1
|
Chen XY, Cheng AY, Wang ZY, Jin JM, Lin JY, Wang B, Guan YY, Zhang H, Jiang YX, Luan X, Zhang LJ. Dbl family RhoGEFs in cancer: different roles and targeting strategies. Biochem Pharmacol 2024; 223:116141. [PMID: 38499108 DOI: 10.1016/j.bcp.2024.116141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Small Ras homologous guanosine triphosphatase (Rho GTPase) family proteins are highly associated with tumorigenesis and development. As intrinsic exchange activity regulators of Rho GTPases, Rho guanine nucleotide exchange factors (RhoGEFs) have been demonstrated to be closely involved in tumor development and received increasing attention. They mainly contain two families: the diffuse B-cell lymphoma (Dbl) family and the dedicator of cytokinesis (Dock) family. More and more emphasis has been paid to the Dbl family members for their abnormally high expression in various cancers and their correlation to poor prognosis. In this review, the common and distinctive structures of Dbl family members are discussed, and their roles in cancer are summarized with a focus on Ect2, Tiam1/2, P-Rex1/2, Vav1/2/3, Trio, KALRN, and LARG. Significantly, the strategies targeting Dbl family RhoGEFs are highlighted as novel therapeutic opportunities for cancer.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ao-Yu Cheng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zi-Ying Wang
- School of Biological Engineering, Tianjin University of Science&Technology, Tianjin 301617, China
| | - Jin-Mei Jin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Yi Lin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bei Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying-Yun Guan
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Hao Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Xin Jiang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Ginn L, Maltas J, Baker MJ, Chaturvedi A, Wilson L, Guilbert R, Amaral FMR, Priest L, Mole H, Blackhall F, Diamantopoulou Z, Somervaille TCP, Hurlstone A, Malliri A. A TIAM1-TRIM28 complex mediates epigenetic silencing of protocadherins to promote migration of lung cancer cells. Proc Natl Acad Sci U S A 2023; 120:e2300489120. [PMID: 37748077 PMCID: PMC10556593 DOI: 10.1073/pnas.2300489120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/03/2023] [Indexed: 09/27/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths. Its high mortality is associated with high metastatic potential. Here, we show that the RAC1-selective guanine nucleotide exchange factor T cell invasion and metastasis-inducing protein 1 (TIAM1) promotes cell migration and invasion in the most common subtype of lung cancer, non-small-cell lung cancer (NSCLC), through an unexpected nuclear function. We show that TIAM1 interacts with TRIM28, a master regulator of gene expression, in the nucleus of NSCLC cells. We reveal that a TIAM1-TRIM28 complex promotes epithelial-to-mesenchymal transition, a phenotypic switch implicated in cell migration and invasion. This occurs through H3K9me3-induced silencing of protocadherins and by decreasing E-cadherin expression, thereby antagonizing cell-cell adhesion. Consistently, TIAM1 or TRIM28 depletion suppresses the migration of NSCLC cells, while migration is restored by the simultaneous depletion of protocadherins. Importantly, high nuclear TIAM1 in clinical specimens is associated with advanced-stage lung adenocarcinoma, decreased patient survival, and inversely correlates with E-cadherin expression.
