesenchymal cell types. Interestingly, the nanopatterning selectively reduced the proliferative rate of mesenchymal cells while increasing the expression of adhesion protein in epithelial type cells. Additionally, co-cultured cells on the nanopatterning were not negatively affected in terms of cell function metabolic ability or cell survival. This is in contrast to conventional co-culturing methods such as ultraviolet or chemical treatments. The nanopatterning appears to be an effective environment for mesenchymal co-cultures with typically low proliferative rates cells such as astrocytes, neurons, melanocytes, and fibroblasts without using potentially damaging treatments.
Collapse
|
9
|
Han JB, Huang ML, Li F, Yang R, Chen SM, Tao ZZ. MiR-214 Mediates Cell Proliferation and Apoptosis of Nasopharyngeal Carcinoma Through Targeting Both WWOX and PTEN. Cancer Biother Radiopharm 2020; 35:615-625. [PMID: 32101017 PMCID: PMC7578184 DOI: 10.1089/cbr.2019.2978] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background: This study aimed to investigate interactions between miR-214, PTEN, and WWOX and their effect on AKT signaling during the NPC progression. Nasopharyngeal carcinoma (NPC) was highly prevalent with poor prognosis among the patients. MiR-214 reported as an important NPC biomarker was associated with regulation of biological functions. Methods: 5–8F and 6–10B NPC cells were transfected with miR-214 inhibitor. MTT and colony formation assays were performed to assess cell proliferation. PI staining assay was performed to determine distribution of cell cycle. Annexin-V/PI staining assay was used to evaluate cell apoptosis in NPC. The effects of miR-214 inhibitor on the expression levels of PTEN, WWOX, AKT signaling pathway, cell-cycle-, and apoptosis-associated proteins were assessed by Western blotting or qRT-PCR assay. PTEN and WWOX were knocked down using the corresponding shRNA to investigate their effects on miR-214 inhibitor involved in proapoptosis and antiproliferation mechanisms in NPC. Results: Inhibition of miR-214 suppressed cell growth and induced apoptosis of 5–8F and 6–10B cells. MiR-214 regulated the expression of both PTEN and WWOX through targeting the 3′-UTR. Inhibition of miR-214 promoted WWOX and PTEN expression, inactivated AKT signaling pathway, and regulated cell-cycle- and apoptosis-associated proteins. Knockdown of PTEN or WWOX reversed effects of miR-214 inhibitor on AKT signaling, cell proliferation, and apoptosis. Conclusion: MiR-214 was suggested to induce cell proliferation and inhibit cell apoptosis of NPC through directly targeting both PTEN and WWOX, which provided a novel therapeutic target for clinical treatment of NPC.
Collapse
Affiliation(s)
- Ji-Bo Han
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Mao-Ling Huang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Fen Li
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
10
|
Shiraishi RD, Miyashita S, Yamashita M, Adachi T, Shimoda MM, Owa T, Hoshino M. Expression of transcription factors and signaling molecules in the cerebellar granule cell development. Gene Expr Patterns 2019; 34:119068. [PMID: 31437514 DOI: 10.1016/j.gep.2019.119068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/29/2019] [Accepted: 08/14/2019] [Indexed: 01/16/2023]
Abstract
Cerebellar granule cell precursors (GCPs) and granule cells (GCs) constitute a good model system to investigate proliferation of neural precursors and differentiation of neurons. During development, GCPs proliferate in the outer external granule cell layer (outer EGL) and then exit the cell cycle in the inner EGL to become GCs, which inwardly migrate to the inner granule cell layer (IGL). Misregulation of GCP proliferation or GC differentiation leads to maldevelopment of the cerebellum and the formation of a cerebellar tumor, medulloblastoma. Despite many efforts in this field, the mechanisms underlying GC development remain elusive. In this study, we performed detailed immunostaining in the developing cerebellum, with particular focus on GCPs and GCs, looking at several transcription factors, signaling molecules, cell cycle regulators, some of which are known to regulate neural development. Interestingly, we found distinct distribution patterns of certain proteins within the outer and inner EGL, suggesting the existence of subpopulations of GCPs and GCs in those layers. This study provides a basis for future studies on the cerebellar GC development and medulloblastoma.
Collapse
Affiliation(s)
- Ryo D Shiraishi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan; Department of NCNP Brain Function and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, TMDU, Tokyo, 113- 8510, Japan
| | - Sathoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Mariko Yamashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan; Department of NCNP Brain Function and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, TMDU, Tokyo, 113- 8510, Japan
| | - Toma Adachi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advance Science and Engineering, TWIns, Waseda University, Tokyo, 162-8480, Japan
| | - Mana M Shimoda
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advance Science and Engineering, TWIns, Waseda University, Tokyo, 162-8480, Japan
| | - Tomoo Owa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan.
| |
Collapse
|
11
|
Sutariya V, Tur J, Kelly S, Halasz K, Chapalamadugu KC, Nimbalkar R, Pathak YV, Weigel R, Daviau T, Webb T, Cacace J, Brotto M, Tipparaju SM. Nanodrug delivery platform for glucocorticoid use in skeletal muscle injury. Can J Physiol Pharmacol 2018; 96:681-689. [PMID: 29756463 DOI: 10.1139/cjpp-2017-0795] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glucocorticoids are utilized for their anti-inflammatory properties in the skeletal muscle and arthritis. However, the major drawback of use of glucocorticoids is that it leads to senescence and toxicity. Therefore, based on the idea that decreasing particle size allows for increased surface area and bioavailability of the drug, in the present study, we hypothesized that nanodelivery of dexamethasone will offer increased efficacy and decreased toxicity. The dexamethasone-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles were prepared using nanoprecipitation method. The morphological characteristics of the nanoparticles were studied under scanning electron microscope. The particle size of nanoparticles was 217.5 ± 19.99 nm with polydispersity index of 0.14 ± 0.07. The nanoparticles encapsulation efficiency was 34.57% ± 1.99% with in vitro drug release profile exhibiting a sustained release pattern over 10 days. We identified improved skeletal muscle myoblast performance with improved closure of the wound along with increased cell viability at 10 nmol/L nano-dexamethasone-PLGA. However, dexamethasone solution (1 μmol/L) was injurious to cells because the migration efficiency was decreased. In addition, the use of dexamethasone nanoparticles decreased lipopolysaccharide-induced lactate dehydrogenase release compared with dexamethasone solution. Taken together, the present study clearly demonstrates that delivery of PLGA-dexamethasone nanoparticles to the skeletal muscle cells is beneficial for treating inflammation and skeletal muscle function.
Collapse
Affiliation(s)
- Vijaykumar Sutariya
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Jared Tur
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Shannon Kelly
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Kathleen Halasz
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Kalyan C Chapalamadugu
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Rohini Nimbalkar
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yashwant V Pathak
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Robert Weigel
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Todd Daviau
- b CoreRx, Inc., 14205 Myerlake Cir, Clearwater, FL 33760, USA
| | - Travis Webb
- b CoreRx, Inc., 14205 Myerlake Cir, Clearwater, FL 33760, USA
| | - Janice Cacace
- b CoreRx, Inc., 14205 Myerlake Cir, Clearwater, FL 33760, USA
| | - Marco Brotto
- c Bone-Muscle Collaborative Sciences, College of Nursing and Health Innovation, University of Texas at Arlington, TX 76010, USA
| | - Srinivas M Tipparaju
- a Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
12
|
Chen X, Chen J. miR-3188 Regulates Cell Proliferation, Apoptosis, and Migration in Breast Cancer by Targeting TUSC5 and Regulating the p38 MAPK Signaling Pathway. Oncol Res 2018; 26:363-372. [PMID: 28560951 PMCID: PMC7844837 DOI: 10.3727/096504017x14953948675421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This study intended to investigate the effects of miR-3188 on breast cancer and to reveal the possible molecular mechanisms. miR-3188 was upregulated and TUSC5 was downregulated in breast cancer tissues and MCF-7 cells compared to normal tissue and MCF-10 cells. After MCF-7 cells were transfected with miR-3188 inhibitor, cell proliferation and migration were inhibited, whereas apoptosis was promoted. Luciferase reporter assay suggested that TUSC5 was a target gene of miR-3188. In addition, miR-3188 overexpression increased the p-p38 expression, while miR-3188 suppression decreased the p-p38 expression significantly. miR-3188 regulated breast cancer progression via the p38 MAPK signaling pathway. In conclusion, miR-3188 affects breast cancer cell proliferation, apoptosis, and migration by targeting TUSC5 and activating the p38 MAPK signaling pathway. miR-3188 may serve as a potential therapeutic agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xiaowen Chen
- *Department of Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, P.R. China
| | - Jianli Chen
- †The Third Department of Medical Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China
| |
Collapse
|
13
|
Peng F, Li Q, Sun JY, Luo Y, Chen M, Bao Y. PFKFB3 is involved in breast cancer proliferation, migration, invasion and angiogenesis. Int J Oncol 2018; 52:945-954. [PMID: 29393396 DOI: 10.3892/ijo.2018.4257] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/29/2017] [Indexed: 11/05/2022] Open
Abstract
6-Phosphofructo 2-kinase/fructose 2, 6-bisphosphatase 3 (PFKFB3) has been reported to be overexpressed in human cancer tissues and to promote the proliferation and migration of cancer cells. However, the role of PFKFB3 in the progression and prognosis of breast cancer is not yet fully understood. In the present study, we investigated the specific role of PFKFB3 in breast cancer progression and its preliminary mechanisms of action. We first used an immunohistochemistry assay to determine that PFKFB3 was highly expressed in breast cancer tissues and that this high level of expression was involved in the poor overall survival of patients with breast cancer. In addition, the suppression of PFKFB3 by lentiviruses carrying shRNA against PFKFB3 (shPFKFB3) subsequently inhibited breast cancer cell (MDA-MB-231 and MDA-MB-468) proliferation, migration and invasion, and induced cell cycle G1 and S phase arrest in vitro. Moreover, PFKFB3 inhibition decreased p-AKT and increased p27 expression levels in breast cancer cells. Furthermore, PFKFB3 suppression inhibited breast cancer cell tumor xenograft growth in nude mice. We also found that PFKFB3 inhibition suppressed vascular endothelial growth factor α (VEGFα) protein expression and inhibited the angiogenic activity of human umbilical vein endothelial cells (HUVECs). On the whole, our results indicate that PFKFB3 is involved in the proliferation, migration, invasion and angiogenesis of breast cancer.
