1
|
Wang M, Zhu L, Yang X, Li J, Liu Y, Tang Y. Targeting immune cell types of tumor microenvironment to overcome resistance to PD-1/PD-L1 blockade in lung cancer. Front Pharmacol 2023; 14:1132158. [PMID: 36874015 PMCID: PMC9974851 DOI: 10.3389/fphar.2023.1132158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Lung cancer is the common malignant tumor with the highest mortality rate. Lung cancer patients have achieved benefits from immunotherapy, including immune checkpoint inhibitors (ICIs) therapy. Unfortunately, cancer patients acquire adaptive immune resistance, leading to poor prognosis. Tumor microenvironment (TME) has been demonstrated to play a critical role in participating in acquired adaptive immune resistance. TME is associated with molecular heterogeneity of immunotherapy efficacy in lung cancer. In this article, we discuss how immune cell types of TME are correlated with immunotherapy in lung cancer. Moreover, we describe the efficacy of immunotherapy in driven gene mutations in lung cancer, including KRAS, TP53, EGFR, ALK, ROS1, KEAP1, ZFHX3, PTCH1, PAK7, UBE3A, TNF-α, NOTCH, LRP1B, FBXW7, and STK11. We also emphasize that modulation of immune cell types of TME could be a promising strategy for improving adaptive immune resistance in lung cancer.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lijie Zhu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoxu Yang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiahui Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Ying Tang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
2
|
Effects of Ozone on Hippocampus BDNF and Fos Expressions in Rats with Chronic Compression of Dorsal Root Ganglia. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5572915. [PMID: 34869766 PMCID: PMC8642004 DOI: 10.1155/2021/5572915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022]
Abstract
The effects of ozone on hippocampal expression levels of brain-derived neurotrophic factor (BDNF) and c-fos protein (Fos) were evaluated in rats with chronic compression of dorsal root ganglia (CCD). Forty-eight adult female Sprague-Dawley rats were randomly divided into the following 4 groups (n = 12): sham operation (sham group), CCD group, CCD with 20 μg/ml of ozone (CCD + AO3 group), and CCD with 40 μg/ml of ozone (CCD + BO3 group). Except the sham group, unilateral L5 dorsal root ganglion (DRG) compression was performed on all other groups. On days 1, 2, and 4 after the operation, the CCD + AO3 and CCD + BO3 groups were injected with 100 μl of ozone with concentrations of 20 and 40 μg/ml, respectively. Thermal withdrawal latencies (TWLs) and mechanical withdrawal thresholds (MWTs) were measured at various time points before and after the operation. BDNF and Fos expressions were examined in the extracted hippocampi using immunohistochemistry. The TWLs and MWTs of CCD model rats that received ozone were lower with decreased BDNF and increased Fos expression levels, on day 21 after the operation, compared to those of the sham group (P < 0.05). The TWLs and MWTs of the CCD + AO3 and CCD + BO3 groups were higher with increased BDNF and decreased Fos expression levels, on day 21 after the operation, compared to those of the CCD group (P < 0.05). The TWLs were longer and the MWTs were higher in the CCD + BO3 group at each time point with increased BDNF and decreased Fos expression levels, on day 21 after the operation, compared to those of the CCD + AO3 group (P < 0.05). Our results revealed that ozone can relieve the neuropathic pain caused by the pathological neuralgia resulting from DRG compression in rats. The mechanism of action for ozone is likely associated with changes in BDNF and Fos expression levels in the hippocampus.