Collapse
Affiliation(s)
- Lucy Ginn
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Joe Maltas
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Martin J. Baker
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Anshuman Chaturvedi
- The Christie National Health Service Foundation Trust, ManchesterM20 4BX, United Kingdom
| | - Leah Wilson
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Ryan Guilbert
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Fabio M. R. Amaral
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Lynsey Priest
- The Christie National Health Service Foundation Trust, ManchesterM20 4BX, United Kingdom
| | - Holly Mole
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, ManchesterM13 9PT, United Kingdom
| | - Fiona Blackhall
- The Christie National Health Service Foundation Trust, ManchesterM20 4BX, United Kingdom
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, ManchesterM13 9PT, United Kingdom
| | - Zoi Diamantopoulou
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Tim C. P. Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| | - Adam Hurlstone
- Division of Immunology, Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, ManchesterM13 9PT, United Kingdom
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, ManchesterM20 4BX, United Kingdom
| |
Collapse
|
3
|
Editor's Note: Balanced Tiam1-Rac1 and RhoA Drives Proliferation and Invasion of Pancreatic Cancer Cells. Mol Cancer Res 2022; 20:1822. [DOI: 10.1158/1541-7786.mcr-22-0795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
4
|
Seyed Salehi A, Parsa-Nikoo N, Roshan-Farzad F, Shams R, Fathi M, Asaszadeh Aghdaei H, Behmanesh A. MicroRNA-125a-3p, -4530, and -92a as a Potential Circulating MicroRNA Panel for Noninvasive Pancreatic Cancer Diagnosis. DISEASE MARKERS 2022; 2022:8040419. [PMID: 36254252 PMCID: PMC9569215 DOI: 10.1155/2022/8040419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 12/24/2022]
Abstract
MicroRNA (miRNA) expression dysregulations in pancreatic ductal adenocarcinoma (PDAC) have been studied widely for their diagnostic and prognostic utility. By the use of bioinformatics-based methods, in our previous study, we identified some potential miRNA panels for diagnosis of pancreatic cancer patients from noncancerous controls (the screening stage). In this report, we used 142 plasma samples from people with and without pancreatic cancer (PC) to conduct RT-qPCR differential expression analysis to assess the strength of the first previously proposed diagnostic panel (consisting of miR-125a-3p, miR-4530, and miR-92a-2-5p). As the result, we identified significant upregulation for all the three considered miRNAs in the serum of PC patients. After that, a three-miRNA panel in serum was developed. The area under the receiver operating characteristic curves (AUC) for the panel were 0.850, 0.910, and 0.86, respectively, indicating that it had a higher diagnostic value than individual miRNAs. Therefore, we detected a promising three-miRNA panel in the plasma for noninvasive PC diagnosis (miR-125a-3p, miR-4530, and miR-92a-2-5p).
Collapse
Affiliation(s)
- Ali Seyed Salehi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology, Faculty of Biological Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Negar Parsa-Nikoo
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology, Faculty of Biological Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Farnaz Roshan-Farzad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology, Faculty of Biological Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Fathi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asaszadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Behmanesh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Förster resonance energy transfer biosensors for fluorescence and time-gated luminescence analysis of rac1 activity. Sci Rep 2022; 12:5291. [PMID: 35351946 PMCID: PMC8964680 DOI: 10.1038/s41598-022-09364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/08/2022] [Indexed: 11/24/2022] Open
Abstract
Genetically encoded, Förster resonance energy transfer (FRET) biosensors enable live-cell optical imaging of signaling molecules. Small conformational changes often limit the dynamic range of biosensors that combine fluorescent proteins (FPs) and sensing domains into a single polypeptide. To address this, we developed FRET and lanthanide-based FRET (LRET) biosensors of Rac1 activation with two key features that enhance sensitivity and dynamic range. For one, alpha helical linker domains separate FRET partners and ensure a large conformational change and FRET increase when activated Rac1 at the biosensor C-terminus interacts with an amino-terminal Rac binding domain. Incorporation of a luminescent Tb(III) complex with long (~ ms) excited state lifetime as a LRET donor enabled time-gated luminescence measurements of Rac1 activity in cell lysates. The LRET dynamic range increased with ER/K linker length up to 1100% and enabled robust detection of Rac1 inhibition in 96-well plates. The ER/K linkers had a less pronounced, but still significant, effect on conventional FRET biosensors (with FP donors and acceptors), and we were able to dynamically image Rac1 activation at cell edges using fluorescence microscopy. The results herein highlight the potential of FRET and LRET biosensors with ER/K linkers for cell-based imaging and screening of protein activities.