Collapse
Affiliation(s)
- Fang Peng
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qiang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jia-Yuan Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Ying Luo
- Department of Clinical Laboratory, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ming Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, P.R. China
| | - Yong Bao
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
14
|
Illes A, Opper B, Reglodi D, Kerenyi M, Czetany P, Boronkai A, Schafer E, Toth G, Fabian E, Horvath G. Effects of pituitary adenylate cyclase activating polypeptide on small intestinal INT 407 cells. Neuropeptides 2017; 65:106-113. [PMID: 28698051 DOI: 10.1016/j.npep.2017.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 12/13/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is an endogenous neuropeptide having a widespread distribution both in the nervous system and peripheral organs including the gastrointestinal tract. It has been shown to exert actions on intestinal functions, mainly affecting glandular secretion and motility. PACAP has several different effects on cell survival depending on the cell type and the applied stimulus. Its influences on small intestinal epithelial cells are not yet elucidated, therefore the aim of the present study was to investigate the effects of PACAP on intestinal epithelial cells having high turnover (INT 407) against different harmful stimuli, such as oxidative stress, in vitro hypoxia and gamma radiation. We tested the effect of PACAP on proliferation and cell survival using MTT assay. Moreover, various cancer-related factors were evaluated by oncology array. PACAP did not influence the proliferation rate of INT 407 cells. Its cell survival-enhancing effect could be detected against oxidative stress, but not against in vitro hypoxia or gamma irradiation. Clonogenic survival assay was performed to analyze the effect of PACAP on clonogenic potential of cells exposed to gamma radiation. Surprisingly, PACAP enhanced the clone-forming ability decrease induced by irradiation. Western blot analysis of ERK1/2 phosphorylation was performed in order to obtain further information on the molecular background. Our data showed phospho-ERK1/2 suppression of PACAP in irradiated cells. Furthermore, the role of endogenous PACAP against oxidative stress was also investigated performing ADCYAP1 small interfering RNA transfection. We found significant difference in the cell vulnerability between cells undergoing silencing and cells without transfection suggesting the protective role of the endogenously present PACAP against oxidative stress in INT 407 cells. In summary, PACAP seems to be able to exert contradictory effects in INT 407 cells depending on the applied stressor, suggesting its regulatory role in the cellular household.
Collapse
Affiliation(s)
- A Illes
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs, Hungary; 1st Department of Internal Medicine, University of Pecs, Hungary
| | - B Opper
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs, Hungary
| | - D Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs, Hungary
| | - M Kerenyi
- Medical Microbiology and Immunology, University of Pecs, Hungary
| | - P Czetany
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs, Hungary
| | - A Boronkai
- Oncotherapy, University of Pecs, Hungary
| | - E Schafer
- Department of Gastroenterology, Medical Centre, Hungarian Defence Forces, Budapest, Hungary
| | - G Toth
- Medical Chemistry, University of Szeged, Hungary
| | - E Fabian
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs, Hungary
| | - G Horvath
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs, Hungary.
| |
Collapse
|
15
|
Niu J, Zhao X, Liu Q, Yang J. Knockdown of MSI1 inhibited the cell proliferation of human osteosarcoma cells by targeting p21 and p27. Oncol Lett 2017; 14:5271-5278. [PMID: 29113163 PMCID: PMC5661380 DOI: 10.3892/ol.2017.6870] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 07/13/2017] [Indexed: 11/18/2022] Open
Abstract
Osteosarcoma is the most common type of primary bone cancer in children and adolescents, but its mechanism remains unclear. Musashi RNA-binding protein 1 (MSI1) is highly expressed in certain cancer types and functions as a putative progenitor/stem cell marker. In the present study, it was demonstrated that MSI1 expression in osteosarcoma tissue was higher compared with in the paraneoplastic tissue samples. Knockdown of MSI1 using shRNA in MG-63 and HOS cells inhibited cell proliferation in vitro and tumor formation in vivo, suggesting that MSI1 serves an essential role in osteosarcomagenesis. Further investigations demonstrated that the knockdown of MSI1 leads to the cell cycle arrest at G0/G1 phase, and the upregulation of p21 and p27 protein expression in osteosarcoma cells. Additionally, luciferase assays demonstrated that MSI1 can bind to the 3′ untranslated regions of p21 and p27 mRNA. In conclusion, the results of the present study suggest that the knockdown of MSI11 can suppress cell proliferation of osteosarcoma by targeting p21 and p27 and subsequently inhibiting cell cycle progression.
Collapse
Affiliation(s)
- Jianbing Niu
- Department of Bone and Joint Surgery, Shandong Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Xiulian Zhao
- Department of Kidney and Chinese Medicine, Shandong Jinxiang County People's Hospital, Jinxiang, Shandong 272200, P.R. China
| | - Qingsheng Liu
- Department of Bone and Joint Surgery, Shandong Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Jinsan Yang
- Department of Bone and Joint Surgery, Shandong Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
16
|
Ma X, Zhao J, Yang F, Liu H, Qi W. Ubiquitin conjugating enzyme E2 L3 promoted tumor growth of NSCLC through accelerating p27kip1 ubiquitination and degradation. Oncotarget 2017; 8:84193-84203. [PMID: 29137415 PMCID: PMC5663587 DOI: 10.18632/oncotarget.20449] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/26/2017] [Indexed: 02/03/2023] Open
Abstract
The molecular pathogenesis of human lung cancer has not been completely clarified. Here, we reported that UBE2L3, a member of the ubiquitin-conjugating enzymes (E2s), were overexpressed in non-small-cell lung cancer (NSCLC) tissues compared with the non-tumor tissues. High expression of UBE2L3 was correlated with advanced tumor stage and adverse outcomes. Knockdown of UBE2L3 inhibited NSCLC cell growth while ectopic expression of UBE2L3 promoted NSCLC cell growth in a cell cycle dependent manner. The results of subcutaneous tumor xenograft studies revealed that knockdown of UBE2L3 attenuated the in vivo tumor growth. Mechanistically, we observed that UBE2L3 could interact with F-box protein Skp2, a member of the SCF (Skp2) ubiquitin ligase complex, and thus promoted the ubiquitination and proteasomal degradation of p27kip1. Furthermore, NSCLC cases with high level of UBE2L3 and low level of p27kip1 had worst prognosis, suggesting that combination of UBE2L3 and p27kip1 is a more powerful prognostic marker for NSCLC patients. Taken together, the current study presented a novel marker for predicting prognosis and a potential therapeutic target for NSCLC patients.