Collapse
|
3
|
Chen H, Gao J, Xu Q, Wan D, Zhai W, Deng L, Qie R. MiR-145-5p modulates lipid metabolism and M2 macrophage polarization by targeting PAK7 and regulating β-catenin signaling in hyperlipidemia. Can J Physiol Pharmacol 2021; 99:857-863. [PMID: 34143694 DOI: 10.1139/cjpp-2020-0539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The present study aims to explore the role of microRNA 145-5p (miR-145-5p) in hyperlipidemia. Using bioinformatics tools and a wide range of function and mechanism assays, we attempted to understand the specific function and potential mechanism of miR-145-5p in hyperlipidemia. A cholesterol-enriched diet induced an increase of serum cholesterol and triacylglycerol but a decrease of serum high-density lipoprotein. MiR-145-5p level was decreased in hyperlipidemia rat models. MiR-145-5p regulated lipid metabolism by antagonizing the alteration of high-density lipoprotein, cholesterol, and triacylglycerol in serum mediated by a cholesterol-enriched diet. In mechanism, miR-145-5p directly bound with p21 protein (RAC1)-activated kinase 7 (PAK7) and negatively regulated mRNA and protein levels of PAK7 in THP-1 cells. Furthermore, miR-145-5p level was negatively associated with PAK7 level in rat cardiac tissues. Finally, overexpression of PAK7 reversed the effects of miR-145-5p on β-catenin activation and M2 macrophages polarization in THP-1 cells. In conclusion, MiR-145-5p modulated lipid metabolism and M2 macrophage polarization by targeting PAK7 and regulating β-catenin signaling in hyperlipidemia, which may provide a potential biomarker for the treatment of hyperlipidemia-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Huijun Chen
- Department of Cardiology, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Jing Gao
- Department of Cardiology, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Qian Xu
- Department of Cardiology, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Dongmei Wan
- Department of Cardiology, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Wenji Zhai
- Department of Cardiology, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Limei Deng
- Department of Cardiology, Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Rui Qie
- Department of Emergency, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| |
Collapse
|
4
|
Gu YF, Kong LT. Inhibiting p21-activated kinase (PAK7) enhances radiosensitivity in hepatocellular carcinoma. Hum Exp Toxicol 2021; 40:2202-2214. [PMID: 34165002 DOI: 10.1177/09603271211027948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In light of the upregulation of p21-activated kinase (PAK7) in a variety of cancers, including hepatocellular carcinoma (HCC), we aimed to investigate the effect of PAK7 on the sensitivity of HCC cells to radiotherapy. METHODS PAK7 expression was determined in normal adult liver epithelial THLE-2 and human HCC cell lines. The effect of ionizing radiation (IR) on the HCC cell viability was evaluated by Sulforhodamine B (SRB) assay. HCC cell lines Mahlavu and Huh7 were chosen to assess the effect of PAK7 shRNAs on the viability, clone formation, apoptosis, cycle distribution and γ-H2AX expression after exposure to IR. RESULTS As compared to THLE-2 cells, PAK7 was upregulated in poorly differentiated Mahlavu and SK-Hep-1 cells, but moderately or lowly expressed in well-differentiated Huh7 and HepG2 cells. HCC cells with moderate or low expression of PAK7 presented a decreased viability at 2 Gy IR, which had no significant effect on PAK7high HCC cells. Mahlavu and Huh7 cells transfected with PAK7 shRNAs showed increased inhibitory effect of IR on viability. In addition, PAK7 shRNAs reduced clone formation, facilitated the cell apoptosis, arrested cells at G2/M phase, and increased γ-H2AX expression. Moreover, changes above were more evident in the HCC cells co-treated with IR and PAK7 shRNAs. CONCLUSION PAK7 downregulation could inhibit the viability, promote the apoptosis, arrest cells in G2/M phase, and induce the DNA damage in HCC cells, thereby enhancing the radiosensitivity in HCC.
Collapse
Affiliation(s)
- Y-F Gu
- Department of Radiotherapy Second Ward, YanTaiShan hospital, YanTai, Shandong, People's Republic of China
| | - L-T Kong
- Department of Emergency, YanTaiShan Hospital, YanTai, Shandong, People's Republic of China
| |
Collapse
|
5
|
A New Computational Approach to Evaluating Systemic Gene–Gene Interactions in a Pathway Affected by Drug LY294002. Processes (Basel) 2020. [DOI: 10.3390/pr8101230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this study, we investigate how drugs systemically affect genes via pathways by integrating information from interactions between chemical compounds and molecular expression datasets, and from pathway information such as gene sets using mathematical models. First, we adopt drug-induced gene expression datasets; then, employ gene set enrichment analysis tools for selecting candidate enrichment pathways; and lastly, implement the inverse algorithm package for identifying gene–gene regulatory networks in a pathway. We tested LY294002-induced datasets of the MCF7 breast cancer cell lines, and found a CELL CYCLE pathway with 101 genes, ERBB signaling pathway consisting of 82 genes, and MTOR pathway consisting of 45 genes. We consider two interactions: quantity strength depending on number of interactions, and quality strength depending on weight of interaction as positive (+) and negative (−) interactions. Our methods revealed ANAPC1-CDK6 (−0.412) and ORC2L- CHEK1(0.951) for the CELL CYCLE pathway; INS-RPS6 (−3.125) and PRKAA2-PRKAA2 (+1.319) for the MTOR pathway; and CBLB-RPS6KB1 (−0.141), RPS6KB1-CBLC (+0.238) for the ERBB signaling pathway to be top quality interactions. Top quantity interactions discovered include 12; the CDC (−,+) gene family for the CELL CYCLE pathway, 20; PIK3 (−), 23; PIK3CG (+) for the MTOR pathway, 11; PAK (−), 10; PIK3 (+) for the ERBB signaling pathway.