Collapse
|
6
|
Wang J, Yuan L, Xu X, Zhang Z, Ma Y, Hong L, Ma J. Rho-GEF Trio regulates osteosarcoma progression and osteogenic differentiation through Rac1 and RhoA. Cell Death Dis 2021; 12:1148. [PMID: 34893584 PMCID: PMC8664940 DOI: 10.1038/s41419-021-04448-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 11/08/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone tumor. Its high mortality rate and metastasis rate seriously threaten human health. Currently, the treatment has reached a plateau, hence we urgently need to explore new therapeutic directions. In this paper, we found that Trio was highly expressed in osteosarcoma than normal tissues and promoted the proliferation, migration, and invasion of osteosarcoma cells. Furthermore, Trio inhibited osteosarcoma cells' osteogenic differentiation in vitro and accelerated the growth of osteosarcoma in vivo. Given Trio contains two GEF domains, which have been reported as the regulators of RhoGTPases, we further discovered that Trio could regulate osteosarcoma progression and osteogenic differentiation through activating RhoGTPases. In summary, all our preliminary results showed that Trio could be a potential target and prognostic marker of osteosarcoma.
Collapse
Affiliation(s)
- Junyi Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, 210029, Nanjing, China
| | - Lichan Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, 210029, Nanjing, China
| | - Xiaohong Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, 210029, Nanjing, China
| | - Zhongyin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, 210029, Nanjing, China
| | - Yuhuan Ma
- Nanjing Foreign Language School, 210008, Nanjing, Jiangsu, China
| | - Leilei Hong
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, 210029, Nanjing, China
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, 210029, Nanjing, China.
| |
Collapse
|
7
|
Li F, Zhou YD, Liu J, Cai JD, Liao ZM, Chen GQ. RBP-J promotes cell growth and metastasis through regulating miR-182-5p-mediated Tiam1/Rac1/p38 MAPK axis in colorectal cancer. Cell Signal 2021; 87:110103. [PMID: 34339855 DOI: 10.1016/j.cellsig.2021.110103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND RBP-J is involved in number of cellular processes. However, the potential mechanisms of RBP-J on colorectal cancer (CRC) development have not been clearly defined. In this study, we aimed to investigate the role and molecular mechanism of RBP-J in CRC. METHODS The expression levels of RBP-J and Tiam1 in CRC tissues and cells were evaluated by RT-qPCR or western blot. RBP-J was knocked down with sh-RBP-J or overexpressed by pcDNA3.1-RBP-J in CRC cells. Cell proliferation, migration and invasion abilities were analyzed by MTT, wound healing, and transwell assay, respectively. CHIP-qPCR, RIP and dual luciferase reporter assays were performed to confirm the interaction between miR-182-5p and RBP-J or Tiam1. Expression levels of p-p38 MAPK, p38 MAPK, Slug-1, Twist1 and MMP-9 were analyzed by western blot. G-LISA test was used to detect Rac1 activity. RESULTS Our results showed that the expression of RBP-J and Tiam1 was significantly up-regulated in CRC tissues and cells. RBP-J overexpression promoted proliferation, migration and invasion of CRC cells. Moreover, RBP-J was found to directly target miR-182-5p promoter and positively regulate the Tiam1/Rac1/p38 MAPK signaling pathway in CRC cells. It was also proved that miR-182-5p can bind Tiam1 directly. Furthermore, experiments revealed that RBP-J could promote CRC cell proliferation, migration and invasion via miR-182-5p-mediated Tiam1/Rac1/p38 MAPK axis. In addition, knockdown of RBP-J reduced tumor growth and metastasis in CRC mice. CONCLUSION RBP-J regulates CRC cell growth and metastasis through miR-182-5p mediated Tiam1/Rac1/p38 MAPK signaling pathway, implying potential novel therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, The Fourth Hospital of Changsha City, Changsha 410006, Hunan Province, PR China
| | - Ya-Dong Zhou
- Department of Pathology, The Fourth Hospital of Changsha City, Changsha 410006, Hunan Province, PR China
| | - Jiao Liu
- Department of Pathology, The Fourth Hospital of Changsha City, Changsha 410006, Hunan Province, PR China
| | - Jiao-Di Cai
- Department of Pathology, The Fourth Hospital of Changsha City, Changsha 410006, Hunan Province, PR China
| | - Zhi-Ming Liao
- Department of Pathology, The Fourth Hospital of Changsha City, Changsha 410006, Hunan Province, PR China
| | - Guo-Qun Chen
- Department of Pathology, The Fourth Hospital of Changsha City, Changsha 410006, Hunan Province, PR China.