Collapse
Affiliation(s)
- Xingjie Ma
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Junjie Zhao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fan Yang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Haitao Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Weibo Qi
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
17
|
Linkages between changes in the 3D organization of the genome and transcription during myotube differentiation in vitro. Skelet Muscle 2017; 7:5. [PMID: 28381300 PMCID: PMC5382473 DOI: 10.1186/s13395-017-0122-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/16/2017] [Indexed: 12/21/2022] Open
Abstract
Background The spatial organization of eukaryotic genomes facilitates and reflects the underlying nuclear processes that are occurring in the cell. As such, the spatial organization of a genome represents a window on the genome biology that enables analysis of the nuclear regulatory processes that contribute to mammalian development. Methods In this study, Hi-C and RNA-seq were used to capture the genome organization and transcriptome in mouse muscle progenitor cells (C2C12 myoblasts) before and after differentiation to myotubes, in the presence or absence of the cytidine analogue AraC. Results We observed significant local and global developmental changes despite high levels of correlation between the myotubes and myoblast genomes. Notably, the genes that exhibited the greatest variation in transcript levels between the different developmental stages were predominately within the euchromatic compartment. There was significant re-structuring and changes in the expression of replication-dependent histone variants within the HIST1 locus. Finally, treating terminally differentiated myotubes with AraC resulted in additional changes to the transcriptome and 3D genome organization of sets of genes that were all involved in pyroptosis. Conclusions Collectively, our results provide evidence for muscle cell-specific responses to developmental and environmental stimuli mediated through a chromatin structure mechanism. Electronic supplementary material The online version of this article (doi:10.1186/s13395-017-0122-1) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Zhan P, Xi G, Zhang B, Wu Y, Liu H, Liu Y, Xu W, Zhu Q, Cai F, Zhou Z, Miu Y, Wang X, Jin J, Li Q, Lv T, Song Y. NCAPG2 promotes tumour proliferation by regulating G2/M phase and associates with poor prognosis in lung adenocarcinoma. J Cell Mol Med 2017; 21:665-676. [PMID: 27862966 PMCID: PMC5345611 DOI: 10.1111/jcmm.13010] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/19/2016] [Indexed: 12/22/2022] Open
Abstract
NCAPG2 is a component of the condensin II complex and contributes to chromosome segregation via microtubule-kinetochore attachment during mitosis. It is well known that NCAPG2 plays a critical role in cell mitosis; however, the role of altered NCAPG2 expression and its transcriptional regulatory function in cancer development remains mostly unknown. Here, for the first time we reported that NCAPG2 was evidently increased in non-small cell lung cancer tissues compared to adjacent normal lung tissues. Clinicopathological data analysis showed that NCAPG2 overexpression was significantly correlated with lymph node metastasis and pathologic-Tumour Nodes Metastasen stages, and was an independent prognostic factor in lung adenocarcinoma patients. Moreover, siRNA-mediated knockdown of NCAPG2 could inhibit tumour cell growth of lung adenocarcinoma cells (A549 and H1299) in vitro and could significantly lead to cell cycle arrest in the G2 phase. Furthermore, we found that NCAPG2 silencing significantly decreased the expression levels of G2/M phase cell cycle-related protein expressions (Cyclin B1, Cdc2) and increased the expression levels of p27 and p21 through Western blot analysis. Taken together, we demonstrated that increased NCAPG2 expression could regulate cell proliferation and identified as a poor prognostic biomarker in lung adenocarcinoma.
Collapse
Affiliation(s)
- Ping Zhan
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
- Department of Respiratory MedicineNanjing Chest HospitalMedical School of Southeast UniversityNanjingChina
| | - Guang‐min Xi
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Bin Zhang
- Department of GastroenterologyThe Affiliated Drum Tower Hospital of Nanjing University, Medical SchoolNanjingJiangsuChina
| | - Ying Wu
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Hong‐bing Liu
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Ya‐fang Liu
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Wu‐jian Xu
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Qingqing Zhu
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Feng Cai
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Ze‐jun Zhou
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Ying‐ying Miu
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Xiao‐xia Wang
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Jia‐jia Jin
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Qian Li
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Tang‐feng Lv
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| | - Yong Song
- Department of Respiratory MedicineJinling HospitalNanjing University School of MedicineNanjingChina
| |
Collapse
|
19
|
Abstract
Uterine carcinosarcomas (UCS) are rare (3-4%) but highly aggressive, accounting for a disproportionately high (16.4%) mortality among uterine malignancies. Transforming growth factor beta (TGFβ) is a multifunctional cytokine that regulates important cellular processes including epithelial-mesenchymal transition (EMT). Existence of biphasic elements and a report demonstrating amplification of TGFβ at 19q13.1 prompted us to investigate the role of TGFβ signaling in UCS. Here we demonstrated the components of TGFβ pathway are expressed and functional in UCS. TGFβ-I induced significant Smad2/3 phosphorylation, migration and EMT responses in UCS cell lines which could be attenuated by the TGFβ receptor I (TGFβR-I) or TGFβ receptor I/II (TGFβR-I/II) inhibitor developed by Eli Lilly and company. Importantly, TGFβ-I induced proliferation was c-Myc dependent, likely through activation of cell cycle. c-Myc was induced by nuclear translocation of nuclear factor of activated T cells (NFAT-1) in response to TGFβ-I. Inhibition of NFAT-1 or TGFβR-I blocked c-Myc induction, cell cycle progression and proliferation in UCS. In corroboration, mRNA levels of c-Myc were elevated in recurrent versus the non-recurrent UCS patient samples. Interestingly, in the absence of exogenous TGFβ the TGFβR-I/II inhibitor enhanced proliferation likely through non-Smad pathways. Thus, inhibition of TGFβR-I could be efficacious in treatment of UCS.
Collapse
|
20
|
Wang D, Han S, Peng R, Jiao C, Wang X, Yang X, Yang R, Li X. Depletion of histone demethylase KDM5B inhibits cell proliferation of hepatocellular carcinoma by regulation of cell cycle checkpoint proteins p15 and p27. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:37. [PMID: 26911146 PMCID: PMC4766611 DOI: 10.1186/s13046-016-0311-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/19/2016] [Indexed: 01/12/2023]
Abstract
Background KDM5B is a jmjc domain-containing histone demethylase which remove tri-, di-, and monomethyl groups from histone H3 lysine 4 (H3K4). KDM5B has been determined as an oncogene in many malignancies. However, its expression and role in hepatocellular carcinoma (HCC) remain unknown. Methods We detected the expression of KDM5B in HCC tissues and cell lines. Cell proliferation was performed to reveal the role of KDM5B depletion on HCC cells both in vivo and in vitro. Flow cytometry was used to analyze the cell cycle and chip analysis was conducted to determine the direct target of KDM5B. Results KDM5B is frequently up-regulated in HCC specimens compared with adjacent normal tissues and its expression level was significantly correlated with tumor size, TNM stage, and Edmondson grade. Moreover, Kaplan-Meier survival analysis showed that patients with high levels of KDM5B expression had a relatively poor prognosis. Knockdown of KDM5B notably inhibits HCC cell proliferation both in vivo and in vitro via arresting the cell cycle at G1/S phase partly through up-regulation of p15 and p27. Further molecular mechanism study indicates that silencing of KDM5B promotes p15 and p27 expression by increasing histone H3K4 trimethylation in their promoters. Conclusions KDM5B could be a potentially therapeutic target, which provides a rationale for the development of histone demethylase inhibitors as a strategy against HCC. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0311-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dong Wang
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Sheng Han
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Rui Peng
- Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, China
| | - Chenyu Jiao
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xing Wang
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xinxiang Yang
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Renjie Yang
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xiangcheng Li
- Liver Transplantation Center, Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
21
|
Xu Y, Li C, Sun J, Li J, Gu X, Xu W. Antitumor effects of galectin-3 inhibition in human renal carcinoma cells. Exp Biol Med (Maywood) 2016; 241:1365-73. [PMID: 26846978 DOI: 10.1177/1535370216630839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/11/2016] [Indexed: 11/17/2022] Open
Abstract
Galectins are thought to be prognosticators for survival in renal cell cancer. However, the biological activity of galectin-3 (Gal-3) in renal carcinoma cells is still debated. In this study, immunohistochemical staining confirmed a high expression of Gal-3 in tumor tissue from renal cell carcinoma. Critically, Gal-3 expression was related to tumor cell differentiation. Consistent with Gal-3 expression in renal cell cancer, strong expression of Gal-3 was also observed in several renal tumor cell lines but not in normal renal cells. A Gal-3 high-expression cell line Caki-1 was chosen to study the biological activity of Gal-3. Using short hairpin RNA method, Gal-3 expression in Caki-1 cells was knocked down. We evidenced that Gal-3 knockdown inhibited cell proliferation and invasion, induced Caspase-3-dependent apoptosis and arrested cell cycle at G1 phase. Mechanically, Cyclin D1 expression decreased, but p27 increased after Gal-3 knockdown. Taken together, these results suggest that Gal-3 is related to the development of renal cell cancer and could serve as a target to therapy renal cell cancer.
Collapse
Affiliation(s)
- Yangyang Xu
- Cancer Hospital of Harbin Medical University, Harbin, 150000, China
| | - Changfu Li
- Cancer Hospital of Harbin Medical University, Harbin, 150000, China
| | - Jiahang Sun
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jingshu Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xin Gu
- Cancer Hospital of Harbin Medical University, Harbin, 150000, China
| | - Wanhai Xu
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
22
|
Lewandowska U, Fichna J, Gorlach S. Enhancement of anticancer potential of polyphenols by covalent modifications. Biochem Pharmacol 2016; 109:1-13. [PMID: 26776305 DOI: 10.1016/j.bcp.2015.12.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 12/23/2015] [Indexed: 12/23/2022]
Abstract
As evidenced by a growing number of respective clinical trials, a promising and increasingly valued approach to cancer prevention is chemoprevention which is based on using synthetic, semisynthetic, or natural compounds with the aim of preventing, delaying, arresting, or reversing carcinogenesis. Research carried out in the last two decades indicates that natural polyphenols isolated from plants (as well as their derivatives and synthetic analogs) exhibit pleiotropic actions toward cancer cells and therefore they could be used in both cancer prevention and therapy. This review discusses selected covalent modifications of polyphenols as a means for increasing their anticancer potential in relation to the parent compounds. The modifications include hydroxylation, methylation, acylation, and galloylation, among others. They were demonstrated to enhance cytotoxic, pro-oxidant, antiproliferative, proapoptotic, proautophagic, and antimigratory activities of phenolics toward various cancer cell lines in vitro. Importantly, some derivatives proved to suppress tumor growth and metastasis in animal models more strongly than the parent compounds. Some of the above-mentioned covalent modifications were also shown to increase absorption and tissue distribution of tested phenolic compounds in vivo. Anticancer clinical trials with polyphenol derivatives therefore seem warranted.