Collapse
|
6
|
Nunnari G, Sanfilippo C, Castrogiovanni P, Imbesi R, Li Volti G, Barbagallo I, Musumeci G, Di Rosa M. Network perturbation analysis in human bronchial epithelial cells following SARS-CoV2 infection. Exp Cell Res 2020; 395:112204. [PMID: 32735892 PMCID: PMC7386311 DOI: 10.1016/j.yexcr.2020.112204] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 11/24/2022]
Abstract
Background SARS-CoV2, the agent responsible for the current pandemic, is also causing respiratory distress syndrome (RDS), hyperinflammation and high mortality. It is critical to dissect the pathogenetic mechanisms in order to reach a targeted therapeutic approach. Methods In the present investigation, we evaluated the effects of SARS-CoV2 on human bronchial epithelial cells (HBEC). We used RNA-seq datasets available online for identifying SARS-CoV2 potential genes target on human bronchial epithelial cells. RNA expression levels and potential cellular gene pathways have been analyzed. In order to identify possible common strategies among the main pandemic viruses, such as SARS-CoV2, SARS-CoV1, MERS-CoV, and H1N1, we carried out a hypergeometric test of the main genes transcribed in the cells of the respiratory tract exposed to these viruses. Results The analysis showed that two mechanisms are highly regulated in HBEC: the innate immunity recruitment and the disassembly of cilia and cytoskeletal structure. The granulocyte colony-stimulating factor (CSF3) and dynein heavy chain 7, axonemal (DNAH7) represented respectively the most upregulated and downregulated genes belonging to the two mechanisms highlighted above. Furthermore, the carcinoembryonic antigen-related cell adhesion molecule 7 (CEACAM7) that codifies for a surface protein is highly specific of SARS-CoV2 and not for SARS-CoV1, MERS-CoV, and H1N1, suggesting a potential role in viral entry. In order to identify potential new drugs, using a machine learning approach, we highlighted Flunisolide, Thalidomide, Lenalidomide, Desoximetasone, xylazine, and salmeterol as potential drugs against SARS-CoV2 infection. Conclusions Overall, lung involvement and RDS could be generated by the activation and down regulation of diverse gene pathway involving respiratory cilia and muscle contraction, apoptotic phenomena, matrix destructuration, collagen deposition, neutrophil and macrophages recruitment. SARS-CoV2 causing respiratory distress syndrome, hyperinflammation and high mortality. In NHBEC, SARS-CoV2 highly regulated the innate immunity recruitment and the disassembly of cilia and cytoskeletal structure. The granulocyte colony-stimulating factor (CSF3) is the most upregulated gene by SARS-CoV2. The dynein heavy chain 7, axonemal (DNAH7) represented the most downregulated genes by SARS-CoV2. Flunisolide, Thalidomide, Lenalidomide, Desoximetasone, xylazine, and salmeterol as potential drugs against SARS-CoV-2.
Collapse
Affiliation(s)
- Giuseppe Nunnari
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| | - Cristina Sanfilippo
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124, Messina, Italy.
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy.
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy.
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy.
| | - Ignazio Barbagallo
- Department of Drug Sciences, University of Catania, Viale Andrea Doria, 6, 95125, Catania, Italy.
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy.
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy.
| |
Collapse
|
7
|
Bao Z, Ji W, Yang Y, Chen Z, Li Z, Wang K, Lu T, Yu Y, Xia W, Lu S. PAK5 promotes the cell stemness ability by phosphorylating SOX2 in lung squamous cell carcinomas. Exp Cell Res 2020; 395:112187. [PMID: 32721391 DOI: 10.1016/j.yexcr.2020.112187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 01/01/2023]
Abstract
Growing evidences suggest that the overexpression of p21-activated kinase 5 (PAK5) plays an important role in various tumor progression. However, the role of PAK5 and its downstream target gene(s) in lung squamous cell carcinomas (LUSC) are waiting to be elucidated. TCGA data were utilized to evaluate the expression levels of PAK5 in LUSC. We then explored the role of PAK5 in maintaining the stem-like phenotype of lung squamous cancer cells through RT-PCR, flow cytometry, oncosphere-forming assay. In addition, co-immunoprecipitation, western blotting and immunofluorescence assays were used to determine SOX2 as a novel effector of PAK5. Xenograft models in nude mice were established to explore the roles of PAK5 in lung cancer growth. In this study, we have shown that PAK5 is overexpressed in LUSC tissues. The absence of PAK5 abolishes self-renewal ability of LUSC cells by decreasing the expression and phosphorylation of SOX2 in vitro and in vivo. In xenograft models, knockdown or pharmacological inhibition of PAK5 suppressed the tumor growth and metastasis of lung squamous cancer cells in vivo. Taken together, our findings suggest that the PAK5-mediated SOX2 phosphorylation promoted the cancer stem cell-like phenotype of LUSC cells. PAK5 inhibition may be a promising target in the treatment of SOX2 positive lung squamous cell cancer.