| |
Collapse
|
8
|
Maltas J, Reed H, Porter A, Malliri A. Mechanisms and consequences of dysregulation of the Tiam family of Rac activators in disease. Biochem Soc Trans 2020; 48:2703-2719. [PMID: 33200195 DOI: 10.1042/bst20200481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022]
Abstract
The Tiam family proteins - Tiam1 and Tiam2/STEF - are Rac1-specific Guanine Nucleotide Exchange Factors (GEFs) with important functions in epithelial, neuronal, immune and other cell types. Tiam GEFs regulate cellular migration, proliferation and survival, mainly through activating and directing Rac1 signalling. Dysregulation of the Tiam GEFs is significantly associated with human diseases including cancer, immunological and neurological disorders. Uncovering the mechanisms and consequences of dysregulation is therefore imperative to improving the diagnosis and treatment of diseases. Here we compare and contrast the subcellular localisation and function of Tiam1 and Tiam2/STEF, and review the evidence for their dysregulation in disease.
Collapse
Affiliation(s)
- Joe Maltas
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, U.K
| | - Hannah Reed
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, U.K
| | - Andrew Porter
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, U.K
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, U.K
| |
Collapse
|
9
|
Li M, Zhao J, Li X, Chen Y, Feng C, Qian F, Liu Y, Zhang J, He J, Ai B, Ning Z, Liu W, Bai X, Han X, Wu Z, Xu X, Tang Z, Pan Q, Xu L, Li C, Wang Q, Li E. HiFreSP: A novel high-frequency sub-pathway mining approach to identify robust prognostic gene signatures. Brief Bioinform 2020; 21:1411-1424. [PMID: 31350847 DOI: 10.1093/bib/bbz078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/19/2019] [Accepted: 06/04/2019] [Indexed: 02/05/2023] Open
Abstract
With the increasing awareness of heterogeneity in cancers, better prediction of cancer prognosis is much needed for more personalized treatment. Recently, extensive efforts have been made to explore the variations in gene expression for better prognosis. However, the prognostic gene signatures predicted by most existing methods have little robustness among different datasets of the same cancer. To improve the robustness of the gene signatures, we propose a novel high-frequency sub-pathways mining approach (HiFreSP), integrating a randomization strategy with gene interaction pathways. We identified a six-gene signature (CCND1, CSF3R, E2F2, JUP, RARA and TCF7) in esophageal squamous cell carcinoma (ESCC) by HiFreSP. This signature displayed a strong ability to predict the clinical outcome of ESCC patients in two independent datasets (log-rank test, P = 0.0045 and 0.0087). To further show the predictive performance of HiFreSP, we applied it to two other cancers: pancreatic adenocarcinoma and breast cancer. The identified signatures show high predictive power in all testing datasets of the two cancers. Furthermore, compared with the two popular prognosis signature predicting methods, the least absolute shrinkage and selection operator penalized Cox proportional hazards model and the random survival forest, HiFreSP showed better predictive accuracy and generalization across all testing datasets of the above three cancers. Lastly, we applied HiFreSP to 8137 patients involving 20 cancer types in the TCGA database and found high-frequency prognosis-associated pathways in many cancers. Taken together, HiFreSP shows higher prognostic capability and greater robustness, and the identified signatures provide clinical guidance for cancer prognosis. HiFreSP is freely available via GitHub: https://github.com/chunquanlipathway/HiFreSP.