Collapse
Affiliation(s)
| | - Jakub Fichna
- Department of Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Sylwia Gorlach
- Department of Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
23
|
Liu X, Pan L, Liang J, Li J, Wu S. Cryptotanshinone inhibits proliferation and induces apoptosis via mitochondria-derived reactive oxygen species involving FOXO1 in estrogen receptor-negative breast cancer Bcap37 cells. RSC Adv 2016. [DOI: 10.1039/c5ra22523j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cryptotanshinone inhibits proliferation and induces apoptosis of the estrogen receptor-negative breast cancer Bcap37 cells via FOXO1 inhibition and ROS-mediated PI3K/AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Xiaoman Liu
- Research Center of Siyuan Natural Pharmacy and Biotoxicology
- College of Life Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Lili Pan
- Research Center of Siyuan Natural Pharmacy and Biotoxicology
- College of Life Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Junling Liang
- Research Center of Siyuan Natural Pharmacy and Biotoxicology
- College of Life Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Jinhui Li
- Institute of Agrobiology and Environmental Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Shihua Wu
- Research Center of Siyuan Natural Pharmacy and Biotoxicology
- College of Life Sciences
- Zhejiang University
- Hangzhou 310058
- China
| |
Collapse
|
24
|
Jain M, Singh A, Singh V, Maurya P, Barthwal MK. Gingerol Inhibits Serum-Induced Vascular Smooth Muscle Cell Proliferation and Injury-Induced Neointimal Hyperplasia by Suppressing p38 MAPK Activation. J Cardiovasc Pharmacol Ther 2015; 21:187-200. [DOI: 10.1177/1074248415598003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 06/08/2015] [Indexed: 01/07/2023]
Abstract
Purpose: Gingerol inhibits growth of cancerous cells; however, its role in vascular smooth muscle cell (VSMC) proliferation is not known. The present study investigated the effect of gingerol on VSMC proliferation in cell culture and during neointima formation after balloon injury. Method and Results: Rat VSMCs or carotid arteries were harvested at 15 minutes, 30 minutes, 1, 6, 12, and 24 hours of fetal bovine serum (FBS; 10%) stimulation or balloon injury, respectively. Gingerol prevented FBS (10%)-induced proliferation of VSMCs in a dose-dependent manner (50 μmol/L-400 μmol/L). The FBS-induced proliferating cell nuclear antigen (PCNA) upregulation and p27Kip1 downregulation were also attenuated in gingerol (200 μmol/L) pretreated cells. Fetal bovine serum-induced p38 mitogen-activated protein kinase (MAPK) activation, PCNA upregulation, and p27Kip1 downregulation were abrogated in gingerol (200 μmol/L) and p38 MAPK inhibitor (SB203580, 10 μmol/L) pretreated cells. Balloon injury induced time-dependent p38 MAPK activation in the carotid artery. Pretreatment with gingerol (200 μmol/L) significantly attenuated injury-induced p38 MAPK activation, PCNA upregulation, and p27Kip1 downregulation. After 14 days of balloon injury, intimal thickening, neointimal proliferation, and endothelial dysfunction were significantly prevented in gingerol pretreated arteries. In isolated organ bath studies, gingerol (30 nmol/L-300 μmol/L) inhibited phenylephrine-induced contractions and induced dose-dependent relaxation of rat thoracic aortic rings in a partially endothelium-dependent manner. Conclusion: Gingerol prevented FBS-induced VSMC proliferation and balloon injury-induced neointima formation by regulating p38 MAPK. Vasodilator effect of gingerol observed in the thoracic aorta was partially endothelium dependent. Gingerol is thus proposed as an attractive agent for modulating VSMC proliferation, vascular reactivity, and progression of vascular proliferative diseases.
Collapse
Affiliation(s)
- Manish Jain
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Ankita Singh
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Vishal Singh
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Preeti Maurya
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| |
Collapse
|
25
|
CHEN XIAODONG, DENG MIN, MA LI, ZHOU JIANSHENG, XIAO YUZHOU, ZHOU XINSHE, ZHANG CHANGCHUN, WU MIN. Inhibitory effects of forkhead box L1 gene on osteosarcoma growth through the induction of cell cycle arrest and apoptosis. Oncol Rep 2015; 34:265-71. [DOI: 10.3892/or.2015.3969] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/19/2015] [Indexed: 11/05/2022] Open
|
26
|
Lin CH, Li HY, Lee YC, Calkins MJ, Lee KH, Yang CN, Lu PJ. Landscape of Pin1 in the cell cycle. Exp Biol Med (Maywood) 2015; 240:403-8. [PMID: 25662955 DOI: 10.1177/1535370215570829] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pin1 is a peptidyl-prolyl isomerase which plays a critical role in many diseases including cancer and Alzheimer's disease. The essential role of Pin1 is to affect stability, localization or function of phosphoproteins by catalyzing structural changes. Among the collection of Pin1 substrates, many have been shown to be involved in regulating cell cycle progression. The cell cycle disorder caused by dysregulation of these substrates is believed to be a common phenomenon in cancer. A number of recent studies have revealed possible functions of several important Pin1-binding cell cycle regulators. Investigating the involvement of Pin1 in the cell cycle may assist in the development of future cancer therapeutics. In this review, we summarize current knowledge regarding the network of Pin1 substrates and Pin1 regulators in cell cycle progression. In G1/S progression, cyclin D1, RB, p53, p27, and cyclin E are all well-known cell cycle regulators that are modulated by Pin1. During G2/M transition, our lab has shown that Aurora A suppresses Pin1 activity through phosphorylation at Ser16 and cooperates with hBora to modulate G2/M transition. We conclude that Pin1 may be thought of as a molecular timer which modulates cell cycle progression networks.
Collapse
Affiliation(s)
- Cheng-Han Lin
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan 704, Taiwan
| | - Hao-Yi Li
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan 704, Taiwan
| | - Yu-Cheng Lee
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan 704, Taiwan
| | - Marcus J Calkins
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan 704, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 115, Taiwan
| | - Chia-Ning Yang
- Institute of Biotechnology, National University of Kaohsiung, 811, Kaohsiung, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, Medical College, National Cheng Kung University, Tainan 704, Taiwan
| |
Collapse
|
27
|
Wang HC, Tseng YH, Wu HR, Chu FH, Kuo YH, Wang SY. Anti-proliferation Effect on Human Breast Cancer Cells via Inhibition of pRb Phosphorylation by Taiwanin E Isolated from Eleutherococcus trifoliatus. Nat Prod Commun 2014. [DOI: 10.1177/1934578x1400900921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Eleutherococcus trifoliatus has been used as a folk medicine since ancient times, especially as refreshing qi medicines. In our current study, taiwanin E, which possesses strong cytotoxicity, was isolated from the branches of E. trifoliatus by using a bioactivity guided fractionation procedure. Taiwanin E presented a potent anti-proliferation activity on the growth of a human breast adenocarcinoma cell line (MCF-7), with an IC50 value for cytotoxicity of 1.47 μM. Cell cycle analysis revealed that the proportion of cells in the G0/G1 phase increased in a dose-dependent manner (from 79.4% to 90.2%) after 48 h exposure to taiwanin E at a dosage range from 0.5 to 4μM. After treatment with taiwanin E, phosphorylation of retinoblastoma protein (pRb) in MCF-7 cells was inhibited, accompanied by a decrease in the levels of cyclin D1, cyclin D3 and cyclin-dependent kinase 4 (cdk4) and cdk6; in addition, there was an increase in the expression of cyclin-dependent kinase inhibitors p21WAF-1/Cip1 and p27Kip1. The results suggest that taiwanin E inhibits cell cycle progression of MCF-7 at the G0/G1 transition.