Collapse
Affiliation(s)
- Zinan Bao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, 200030, Shanghai, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China
| | - Ying Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China
| | - Zhuo Chen
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, 200030, Shanghai, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China
| | - Kaixuan Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China
| | - Tingting Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China
| | - Weiliang Xia
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Huashan Road 1954, 200030, Shanghai, China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, 20030, Shanghai, China.
| |
Collapse
|
8
|
Zhang J, Zhang Y, Cheng S, Mu Y, Liu Y, Yi X, Jiang D, Ding Y, Zhuang R. LAIR-1 overexpression inhibits epithelial-mesenchymal transition in osteosarcoma via GLUT1-related energy metabolism. World J Surg Oncol 2020; 18:136. [PMID: 32563267 PMCID: PMC7345510 DOI: 10.1186/s12957-020-01896-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a collagen receptor belonging to the immunoglobulin superfamily. Although previous studies have evaluated the biological role of LAIR in solid tumors, the precise mechanisms underlying the functions of LAIR-1 as a regulator of tumor biological functions remain unclear. Methods LAIR-1 expression was evaluated by immunohistochemical analysis using an osteosarcoma (OS) tissue microarray. Wound healing and transwell migration assays were performed to evaluate tumor cell migration. Quantitative real-time polymerase chain reaction (qPCR) and western blotting were conducted to detect the expression of epithelial–mesenchymal transition (EMT)-related molecules. RNA-sequencing (RNA-seq) was conducted to evaluate the mRNA expression profiles after overexpressing LAIR-1 in OS cells. Glucose transporter (Glut)1 expression in OS cells was evaluated by western blotting. Results LAIR-1 expression was significantly different between the T1 and T2 stages of OS tumors, and it inhibited OS cell migration. LAIR-1 expression was inversely correlated with the expression of Twist1, an EMT-associated transcription factor, via the Forkhead box O1 signal transduction pathway. Furthermore, RNA-seq and qPCR demonstrated that the expression of EMT energy metabolism-related molecules was significantly reduced after LAIR-1 overexpression. Conclusions LAIR-1 overexpression decreased the expression of Glut1 and inhibited the expression of EMT-related molecules in OS cells. These findings provide new insights into the molecular mechanism underlying OS progression.
Collapse
Affiliation(s)
- Jinxue Zhang
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, #1 Xinsi Road, Xi'an, 710032, China.,Department of Immunology, Fourth Military Medical University, #129 West Changle Road, Xi'an, 710032, China
| | - Yuan Zhang
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China
| | - Shiyang Cheng
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, #1 Xinsi Road, Xi'an, 710032, China
| | - Yang Mu
- Department of Immunology, Fourth Military Medical University, #129 West Changle Road, Xi'an, 710032, China
| | - Yongming Liu
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, #1 Xinsi Road, Xi'an, 710032, China
| | - Xin Yi
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, #1 Xinsi Road, Xi'an, 710032, China
| | - Dongxu Jiang
- Department of Immunology, Fourth Military Medical University, #129 West Changle Road, Xi'an, 710032, China
| | - Yong Ding
- Orthopedic Department of Tangdu Hospital, Fourth Military Medical University, #1 Xinsi Road, Xi'an, 710032, China.
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, #129 West Changle Road, Xi'an, 710032, China.
| |
Collapse
|
9
|
Dang Y, Guo Y, Ma X, Chao X, Wang F, Cai L, Yan Z, Xie L, Guo X. Systemic analysis of the expression and prognostic significance of PAKs in breast cancer. Genomics 2020; 112:2433-2444. [PMID: 31987914 DOI: 10.1016/j.ygeno.2020.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/24/2019] [Accepted: 01/23/2020] [Indexed: 12/23/2022]
Abstract
PAKs (p21-activated kinases) are reported to play crucial roles in a variety of cellular processes and participate in the progression of human cancers. However, the expression and prognostic values of PAKs remain poorly explored in breast cancers. In our study, we examined the mRNA and protein expression levels of PAKs and the prognostic value. We also analyzed the interaction network, genetic alteration, and functional enrichment of PAKs. The results showed that the mRNA levels of PAK1, PAK2, PAK4 and PAK6 were significantly up-regulated in breast cancer compared with normal tissues, while the reverse trend for PAK3 and PAK5 was found, furthermore, the proteins expression of PAK1, PAK2 and PAK4 in breast cancer tissues were higher than that in normal breast tissues. Survival analysis revealed breast cancer patients with low mRNA expression of PAK3 and PAK5 showed worse RFS, conversely, elevated PAK4 levels predicted worse RFS. In addition, the breast cancer patients with PAKs genetic alterations correlated with worse OS. These results indicated that PAKs might be promising potential biomarkers for breast cancer.