Collapse
Affiliation(s)
- Meng Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jianmei Zhao
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xuecang Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yang Chen
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Chenchen Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Fengcui Qian
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yuejuan Liu
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jian Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Jianzhong He
- Institute of Oncologic Pathology, Shantou University Medical College
| | - Bo Ai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Ziyu Ning
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Wei Liu
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Xuefeng Bai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Xiaole Han
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Zhiyong Wu
- Departments of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University
| | - Xiue Xu
- Institute of Oncologic Pathology, Shantou University Medical College
| | - Zhidong Tang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Qi Pan
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Liyan Xu
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Qiuyu Wang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Enmin Li
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| |
Collapse
|
10
|
Abstract
BACKGROUND A number of studies have attempted to determine the prognostic value of T-cell lymphoma invasion and metastasis-inducing factor 1 (Tiam1) in patients with solid cancers, but the reported results were of inconsistency. Thus, we performed a systematic review and meta-analysis to exhaustively evaluate the prognostic role of Tiam1 expression in patients with solid cancers. METHODS We retrieved literature published in between 1994 and April 22th, 2019 through searching PubMed, Web of Science and China national knowledge infrastructure (CNKI). Hazard ratios (HRs) coupled with 95% confidence intervals (95% CIs) were used to assess the relationship of Tiam1 expression and overall survival (OS), and disease-free survival (DFS). RESULTS A total of 2647 patients with solid cancers in 20 studies were enrolled in our meta-analysis eventually. The pooled results showed that Tiam1 high expression was closely correlated with poor OS (HR = 2.17, 95% CI: 1.80-2.61, P = .000) and DFS (pooled HR = 1.95, 95% CI = 1.58-2.40, P = .000). Moreover, our subgroup analysis and sensitivity analysis demonstrated the reliability and stability of our pooled results. CONCLUSION In conclusion, this meta-analysis confirmed that Tiam1 higher expression positively correlated with OS and DFS, suggesting that Tiam1 may act as a valuable prognostic predictor and therapeutic target for patients with solid cancers. Nevertheless, in future more homogeneous and prospective studies should be performed to further support our findings.
Collapse
|
11
|
Yang C, Ma C, Li Y, Mo P, Yang Y. High Tiam1 expression predicts positive lymphatic metastasis and worse survival in patients with malignant solid tumors: a systematic review and meta-analysis. Onco Targets Ther 2019; 12:5925-5936. [PMID: 31413590 PMCID: PMC6663076 DOI: 10.2147/ott.s191571] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Background Many studies have explored the prognostic value of T-cell lymphoma invasion and metastasis inducing factor 1 (Tiam1) and its association with lymphatic metastasis in malignant solid tumors, but the conclusions remain controversial. Therefore, we performed a meta-analysis to systematically assess the prognostic value of Tiam1 expression and its association with lymphatic metastasis in malignant solid tumors. Methods We searched eligible studies in PubMed, Web of Science and EMBASE databases (from inception up to October 2018). The combined HR with 95% CI was used to estimate the prognostic value of Tiam1 expression. The correlation between Tiam1 expression and lymphatic metastasis was assessed using the combined odds ratio (OR) with 95% CI. Results A total of 17 studies with 2,228 patients with solid tumors were included in this meta-analysis. The overall estimated results showed that high Tiam1 expression was significantly associated with shorter overall survival (HR= 2.08, 95% CI: 1.62-2.68, P<0.01), and disease-free survival (HR = 1.86, 95% CI: 1.49-2.32, P<0.01). Besides, we also found that there was a close relationship between high Tiam1 expression and positive lymphatic metastasis (OR=2.63; 95% CI: 1.79-3.84, P<0.01). Conclusion High Tiam1 expression was significantly associated with shorter survival and positive lymphatic metastasis in patients with malignant solid tumors. Therefore, Tiam1 may be a promising prognostic biomarker and an effective therapeutic target for malignant solid tumors.