Collapse
Affiliation(s)
- Hui-Chun Wang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yen-Hsueh Tseng
- Department of Forestry, National Chung-Hsing University, Taichung, Taiwan
| | - Hui-Rong Wu
- Department of Forestry, National Chung-Hsing University, Taichung, Taiwan
| | - Fang-Hua Chu
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, Taiwan
| | - Yueh-Hsiung Kuo
- Graduate Institute of Chinese Pharmaceutical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Sheng-Yang Wang
- Department of Forestry, National Chung-Hsing University, Taichung, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
- Agricultural Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|
28
|
Haque I, Banerjee S, De A, Maity G, Sarkar S, Majumdar M, Jha SS, McGragor D, Banerjee SK. CCN5/WISP-2 promotes growth arrest of triple-negative breast cancer cells through accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation. Oncogene 2014; 34:3152-63. [PMID: 25132260 DOI: 10.1038/onc.2014.250] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 04/18/2014] [Accepted: 07/03/2014] [Indexed: 02/07/2023]
Abstract
The matricellular protein CCN5/WISP-2 represents a promising target in triple-negative breast cancer (TNBC) because treatment or induced activation of CCN5 in TNBC cells promotes cell growth arrest at the G0/G1 phase, reduces cell proliferation and delays tumor growth in the xenograft model. Our studies found that the p27(Kip1) tumor suppressor protein is upregulated and relocalized to the nucleus from cytoplasm by CCN5 in these cells and that these two events (upregulation and relocalization of p27(Kip1)) are critical for CCN5-induced growth inhibition of TNBC cells. In the absence of CCN5, p27(Kip1) resides mostly in the cytoplasm, which is associated with the aggressive nature of cancer cells. Mechanistically, CCN5 inhibits Skp2 expression, which seems to stabilize the p27(Kip1) protein in these cells. On the other hand, CCN5 also recruits FOXO3a to mediate the transcriptional regulation of p27(Kip1). The recruitment of FOXO3a is achieved by the induction of its expression and activity through shifting from cytoplasm to the nucleus. Our data indicate that CCN5 blocks PI3K/AKT signaling to dephosphorylate at S318, S253 and Thr32 in FOXO3a for nuclear relocalization and activation of FOXO3a. Moreover, inhibition of α6β1 receptors diminishes CCN5 action on p27(Kip1) in TNBC cells. Collectively, these data suggest that CCN5 effectively inhibits TNBC growth through the accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation, and thus, activation of CCN5 may have the therapeutic potential to kill TNBC.
Collapse
Affiliation(s)
- I Haque
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - S Banerjee
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - A De
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - G Maity
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - S Sarkar
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Department of Anatomy and Cell Biology and Department of Pathology, University of Kansas Medical Center, Kansas City, MO, USA
| | - M Majumdar
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - S S Jha
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - D McGragor
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - S K Banerjee
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA [3] Department of Anatomy and Cell Biology and Department of Pathology, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
29
|
Meng B, Wang Y, Li B. Suppression of PAX6 promotes cell proliferation and inhibits apoptosis in human retinoblastoma cells. Int J Mol Med 2014; 34:399-408. [PMID: 24939714 PMCID: PMC4094585 DOI: 10.3892/ijmm.2014.1812] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/03/2014] [Indexed: 12/28/2022] Open
Abstract
The aim of this study was to investigate the role of the transcription factor, PAX6, in the development of retinoblastoma. The expression of endogenous PAX6 was knocked down using PAX6-specific lentivirus in two human retinoblastoma cell lines, SO-Rb50 and Y79. Cell proliferation functional assays and apoptotic assays were performed on the cells in which PAX6 was knocked down. The results revealed that PAX6 knockdown efficiency was significant (P<0.01, n=3) in the SO-Rb50 and Y79 cells. The inhibition of PAX6 reduced tumor cell apoptosis (P<0.05, n=3), but induced cell cycle S phase arrest (SO-Rb50; P<0.05, n=3) and G2/M phase arrest (Y79; P<0.05, n=3). Western blot analysis indicated that the inhibition of PAX6 increased the levels of the anti-apoptotic proteins, Bcl-2, proliferating cell nuclear antigen (PCNA) and CDK1, but reduced the levels of the pro-apoptotic proteins, BAX and p21. In conclusion, our data demonstrate that the suppression of PAX6 increases proliferation and decreases apoptosis in human retinoblastoma cells by regulating several cell cycle and apoptosis biomarkers.
Collapse
Affiliation(s)
- Bo Meng
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, P.R. China
| | - Yisong Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China
| | - Bin Li
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, P.R. China
| |
Collapse
|
30
|
Nejatollahi F, Ranjbar R, Younesi V, Asgharpour M. Deregulation of HER2 downstream signaling in breast cancer cells by a cocktail of anti-HER2 scFvs. Oncol Res 2013; 20:333-40. [PMID: 23924853 DOI: 10.3727/096504013x13657689382734] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is overexpressed in 30% of patients with breast cancer. HER2 targeting is the mainstay of targeted therapy for the treatment of invasive breast cancers. Due to biological and therapeutic advantages, single chain fragment variable (scFv) antibodies have emerged as promising alternative therapeutics. In this study, we assessed the capability of three scFvs against HER2 extracellular domains (II, III, IV) in deregulation of some key signaling mediators that have important roles in growth, survival, angiogenesis, and cell migration of breast tumor cells. Downregulation of activated Akt (p-Akt), increase of p27 protein levels, and downregulation of HER1, HER2, HER3 and epidermal growth factor (EGF), CXCR3, CXCL10, and MMP2 were observed following treatment of breast cancer cells (SKBR3 cell line) with the scFvs and their combination. Our results suggest that the combination of the three scFvs could be considered as an effective cocktail on HER2 tumorgenic signaling pathways that leads to tumor growth suppression and death.
Collapse
|
31
|
Forte G, Fortuna CG, Salerno L, Modica MN, Siracusa MA, Cardile V, Romeo G, Bulbarelli A, Lonati E, Pittalà V. Antitumor properties of substituted (αE)-α-(1H-indol-3-ylmethylene)benzeneacetic acids or amides. Bioorg Med Chem 2013; 21:5233-45. [PMID: 23871443 DOI: 10.1016/j.bmc.2013.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/17/2013] [Accepted: 06/12/2013] [Indexed: 11/25/2022]
Abstract
A novel class of indole derivatives characterized by a (αE)-α-(1H-indol-3-ylmethylene)benzeneacetic acid or amide scaffold was synthesized. These derivatives, assayed for cell-growth inhibition activity against a panel of six different tumor cell lines, showed strong antiproliferative activity and selectivity mainly towards DU145 cell line. In particular, compounds 2d-m and 5 stand out for their cell growth inhibitory activity and, among them, compound 2d emerged for its selectivity towards DU145 with respect to other tested tumor cell lines. DU145 treated with 1μM of 2d for 72h showed p21(Cip1) induction and suppression of Akt signaling together with induction of Rb. From a computational point of view, two different approaches were used in order to study topology and electronic properties of the novel compounds and to shed light on their drug-likeness properties. Firstly, topological and electronic features of the compounds endowed with the most relevant biological activity were deepened; in parallel, some ADME properties like solubility and permeability were predicted.
Collapse
Affiliation(s)
- Giuseppe Forte
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, viale A. Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lee E, Son JE, Byun S, Lee SJ, Kim YA, Liu K, Kim J, Lim SS, Park JHY, Dong Z, Lee KW, Lee HJ. CDK2 and mTOR are direct molecular targets of isoangustone A in the suppression of human prostate cancer cell growth. Toxicol Appl Pharmacol 2013; 272:12-20. [PMID: 23707764 DOI: 10.1016/j.taap.2013.04.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 01/06/2023]
Abstract
Licorice extract which is used as a natural sweetener has been shown to possess inhibitory effects against prostate cancer, but the mechanisms responsible are poorly understood. Here, we report a compound, isoangustone A (IAA) in licorice that potently suppresses the growth of aggressive prostate cancer and sought to clarify its mechanism of action. We analyzed its inhibitory effects on the growth of PTEN-deleted human prostate cancer cells, in vitro and in vivo. Administration of IAA significantly attenuated the growth of prostate cancer cell cultures and xenograft tumors. These effects were found to be attributable to inhibition of the G1/S phase cell cycle transition and the accumulation of p27(kip1). The elevated p27(kip1) expression levels were concurrent with the decrease of its phosphorylation at threonine 187 through suppression of CDK2 kinase activity and the reduced phosphorylation of Akt at Serine 473 by diminishing the kinase activity of the mammalian target of rapamycin (mTOR). Further analysis using recombinant proteins and immunoprecipitated cell lysates determined that IAA exerts suppressive effects against CDK2 and mTOR kinase activity by direct binding with both proteins. These findings suggested that the licorice compound IAA is a potent molecular inhibitor of CDK2 and mTOR, with strong implications for the treatment of prostate cancer. Thus, licorice-derived extracts with high IAA content warrant further clinical investigation for nutritional sources for prostate cancer patients.
Collapse
Affiliation(s)
- Eunjung Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 151-921, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sirinian C, Symeonidis A, Giannakoulas N, Zolota V, Melachrinou M. Overexpression of phosphorylated p27 Kip1 at threonine 187 may predict outcome in aggressive B-cell lymphomas. Leuk Lymphoma 2011; 52:814-22. [PMID: 21338280 DOI: 10.3109/10428194.2011.555026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Phosphorylation of p27(Kip1) at threonine 187 (pThr187-p27(Kip1)) occurs frequently in the development of human tumors, directing protein polyubiquitination and subsequent proteasomal degradation. We investigated the immunoexpression of p27(Kip1) and pThr187-p27(Kip1) in 126 B-cell lymphomas and their relation to proliferative activity and clinical parameters. Increased levels of p27(Kip1) and pThr187-p27(Kip1) were significantly correlated with indolent and aggressive lymphomas, respectively (p < 0.001). pThr187-p27(Kip1) expression showed a strong positive correlation with proliferation index in aggressive (p = 0.01) and indolent (p < 0.001) subgroups. Survival analysis revealed that pThr187-p27(Kip1) was an unfavorable prognostic factor for disease-free (p = 0.019) and overall survival (p = 0.003) in aggressive lymphomas. Cox regression analysis demonstrated that the prognostic value of pThr187-p27(Kip1) was independent of the international prognostic index (IPI) score, tumor stage, patient age, and serum lactate dehydrogenase (LDH) level. Overall, our results suggest that high levels of pThr187-p27(Kip1) may predict a worse clinical outcome in patients with aggressive lymphomas.