Collapse
Affiliation(s)
- Yifang Dang
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Ying Guo
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiaoyu Ma
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiaoyu Chao
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Fei Wang
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Linghao Cai
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Zhongyi Yan
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Longxiang Xie
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiangqian Guo
- Department of Predictive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| |
Collapse
|
10
|
Huo FC, Pan YJ, Li TT, Mou J, Pei DS. PAK5 promotes the migration and invasion of cervical cancer cells by phosphorylating SATB1. Cell Death Differ 2018; 26:994-1006. [PMID: 30082769 DOI: 10.1038/s41418-018-0178-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023] Open
Abstract
p21-activated kinase 5 (PAK5) is involved in several oncogenic signaling pathways and its amplification or overexpression has been found in various types of cancer; however, the pathophysiologic role of PAK5 in cervical cancer (CC) remains elusive. This study aims to elucidate the effects of PAK5 on CC metastasis and its specific regulation mechanism. We performed western blotting and immunohistochemistry (IHC) analysis and found that the expression levels of PAK5 were significantly upregulated in CC cells and tissues. In addition, statistical analysis via IHC showed that increased PAK5 significantly correlated with CC progression. Mn2+-Phos-tag SDS-PAGE, western blotting, immunofluorescence and dual luciferase reporter assays were utilized to determine the involvement of SATB1 in PAK5-mediated epithelial-mesenchymal transition (EMT). We found that PAK5-mediated special AT-rich binding protein-1 (SATB1) phosphorylation on Ser47 initiated EMT cascade and promoted migration and invasion of CC cells. Furthermore, overexpression of PAK5 induced lung metastasis of CC cells in xenograft modes. Taken together, we conclude that PAK5 is a novel prognostic indicator and plays an important role in the CC metastasis.
Collapse
Affiliation(s)
- Fu-Chun Huo
- Department of pathology, Xuzhou Medical University, Xuzhou, 221004, China.,Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, 221002, China
| | - Yao-Jie Pan
- Department of Oncology, The Affiliated Yancheng Hospital of Medicine School of Southeast University, Yancheng, 224001, China
| | - Tong-Tong Li
- Department of pathology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jie Mou
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Dong-Sheng Pei
- Department of pathology, Xuzhou Medical University, Xuzhou, 221004, China. .,Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
11
|
Li K, Xu X, He Y, Tian Y, Pan W, Xu L, Ma Y, Gao Y, Gao J, Qi Y, Wei L, Zhang J. P21-activated kinase 7 (PAK7) interacts with and activates Wnt/β-catenin signaling pathway in breast cancer. J Cancer 2018; 9:1821-1835. [PMID: 29805709 PMCID: PMC5968771 DOI: 10.7150/jca.24934] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 02/22/2018] [Indexed: 12/30/2022] Open
Abstract
Background: Breast cancer is the highest incidence of tumor in women, which seriously threaten women's health. The occurrence and progression of breast cancer is linked to inactivation or downregulation of tumor suppressors, and activation or upregulation of oncogenes. However, the mechanism of PAK7 involving in the occurrence and progression of breast cancer is not yet fully understood. Methods: PAK7 expression was analyzed by RT-qPCR and immunohistochemistry and correlated with clinicopatholgical parameters in breast cancer tissue microarray. The effects of PAK7 on breast cancer cells were detected by CCK-8 assay, colon formation assay, wound healing and transwell assays, and flow cytometry. The relationship between PAK7 and Wnt/β-catenin signaling pathway was determined by western blotting, TOP/FOP flash, co-Immunoprecipitation and co-localization assays. Results: PAK7 expression was significantly increased in breast cancer tissues and positively correlated with pathological differentiation and TNM stage of breast cancer. Overexpression of PAK7 could significantly promote proliferation and migration of breast cancer cells, and inhibit apoptosis. In contrast, PAK7 knockdown significantly inhibited the proliferation and migration of breast cancer cells and promoted apoptosis. In addition, PAK7 could activate Wnt/β-catenin signaling pathway in breast cancer cells. Further study found that PAK7 could directly bind to GSK3β and β-catenin, and regulate β-catenin degradation by phosphorylating GSK3β. Conclusions: Our study demonstrated that PAK7, as an oncogene, involved in breast cancer progression by activating the Wnt/β-catenin signaling pathway, suggesting that the potential applicability of PAK7 as a target for breast cancer treatment.