Collapse
Affiliation(s)
- Caixia Yang
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Chenlin Ma
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Yingchun Li
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Peng Mo
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Yusheng Yang
- Department of Pathology, Ninbo Yinzhou No. 2 Hospital, Ninbo, People's Republic of China
| |
Collapse
|
12
|
Liu Y, Wang X, Jiang X, Yan P, Zhan L, Zhu H, Wang T, Wen J. Tumor-suppressive microRNA-10a inhibits cell proliferation and metastasis by targeting Tiam1 in esophageal squamous cell carcinoma. J Cell Biochem 2019; 120:7845-7857. [PMID: 30426564 DOI: 10.1002/jcb.28059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Aberrant microRNAs (miRNAs) expressions could contribute to the progression of numerous cancers, including esophageal squamous cell carcinoma, while miR-10a participates in multiple biological processes on cancers. However, the molecular mechanism of miR-10a in esophageal squamous cell carcinoma (ESCC) has not been investigated. Herein, miR-10a was significantly reduced in ESCC clinical tissues and ESCC cell lines (EC109 and TE-3). In addition, immunohistochemistry indicated that the expressions of α-SMA, Ki-67, and PCNA in tumor tissues were higher than that of controls. In vitro, overexpression of miR-10a dramatically suppressed cell proliferation and enhanced cell apoptosis, while the decrease of miR-10a expressed the opposite outcome. Specially, overexpression of miR-10a caused a G0/G1 peak accumulation. Moreover, miR-10a also negatively regulated ESCC cell migration and invasion. Furthermore, targetscan bioinformatics predictions and the dual-luciferase assay confirmed that Tiam1 was a direct target gene of miR-10a. The statistical analysis showed Tiam1 was negatively in correlation with miR-10a in ESCC patient samples. And silencing Tiam1 could lead to a decline on cell growth, invasion, and migration in ESCC cell lines, while it could enhance cell apoptosis and cause a G0/G1 peak accumulation. In vivo, it revealed that miR-10a notably decreased the tumor growth and metastasis in xenograft model and pulmonary metastasis model. And it showed a lower expressions of Tiam1 in the miR-10a mimics group by immunohistochemistry. Taken together the results, they indicated that miR-10a might function as a novel tumor suppressor in vitro and in vivo via targeting Tiam1, suggesting miR-10a to be a candidate biomarker for the ESCC therapy.
Collapse
Affiliation(s)
- Yatian Liu
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuesong Jiang
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengwei Yan
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liangliang Zhan
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huanfeng Zhu
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tingting Wang
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Wen
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Dai J, Ma J, Yu B, Zhu Z, Hu Y. [ARTICLE WITHDRAWN] Long Noncoding RNA TUNAR Represses Growth, Migration, and Invasion of Human Glioma Cells Through Regulating miR-200a and Rac1. Oncol Res 2018; 27:107-115. [PMID: 29540255 PMCID: PMC7848266 DOI: 10.3727/096504018x15205622257163] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
THIS ARTICLE WAS WITHDRAWN BY THE PUBLISHERS IN NOVEMBER 2020.