Collapse
Affiliation(s)
- Chaido Sirinian
- Department of Pathology, Division of Hematology, University of Patras Medical School, Patras, Greece
| | | | | | | | | |
Collapse
|
34
|
Krstic AD, Impera L, Guc-Scekic M, Lakic N, Djokic D, Slavkovic B, Storlazzi CT. A complex rearrangement involving cryptic deletion of ETV6 and CDKN1B genes in a case of childhood acute lymphoblastic leukemia. CANCER GENETICS AND CYTOGENETICS 2009; 195:125-31. [PMID: 19963112 DOI: 10.1016/j.cancergencyto.2009.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 06/27/2009] [Accepted: 07/10/2009] [Indexed: 11/30/2022]
Abstract
We report on a case of childhood B-cell lineage acute lymphoblastic leukemia (ALL). Conventional cytogenetic analysis at diagnosis showed the karyotype: 47,XY,add(3)(q?),-12,+2mar[4]/46,XY[18]. Fluorescence in situ hybridization (FISH) revealed a complex rearrangement: 47,XY,der(3)(3pter->3q29::12q13->12q24.33::12p13.31->12p13.2::12q24.33->12qter),der(12)(12pter->12p13.31::12p12.3->12q12::3q29->3qter),+del(21)(q?). The derivative chromosome 3 arose likely from multiple events due to clonal evolution. After insertion of the segment of the short arm of the chromosome 12 to the distal part of the long arm of chromosome 12 [ins(12)(q24.33p13.31p13.2)], a translocation occurred between chromosome 3 and derivative chromosome 12. Additional FISH results disclosed two heterozygous deletions flanking the translocated region on both 12p13.2 approximately p12.3 and 12q12 approximately q13.13. The deleted segment on 12p contains several genes, among the tumor suppressor genes ETV6 and CDKN1B, which are frequently involved in 12p abnormalities in childhood ALL. Thus, the present study documents the loss of both ETV6 and CDKN1B genes accompanying the occurrence of a complex rearrangement involving chromosomes 3 and 12 in a case of childhood ALL.
Collapse
Affiliation(s)
- Aleksandra Drago Krstic
- Laboratory of Medical Genetics, Mother and Child Health Institute Dr. Vukan Cupic, Radoja Dakica 6-8, 11070 Belgrade, Serbia.
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Recent emerging evidence suggests that ING family proteins play roles in carcinogenesis both as oncogenes and tumor suppressor genes depending on the family members and on cell status. Previous results from non-physiologic overexpression experiments showed that all five family members induce apoptosis or cell cycle arrest, thus it had been thought until very recently that all of the family members function as tumor suppressor genes. Therefore restoration of ING family proteins in cancer cells has been proposed as a treatment for cancers. However, ING2 knockdown experiments showed unexpected results: ING2 knockdown led to senescence in normal human fibroblast cells and suppressed cancer cell growth. ING2 is also overexpressed in colorectal cancer, and promotes cancer cell invasion through an MMP13 dependent pathway. Additionally, it was reported that ING2 has two isoforms, ING2a and ING2b. Although expression of ING2a predominates compared with ING2b, both isoforms confer resistance against cell cycle arrest or apoptosis to cancer cells, thus knockdown of both isoforms is critical to remove this resistance. Taken together, these results suggest that ING2 can function as an oncogene in some specific types of cancer cells, indicating restoration of this gene in cancer cells could cause cancer progression. Because knockdown of ING2 suppresses cancer cell invasion and induces apoptosis or cell cycle arrest, ING2 may be an anticancer drug target. In this brief review, we discuss possible clinical applications of ING2 with the latest knowledge of molecular targeted therapies.
Collapse
Affiliation(s)
- M Unoki
- Laboratory for Biomarker, The Institute of Physical and Chemical Research, RIKEN, Tokyo 108-8639, Japan
| | | | | |
Collapse
|
36
|
Huang X, Yang C, Jin C, Luo Y, Wang F, McKeehan WL. Resident hepatocyte fibroblast growth factor receptor 4 limits hepatocarcinogenesis. Mol Carcinog 2009; 48:553-62. [PMID: 19009564 DOI: 10.1002/mc.20494] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor (FGF) family signaling mediates cell-to-cell communication in development and organ homeostasis in adults. Of the FGF receptor (FGFR) isotypes, FGFR4 is the sole resident isotype present in mature parenchymal hepatocytes. FGFR1 that is normally associated with activated nonparenchymal cells appears ectopically in hepatoma cells. Ectopic expression and chronic activity of FGFR1 in hepatocytes accelerates diethylnitrosamine (DEN)-initiated hepatocarcinogenesis by driving unrestrained cell proliferation and tumor angiogenesis. Hepatocyte FGFR4 mediates liver's role in systemic cholesterol/bile acid and lipid metabolism and affects proper hepatolobular restoration after damage without effect on cell proliferation. Here we ask whether FGFR4 plays a role in progression of hepatocellular carcinoma (HCC). We report that although spontaneous HCC was not detected in livers of FGFR4-deficient mice, the ablation of FGFR4 accelerated DEN-induced hepatocarcinogenesis. In contrast to FGFR1 that induced a strong mitogenic response and depressed rate of cell death in hepatoma cells, FGFR4 failed to induce a mitogenic response and increased the rate of cell death. FGFR1 but not FGFR4 induced cyclin D1 and repressed p27 expression. Analysis of activation of Erk, JNK, and PI3K-related AKT signaling pathways indicated that in contrast to FGFR1, FGFR4 failed to sustain Erk activation and did not activate AKT. These differences may underlie the opposing effects of FGFR1 and FGFR4. These results suggest that in contrast to ectopic FGFR1 that is a strong promoter of hepatoma, resident FGFR4 that mediates differentiated hepatocyte metabolic functions also serves to suppress hepatoma progression.
Collapse
Affiliation(s)
- Xinqiang Huang
- Center for Cancer & Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
37
|
Orzáez M, Gortat A, Mondragón L, Bachs O, Pérez-Payá E. ATP-noncompetitive inhibitors of CDK-cyclin complexes. ChemMedChem 2009; 4:19-24. [PMID: 19039815 DOI: 10.1002/cmdc.200800185] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Progression through the cell division cycle is controlled by a family of cyclin-dependent kinases (CDKs), the activity of which depends on their binding to regulatory partners (cyclins A-H). Deregulation of the activity of CDKs has been associated with the development of infectious, neurodegenerative, and proliferative diseases such as Alzheimer's, Parkinson's, or cancer. Most cancer cells contain mutations in the pathways that control the activity of CDKs. This observation led this kinase family to become a central target for the development of new drugs for cancer therapy. A range of structurally diverse molecules has been shown to inhibit the activity of CDKs through their activity as ATP antagonists. Nevertheless, the ATP binding sites on CDKs are highly conserved, limiting the kinase specificity of these inhibitors. Various genetic and crystallographic approaches have provided essential information about the mechanism of formation and activation of CDK-cyclin complexes, providing new ways to implement novel research strategies toward the discovery of new, more effective and selective drugs. Herein we review the progress made in the development of ATP-noncompetitive CDK-cyclin inhibitors.
Collapse
Affiliation(s)
- Mar Orzáez
- Department of Medicinal Chemistry, Centro de Investigación Príncipe Felipe, Avda. Autopista del Saler 16, 46012, Valencia, Spain.
| | | | | | | | | |
Collapse
|
38
|
Markaki EA, Stiakaki E, Zafiropoulos A, Arvanitis DA, Katzilakis N, Dimitriou H, Spandidos DA, Kalmanti M. Mutational analysis of the cell cycle inhibitor Kip1/p27 in childhood leukemia. Pediatr Blood Cancer 2006; 47:14-21. [PMID: 16526056 DOI: 10.1002/pbc.20730] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cyclin-dependent kinases (CDKs) and cyclins, their regulatory subunits, govern cell-cycle progression in eukaryotic cells. Kip1/p27 is the main cyclin-dependent kinase inhibitor, which arrests cell division inhibiting G1-S transition. Kip1/p27 seems to play a critical role in the pathogenesis of several human malignancies and its lower expression has been shown to correlate with a poor prognosis in adult solid tumors. METHODS Bone marrow blasts from 49 children with leukemia, 37 acute lymphoblastic leukemia (ALL), and 12 acute myeloid leukemia (AML) were studied. Exon 3 of Kip1/p27 was amplified using the polymerase chain reaction technique (PCR). Single strand conformational polymorphism and heterodouplex analysis were performed to detect DNA sequence with altered conformations and were subsequently sequenced to document mutations. RESULTS Mutations in Kip1/p27 gene were detected in 2 out of 3 T-ALL, 6 out of 12 AML patients, and only 1 out of 34 B lineage ALL cases. Although the patient groups are small, a highly significant relation of the mutation status with the type of leukemia (P = 0.0037) and the risk group according to treatment protocols (P = 0.00021) was estimated. A statistically significant difference in the white blood count was observed (P = 0.019) between the mutated and non-mutated patient groups although no statistically significant association of the mutation status with the hemoglobin and platelets values, karyotype, age, sex, disease progression, and outcome was determined. CONCLUSIONS Based upon these results, the Kip1/p27 mutations should be considered for further prospective testing as an additional parameter for risk stratification and treatment of childhood leukemia.