Collapse
Affiliation(s)
- Kai Li
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Xiaolong Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yanqi He
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yihao Tian
- Department of Anatomy, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Wenting Pan
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Liu Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yanbin Ma
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yang Gao
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| | - Jingbo Gao
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| | - Yuwen Qi
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| | - Lei Wei
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Jingwei Zhang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
12
|
|
13
|
Zhang YC, Huo FC, Wei LL, Gong CC, Pan YJ, Mou J, Pei DS. PAK5-mediated phosphorylation and nuclear translocation of NF-κB-p65 promotes breast cancer cell proliferation in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:146. [PMID: 29041983 PMCID: PMC5645986 DOI: 10.1186/s13046-017-0610-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
Background Abnormal proliferation is significantly associated with the promotion of malignant tumor. Growing evidence suggest that the signal pathways of p21cdc42/rac1-activated kinase 5 (PAK5) have been found in various tumor progression, however, the role of PAK5 in breast cancer remains largely unclear. Methods We evaluated PAK5 and p65 staining in breast cancer tissues (BCTs) and paired non-cancerous tissues (NTs) using tissue microarray (TMA) technology. The functions of PAK5 were studied in vitro and in vivo. Cell Counting Kit-8 (CCK-8) and flow cytometry were performed to determine proliferation of breast cancer cells. Phosphorylation assay and co-immunoprecipitation (co-IP) were employed to identify the regulation mechanism of p65 by PAK5. The activation of Cyclin D1 promoter was measured with luciferase reporter assay. Xenograft models in nude mice were established to explore the roles of PAK5 in breast cancer growth. Results In this study, we show that PAK5 is highly expressed in breast cancer tissues and the increased PAK5 is significantly associated with breast cancer progression. Overexpression of PAK5 promotes the proliferation and cell-cycle progression by increasing the expression of Cyclin D1 in vitro and in vivo. Mechanistic studies demonstrated that PAK5 can promote the phosphorylation and the nuclear translocation of p65 subunit of nuclear factor-kappaB (NF-κB). Furthermore, p65 can directly bind to the promoter of Cyclin D1 and mediate an increase in its protein expression. Conclusions Taken together, our findings suggest that PAK5 may serve as a potential prognosis marker and therapeutic target for human breast cancer.
Collapse
Affiliation(s)
- Ying-Chun Zhang
- Department of pathology, Xuzhou Medical University, Xuzhou, 221002, China.,Department of Interventional Radiology, Jining No.1 People's Hospital, Jining, Shandong Province, China
| | - Fu-Chun Huo
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, 221002, China
| | - Lu-Lu Wei
- Department of pathology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Chan-Chan Gong
- Department of pathology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Yao-Jie Pan
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jie Mou
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Dong-Sheng Pei
- Department of pathology, Xuzhou Medical University, Xuzhou, 221002, China. .,Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
14
|
Song X, Xie Y, Liu Y, Shao M, Yang W. MicroRNA-492 overexpression exerts suppressive effects on the progression of osteosarcoma by targeting PAK7. Int J Mol Med 2017; 40:891-897. [DOI: 10.3892/ijmm.2017.3046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/24/2017] [Indexed: 11/06/2022] Open
|
15
|
Zhan L, Pan Y, Chen L, Chen Z, Zhang H, Sun C. MicroRNA-526a targets p21-activated kinase 7 to inhibit tumorigenesis in hepatocellular carcinoma. Mol Med Rep 2017; 16:837-844. [PMID: 28560394 DOI: 10.3892/mmr.2017.6658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 01/19/2017] [Indexed: 01/11/2023] Open
Abstract
MicroRNAs belong to a series of noncoding RNAs and have diverse roles in several biological processes. The association between aberrant microRNA expression and tumorigenesis is complex and remains to be fully elucidated. The present study investigated whether microRNA (miR) ‑526a can suppress the progression of hepatocellular carcinoma (HCC) in vitro and in vivo. Reverse transcription‑quantitative polymerase chain reaction, luciferase reporter assay, invasion assay, western blotting and in vivo implantation were used to investigate the potential function of miR‑526a. The present study observed that the level of miR‑526a was downregulated in HCC tissues and well‑established cell lines. In addition, the ectopic introduction of miR‑526a into Huh7 and HepG2 cells significantly attenuated HCC tumorigenesis, including proliferation, migration and invasion. The growth of tumor xenografts was also inhibited following transfection with miR‑526a. Using overlapping strategies, p21‑activated kinase 7 (PAK7) was predicted to be a target for miR‑526a, and this was verified experimentally. An inverse correlation was found between miR‑526a and PAK7 in HCC tissues. The results of the present study revealed a novel function of miR‑526a and may provide crucial insight into therapeutic interventions targeting microRNAs.