Collapse
Affiliation(s)
- E-A Markaki
- Department of Pediatric Hematology-Oncology, University of Crete, Medical School, Heraklion Crete, Greece
| | | | | | | | | | | | | | | |
Collapse
|
39
|
di Pietro A, Vries EGED, Gietema JA, Spierings DCJ, de Jong S. Testicular germ cell tumours: the paradigm of chemo-sensitive solid tumours. Int J Biochem Cell Biol 2005; 37:2437-56. [PMID: 16099193 DOI: 10.1016/j.biocel.2005.06.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Revised: 06/02/2005] [Accepted: 01/27/2005] [Indexed: 11/16/2022]
Abstract
Testicular germ cell tumours (TGCTs) are the most frequent solid malignant tumour in men 20-40 years of age and the most frequent cause of death from solid tumours in this age group. Up to 50% of the patients suffer from metastatic disease at diagnosis. The majority of metastatic testicular cancer patients, in contrast to most other metastatic solid tumours, can be cured with highly effective cisplatin-based chemotherapy. From a genetic point of view, almost all TGCTs in contrast to solid tumours are characterised by the presence of wild type p53. High p53 expression levels are associated with elevated Mdm2 levels and a loss of p21(Waf1/Cip1) expression suggesting a changed functionality of p53. Expression levels of other proteins involved in the regulation of cell cycle progression indicate a deregulated G1-S phase checkpoint in TGCTs. After cisplatin-induced DNA damage, the increasing levels of p53 lead to the trans-activation of a number of genes but not of p21(Waf1/Cip1), preferentially directing TGCT cells into apoptosis or programmed cell death, both via the mitochondrial and the death receptor apoptosis pathways. The sensitivity of TGCTs to chemotherapeutic drugs may lay in the susceptibility of germ cells to apoptosis. Taken together, this provides TGCT as a tumour type model to investigate and understand the molecular determinants of chemotherapy sensitivity of solid tumours. This review aims to summarise the current knowledge on the biological basis of cisplatin-induced apoptosis and response to chemotherapy in TGCTs.
Collapse
Affiliation(s)
- Alessandra di Pietro
- Department of Medical Oncology, Internal Medicine, University of Groningen and University Medical Center Groningen, 9713 GZ Hanzeplein 1, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
40
|
Abstract
Critical cellular processes are regulated, in part, by maintaining the appropriate intracellular levels of proteins. Whereas de novo protein synthesis is a comparatively slow process, proteins are rapidly degraded at a rate compatible with the control of cell cycle transitions and cell death induction. A major pathway for protein degradation is initiated by the addition of multiple 76-amino acid ubiquitin monomers via a three-step process of ubiquitin activation and substrate recognition. Polyubiquitination targets proteins for recognition and processing by the 26S proteasome, a cylindrical organelle that recognizes ubiquitinated proteins, degrades the proteins, and recycles ubiquitin. The critical roles played by ubiquitin-mediated protein turnover in cell cycle regulation makes this process a target for oncogenic mutations. Oncogenes of several common malignancies, for example colon and renal cell cancer, code for ubiquitin ligase components. Cervical oncogenesis by human papillomavirus is also mediated by alteration of ubiquitin ligase pathways. Protein degradation pathways are also targets for cancer therapy, as shown by the successful introduction of bortezomib, an inhibitor of the 26S proteasome. Further work in this area holds great promise toward our understanding and treatment of a wide range of cancers.
Collapse
Affiliation(s)
- Aparna Mani
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Rd NW, Washington, DC 20007-2197, USA
| | | |
Collapse
|
41
|
Stefanaki C, Antoniou C, Stefanaki K, Stratigos A, Constantinidou VV, Argyrakos T, Karentzou O, Katsambas A. Expression of the cyclin-dependent kinase inhibitor p27kip-1 in benign naevi and correlation with Ki-67 proliferative index. Br J Dermatol 2005; 152:373-4. [PMID: 15727660 DOI: 10.1111/j.1365-2133.2005.06312.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Martín-Caballero J, Flores JM, García-Palencia P, Collado M, Serrano M. Different cooperating effect of p21 or p27 deficiency in combination with INK4a/ARF deletion in mice. Oncogene 2004; 23:8231-7. [PMID: 15378017 DOI: 10.1038/sj.onc.1207863] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The control exerted by the INK4a/ARF locus on cellular proliferation is crucial to restrict tumor development. In agreement with this, mice with defects in this locus are highly tumor prone. However, the potential contribution of other pathways in modulating tumorigenesis in the absence of INK4a/ARF is largely unexplored. In the present study, we investigated the consequences of the combined loss of either of two cyclin-dependent kinase inhibitors, p21 and p27, in cooperation with deletion of the INK4a/ARF locus. Our results show a clear differential effect in tumorigenesis depending on the CKI that is absent. The absence of p21 produced no overt alteration of the lifespan of the INK4a/ARF-null mice, although it modified their tumor spectrum, causing a significant increase in the incidence of fibrosarcomas and the appearance of a small number of rhabdomyosarcomas. In contrast, deficiency of p27 resulted in a significant increase in lethality due to accelerated tumor development, especially in the case of T-cell lymphomas. Finally, combined deficiency of INK4a/ARF and p27 resulted in a significant increase in the number of metastatic tumors. These results demonstrate genetically the oncogenic cooperation between defects on INK4a/ARF and p27, which are common alterations in human cancer.
Collapse
Affiliation(s)
- Juan Martín-Caballero
- Molecular Oncology Program, Spanish National Cancer Center (CNIO), Melchor Fernandez Almagro 3, Madrid E-28029, Spain
| | | | | | | | | |
Collapse
|
43
|
Chen CF, Goyette P, Lohnes D. RARgamma acts as a tumor suppressor in mouse keratinocytes. Oncogene 2004; 23:5350-9. [PMID: 15094780 DOI: 10.1038/sj.onc.1207682] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Revised: 02/23/2004] [Accepted: 02/23/2004] [Indexed: 11/09/2022]
Abstract
All-trans retinoic acid (RA), the principle biologically active form of vitamin A, is essential for many developmental process as well as homeostasis in the adult. Many lines of evidence also suggest that RA, acting through the RA receptors (RARs), can also suppress growth of tumors of diverse origin. To assess directly the role of the RARs in a model of epidermal tumorigenesis, we investigated the incidence of tumor formation using keratinocytes lacking specific RAR types. Our data suggest that loss of RARgamma, but not RARalpha, predisposed keratinocytes to v-Ha-Ras-induced squamous cell carcinoma. We also found that ablation of RARgamma, but not RARalpha, abolished RA-induced cell cycle arrest and apoptosis in these keratinocytes. Reconstitution of receptor expression into RAR-null cells restored sensitivity to RA, and reversed the tumorigenic potential of receptor-deficient keratinocytes. These data strongly support a tumor suppressor effect for the RARs, in particular endogenous RARgamma, in murine keratinocytes.
Collapse
Affiliation(s)
- Chang Feng Chen
- Division of Experimental Medicine, McGill University, Quebec, Canada
| | | | | |
Collapse
|
44
|
Philipp-Staheli J, Kim KH, Liggitt D, Gurley KE, Longton G, Kemp CJ. Distinct roles for p53, p27Kip1, and p21Cip1 during tumor development. Oncogene 2003; 23:905-13. [PMID: 14647411 DOI: 10.1038/sj.onc.1207220] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mutations in p53 and reduced expression of the Cdk inhibitor p27Kip1 are frequently observed in a wide variety of human cancers, but it is not known whether alterations in these tumor suppressors interact to influence tumor progression. To address this, we measured tumor latency and spectrum in p53 and p27 single and compound mutant mice. p53-/- (null) mice developed T-cell lymphomas and soft-tissue sarcomas, while p27-/- mice developed adenomas of the pituitary and lung, but with much longer latency. The latency for tumor development in p53-/- p27-/- and p53-/- p27+/- compound mutant mice was significantly reduced, by 15-30%, compared to single mutant p53-/- mice. The tumor spectrum in the compound mutants was similar to that of p53-/- mice, and additional tumors of diverse histotypes. In tumors from p53-/- mice, p27 protein levels were reduced to a greater extent than were mRNA levels, indicating that p27 is downregulated in tumors at the transcriptional as well as post-transcriptional levels. In contrast, mice deficient in another Cdk inhibitor p21Cip1, which is also a transcriptional target and effector of p53, showed only a marginal increase in tumor predisposition in response to ENU treatment. Thus, downregulation of p27 is a common feature in p53-/- tumors. Germline deletion of one or both alleles of p27 accelerates tumor development and associated mortality in p53-/- mice, indicating potent synergy between loss of p27 and p53. Although p21 is functionally similar to both p53 and p27, it plays a lesser role in tumor suppression. These results further highlight the highly cooperative nature of p27 and its central role in tumor suppression.
Collapse
|
45
|
Osipov V, Keating JT, Faul PN, Loda M, Datta MW. Expression of p27 and VHL in renal tumors. Appl Immunohistochem Mol Morphol 2002; 10:344-50. [PMID: 12607603 DOI: 10.1097/00129039-200212000-00010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Renal tumors, in particular clear cell renal cell carcinomas, have an unclear prognosis and metastatic potential. Cell cycle regulators play a key role in cellular proliferation and have been implicated in neoplasia. The cell cycle inhibitor p27 has been associated with prognosis in various tumor types. Recently a reported association between p27 and Von Hippel-Lindau (VHL) gene function has also been noted. We have examined p27 and VHL expression by immunohistochemistry in a panel of kidney tumors and have noted specific and unique patterns of p27 expression in various tumor types. In addition, we have analyzed p27 expression in clear cell type renal cell carcinomas and have noted a significant association between decreasing p27 expression and increasing tumor size, suggesting a relation between renal cell proliferation and loss of p27 function. These findings suggest a role for p27 in the development of various types of renal tumors.
Collapse
Affiliation(s)
- Vladimir Osipov
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | |
Collapse
|
46
|
Lasak JM, Welling DB, Akhmametyeva EM, Salloum M, Chang LS. Retinoblastoma-cyclin-dependent kinase pathway deregulation in vestibular schwannomas. Laryngoscope 2002; 112:1555-61. [PMID: 12352662 DOI: 10.1097/00005537-200209000-00004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The purpose of the study was to identify genes of the retinoblastoma protein (pRb)-cyclin-dependent kinase (CDK) pathway that are deregulated in vestibular schwannomas when compared with normal vestibular nerve tissues. STUDY DESIGN Expression profiles in eight vestibular schwannomas (four sporadic tumors, one neurofibromatosis type 2 tumor, and three cystic tumors) and a paired normal vestibular nerve from one of the eight patients were chosen. Genes examined included the retinoblastoma susceptibility gene (Rb-1); cyclins D1, D2, A, and E; the CDK inhibitors p18, p19, and p27; CDK2 and CDK6; transcription factors E2F-4, E2F-5, and DP-1; and the neurofibromatosis type 2 gene. METHODS Total RNA samples were extracted from normal vestibular nerve and vestibular schwannoma tissues and used to generate radiolabeled complementary DNA (cDNA) samples. Labeled cDNA probes were then hybridized to cDNA microarray filters. The hybridization signal was captured and quantified. Differential gene expression profiles between the normal vestibular nerve and vestibular schwannoma were compared. Real-time polymerase chain reaction and immunohistochemistry were used to further confirm the cDNA microarray data. RESULTS Among genes in the pRb-CDK pathway, CDK2 was substantially underexpressed in seven of the eight vestibular schwannoma tumors examined. Quantitative RNA expression analysis using real-time polymerase chain reaction also showed consistent downregulation of CDK2 in the tumors. Anti-CDK2 antibody stained predominantly in the vestibular nerve and ganglion cells but only weakly in the vestibular schwannoma tissues. CONCLUSIONS The pRb-CDK pathway was altered in all vestibular schwannoma tumors examined, with CDK2 significantly downregulated in seven of the eight tumors. Further investigation into the regulatory mechanisms governing CDK2 expression may lead to a better understanding of vestibular schwannoma tumorigenesis.
Collapse
Affiliation(s)
- John M Lasak
- Department of Otolaryngology, The Ohio State University and Children's Hospital, Columbus, USA
| | | | | | | | | |
Collapse
|
47
|
Wohlschlegel JA, Kutok JL, Weng AP, Dutta A. Expression of geminin as a marker of cell proliferation in normal tissues and malignancies. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:267-73. [PMID: 12107111 PMCID: PMC1850683 DOI: 10.1016/s0002-9440(10)64178-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Geminin interacts with a DNA replication initiation factor, Cdt1p, to suppress initiation of DNA replication in a Xenopus egg extract based cell-free system, leading to the expectation that the protein acts as an inhibitor of cell proliferation. Immunohistochemistry and immunoblotting for geminin, however, reveals that the protein is expressed specifically in proliferating lymphocytes and epithelial cells. This pattern is in contrast to the expression of a bona fide cell cycle inhibitor like p21/WAF1 that is specifically expressed in quiescent cells. Geminin is widely expressed in several malignancies and the number of geminin-expressing cells is directly proportional to the cell proliferation index as measured by Ki-67 expression. Therefore, instead of being a suppressor of cell proliferation, geminin expression is positively correlated with cell proliferation. Consistent with this observation, transient overexpression of wild-type geminin in cancer cells in culture did not produce a cell cycle block. A point mutation in the destruction box of geminin, however, results in a protein that is stabilized in G(1) and capable of arresting cells at the G(1)-S transition.
Collapse
Affiliation(s)
- James A Wohlschlegel
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
48
|
Bai M, Vlachonikolis J, Agnantis NJ, Tsanou E, Dimou S, Nicolaides C, Stefanaki S, Pavlidis N, Kanavaros P, Kanavarous P. Low expression of p27 protein combined with altered p53 and Rb/p16 expression status is associated with increased expression of cyclin A and cyclin B1 in diffuse large B-cell lymphomas. Mod Pathol 2001; 14:1105-13. [PMID: 11706071 DOI: 10.1038/modpathol.3880444] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The expression of the cyclin-dependent kinase inhibitor (CDKI) p27 protein was investigated in relation to (1) the expression of the cell cycle regulators p53, Rb and p16 and (2) the proliferation profile as determined by the expression of Ki67, cyclin A, and cyclin B1 in 80 cases of de novo diffuse large B-cell lymphomas (DLBCL). P27 expression was low/null in large tumor cells in 58/80 cases and intermediate/high in 22/80 cases. Increased expression of p53 protein was observed in 39/80 cases. Decreased expression of Rb and p16 proteins was mutually exclusive and was observed in 5/80 and 14/80 cases, respectively. The analysis of the p27 expression status (low/null versus intermediate/high) with respect to the p53 and/or Rb/p16 expression status showed that low/null p27 expression was significantly correlated with increased p53 expression (P =.018) and showed a strong trend for correlation with concurrent increased p53 expression and decreased Rb or p16 expression (P =.050). These findings suggest a tendency for concurrent alterations of the cell cycle regulators p27, p53, and Rb or p16 in DLBCL, which might result in impaired tumor growth control. Indeed, the analysis of the combined p27/p53/Rb/p16 expression status with respect to the proliferation profile showed that (1) three alterations in the combined p27/p53/Rb/p16 status (i.e., low/null P27 expression, increased expression of p53, and decreased expression of Rb or p16) were significantly correlated with increased expression of cyclin B1 (P =.005) and (2) two or three alterations were significantly correlated with increased expression of cyclin A (P =.014). These findings suggest combined impairment of a complex cell-cycle control network involving the CDK inhibitor p27, the P53 pathway, and the Rb1 pathway, which exerts a cooperative effect resulting in enhanced tumor cell proliferation.
Collapse
Affiliation(s)
- M Bai
- Department of Pathology, Medical Faculty, University of Ioannina, 45110, Ioannina, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Choi J, Means RE, Damania B, Jung JU. Molecular piracy of Kaposi's sarcoma associated herpesvirus. Cytokine Growth Factor Rev 2001; 12:245-57. [PMID: 11325605 DOI: 10.1016/s1359-6101(00)00029-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Kaposi's Sarcoma associated Herpesvirus (KSHV) is the most recently discovered human tumor virus and is associated with the pathogenesis of Kaposi's sarcoma, primary effusion lymphoma, and Multicentric Casttleman's disease. KSHV contains numerous open reading frames with striking homology to cellular genes. These viral gene products play a variety of roles in KSHV-associated pathogenesis by disrupting cellular signal transduction pathways, which include interferon-mediated anti-viral responses, cytokine-regulated cell growth, apoptosis, and cell cycle control. In this review, we will attempt to cover our understanding of how viral proteins deregulate cellular signaling pathways, which ultimately contribute to the conversion of normal cells to cancerous cells.
Collapse
Affiliation(s)
- J Choi
- Department of Microbiology and Molecular Genetics, Tumor Virology Division, New England Regional Primate Research Center, Harvard Medical School, 1 Pine Hill Drive, Southborough, MA 01772, USA
| | | | | | | |
Collapse
|