Collapse
Affiliation(s)
- Lei Zhan
- Department of Biliary‑Hepatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Yaozhen Pan
- Department of Biliary‑Hepatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Ling Chen
- Department of Biliary‑Hepatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Zili Chen
- Department of Biliary‑Hepatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Hong Zhang
- Department of Biliary‑Hepatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Chengyi Sun
- Department of Biliary‑Hepatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| |
Collapse
|
16
|
Zhao ZX, Li X, Liu WD, Liu XZ, Wu SJ, Hu XH. Inhibition of Growth and Metastasis of Tumor in Nude Mice after Intraperitoneal Injection of Bevacizumab. Orthop Surg 2017; 8:234-40. [PMID: 27384733 PMCID: PMC6593177 DOI: 10.1111/os.12236] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/19/2016] [Indexed: 01/16/2023] Open
Abstract
Objective To explore the inhibitory effect of bevacizumab, a vascular endothelial growth factor antibody, on angiogenesis in human osteosarcoma of nude mice. Methods Twenty‐one nude mice were inoculated with red fluorescent protein (RFP)‐labeled human osteosarcoma cell line 143B‐RFP, that is, clones that expressed RFP in the cytoplasm, and randomly assigned to one of three groups: G1 (Control group, injected with saline solution); G2 (intraperitoneal bevacizumab 2 mg/kg twice per week) and G3 (intraperitoneal bevacizumab 5 mg/kg, twice per week). The tumor‐bearing mice were examined in a fluorescence light box that was illuminated periodically. The primary tumors were measured by fluorescence imaging weekly and their volumes calculated. Results The mean tumor volumes were significantly smaller in the G3 (186.4 ± 100.8 mm3) than the control group (587.0 ± 406.8 mm3) (P < 0.05) on Day 31, and again significantly smaller in the G3 (677.3 ± 461.9 mm3) than the control group (3162.6 ± 1529.2 mm3) on Day 38 (P < 0.01). The average tumor volume in the G2 group was 493.5 ± 425.4 mm3 on Day 31 and 1870.1 ± 1524.8 mm3 on Day 38. The effect on tumor volume was greater in the G3 than the G2 group. Three mice in the G2 group, four in the G3 group and four in the control group developed lung metastases that were confirmed by pathological examination; these differences were not statistically significant (P < 0.05). Conclusions Bevacizumab exhibits strong antiangiogenesis activity in experimental osteosarcoma in a nude mouse model but does not influence the incidence of lung metastasis. Our findings may have considerable potential for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Ze-Xue Zhao
- Department of Orthopaedics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Orthopaedics, Huaian First People's Hospital, Nanjing Medical University, Huaian, China
| | - Xiang Li
- Department of Orthopaedics, Huaian First People's Hospital, Nanjing Medical University, Huaian, China
| | - Wei-Dong Liu
- Department of Orthopaedics, Huaian First People's Hospital, Nanjing Medical University, Huaian, China
| | - Xiao-Zhou Liu
- Department of Orthopaedics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Su-Jia Wu
- Department of Orthopaedics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiao-Hui Hu
- Department of Orthopaedics, Huaian First People's Hospital, Nanjing Medical University, Huaian, China
| |
Collapse
|
17
|
PAK5 is auto-activated by a central domain that promotes kinase oligomerization. Biochem J 2016; 473:1777-89. [DOI: 10.1042/bcj20160132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 04/18/2016] [Indexed: 12/23/2022]
Abstract
The present study shows for the first time that self-association of PAK5 in vivo underlies its high basal activity, which contrasts with the inactive state of cellular PAK4. Such PAK5 self-association interferes with the engagement of the auto-inhibitory (AID) with the catalytic domain.
Collapse
|
18
|
Kumar R, Li DQ. PAKs in Human Cancer Progression: From Inception to Cancer Therapeutic to Future Oncobiology. Adv Cancer Res 2016; 130:137-209. [PMID: 27037753 DOI: 10.1016/bs.acr.2016.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since the initial recognition of a mechanistic role of p21-activated kinase 1 (PAK1) in breast cancer invasion, PAK1 has emerged as one of the widely overexpressed or hyperactivated kinases in human cancer at-large, allowing the PAK family to make in-roads in cancer biology, tumorigenesis, and cancer therapeutics. Much of our current understanding of the PAK family in cancer progression relates to a central role of the PAK family in the integration of cancer-promoting signals from cell membrane receptors as well as function as a key nexus-modifier of complex, cytoplasmic signaling network. Another core aspect of PAK signaling that highlights its importance in cancer progression is through PAK's central role in the cross talk with signaling and interacting proteins, as well as PAK's position as a key player in the phosphorylation of effector substrates to engage downstream components that ultimately leads to the development cancerous phenotypes. Here we provide a comprehensive review of the recent advances in PAK cancer research and its downstream substrates in the context of invasion, nuclear signaling and localization, gene expression, and DNA damage response. We discuss how a deeper understanding of PAK1's pathobiology over the years has widened research interest to the PAK family and human cancer, and positioning the PAK family as a promising cancer therapeutic target either alone or in combination with other therapies. With many landmark findings and leaps in the progress of PAK cancer research since the infancy of this field nearly 20 years ago, we also discuss postulated advances in the coming decade as the PAK family continues to shape the future of oncobiology.
Collapse
Affiliation(s)
- R Kumar
- School of Medicine and Health Sciences, George Washington University, Washington, DC, United States; Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram, India.
| | - D-Q Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China; Key Laboratory of Epigenetics in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Sosa-García B, Vázquez-Rivera V, González-Flores JN, Engel BE, Cress WD, Santiago-Cardona PG. The Retinoblastoma Tumor Suppressor Transcriptionally Represses Pak1 in Osteoblasts. PLoS One 2015; 10:e0142406. [PMID: 26555075 PMCID: PMC4640669 DOI: 10.1371/journal.pone.0142406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/21/2015] [Indexed: 12/26/2022] Open
Abstract
We previously characterized the retinoblastoma tumor suppressor protein (Rb) as a regulator of adherens junction assembly and cell-to-cell adhesion in osteoblasts. This is a novel function since Rb is predominantly known as a cell cycle repressor. Herein, we characterized the molecular mechanisms by which Rb performs this function, hypothesizing that Rb controls the activity of known regulators of adherens junction assembly. We found that Rb represses the expression of the p21-activated protein kinase (Pak1), an effector of the small Rho GTPase Rac1. Rac1 is a well-known regulator of adherens junction assembly whose increased activity in cancer is linked to perturbations of intercellular adhesion. Using nuclear run-on and luciferase reporter transcription assays, we found that Pak1 repression by Rb is transcriptional, without affecting Pak1 mRNA and protein stability. Pak1 promoter bioinformatics showed multiple E2F1 binding sites within 155 base pairs of the transcriptional start site, and a Pak1-promoter region containing these E2F sites is susceptible to transcriptional inhibition by Rb. Chromatin immunoprecipitations showed that an Rb-E2F complex binds to the region of the Pak1 promoter containing the E2F1 binding sites, suggesting that Pak1 is an E2F target and that the repressive effect of Rb on Pak1 involves blocking the trans-activating capacity of E2F. A bioinformatics analysis showed elevated Pak1 expression in several solid tumors relative to adjacent normal tissue, with both Pak1 and E2F increased relative to normal tissue in breast cancer, supporting a cancer etiology for Pak1 up-regulation. Therefore, we propose that by repressing Pak1 expression, Rb prevents Rac1 hyperactivity usually associated with cancer and related to cytoskeletal derangements that disrupt cell adhesion, consequently enhancing cancer cell migratory capacity. This de-regulation of cell adhesion due to Rb loss could be part of the molecular events associated with cancer progression and metastasis.
Collapse
Affiliation(s)
- Bernadette Sosa-García
- Department of Basic Sciences, Biochemistry Division, Ponce Health Science University, Ponce, Puerto Rico
| | - Viviana Vázquez-Rivera
- Department of Basic Sciences, Biochemistry Division, Ponce Health Science University, Ponce, Puerto Rico
| | | | - Brienne E. Engel
- Molecular Oncology and Thoracic Oncology Departments, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States of America
| | - W. Douglas Cress
- Molecular Oncology and Thoracic Oncology Departments, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States of America
| | - Pedro G. Santiago-Cardona
- Department of Basic Sciences, Biochemistry Division, Ponce Health Science University, Ponce, Puerto Rico
- * E-mail:
| |
Collapse
|