Collapse
Affiliation(s)
- Jinhua Dai
- *Department of Clinical Laboratory, Ningbo No. 2 Hospital, Ningbo, P.R. China
| | - Jianbo Ma
- *Department of Clinical Laboratory, Ningbo No. 2 Hospital, Ningbo, P.R. China
| | - Bixia Yu
- †Department of Clinical Laboratory, Zhenhai Longsai Hospital, Ningbo, P.R. China
| | - Zhankun Zhu
- *Department of Clinical Laboratory, Ningbo No. 2 Hospital, Ningbo, P.R. China
| | - Yanqin Hu
- †Department of Clinical Laboratory, Zhenhai Longsai Hospital, Ningbo, P.R. China
| |
Collapse
|
14
|
Saponara E, Visentin M, Baschieri F, Seleznik G, Martinelli P, Esposito I, Buschmann J, Chen R, Parrotta R, Borgeaud N, Bombardo M, Malagola E, Caflisch A, Farhan H, Graf R, Sonda S. Serotonin uptake is required for Rac1 activation in Kras-induced acinar-to-ductal metaplasia in the pancreas. J Pathol 2018; 246:352-365. [PMID: 30058725 DOI: 10.1002/path.5147] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 07/03/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), which is the primary cause of pancreatic cancer mortality, is poorly responsive to currently available interventions. Identifying new targets that drive PDAC formation and progression is critical for developing alternative therapeutic strategies to treat this lethal malignancy. Using genetic and pharmacological approaches, we investigated in vivo and in vitro whether uptake of the monoamine serotonin [5-hydroxytryptamine (5-HT)] is required for PDAC development. We demonstrated that pancreatic acinar cells have the ability to readily take up 5-HT in a transport-mediated manner. 5-HT uptake promoted activation of the small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1), which is required for transdifferentiation of acinar cells into acinar-to-ductal metaplasia (ADM), a key determinant in PDAC development. Consistent with the central role played by Rac1 in ADM formation, inhibition of the 5-HT transporter Sert (Slc6a4) with fluoxetine reduced ADM formation both in vitro and in vivo in a cell-autonomous manner. In addition, fluoxetine treatment profoundly compromised the stromal reaction and affected the proliferation and lipid metabolism of malignant PDAC cells. We propose that Sert is a promising therapeutic target to counteract the early event of ADM, with the potential to stall the initiation and progression of pancreatic carcinogenesis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Enrica Saponara
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital of Zurich, Zurich, Switzerland
| | - Francesco Baschieri
- Institute Gustave Roussy, Institut National de la Santé et de la Recherche Médicale (INSERM), Villejuif, France
| | - Gitta Seleznik
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Paola Martinelli
- Institute for Cancer Research, Medical University, Wien, Austria
| | - Irene Esposito
- Institut für Pathologie, University Hospital of Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany
| | - Johanna Buschmann
- Division of Plastic and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Rong Chen
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Rossella Parrotta
- Laboratory of Molecular Oncology, Thorax und Lungen Tumor Zentrum, University Hospital of Zurich, Zurich, Switzerland
| | - Nathalie Borgeaud
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Marta Bombardo
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Ermanno Malagola
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Hesso Farhan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Rolf Graf
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland.,Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Sabrina Sonda
- Department of Visceral and Transplant Surgery, University Hospital of Zurich, Zurich, Switzerland.,Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Poudel KR, Roh-Johnson M, Su A, Ho T, Mathsyaraja H, Anderson S, Grady WM, Moens CB, Conacci-Sorrell M, Eisenman RN, Bai J. Competition between TIAM1 and Membranes Balances Endophilin A3 Activity in Cancer Metastasis. Dev Cell 2018; 45:738-752.e6. [PMID: 29920278 DOI: 10.1016/j.devcel.2018.05.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 04/10/2018] [Accepted: 05/17/2018] [Indexed: 11/19/2022]
Abstract
Normal cells acquire aggressive behavior by modifying signaling pathways. For instance, alteration of endocytosis profoundly impacts both proliferation and migration during tumorigenesis. Here we investigate the mechanisms that enable the endocytic machinery to coordinate these processes. We show that a membrane curvature-sensing protein, endophilin A3, promotes growth and migration of colon cancer cells through two competing mechanisms: an endocytosis pathway that is required for proliferation and a GTPase regulatory pathway that controls cell motility. EndoA3 stimulates cell migration by binding the Rac GEF TIAM1 leading to activation of small GTPases. Competing interactions of EndoA3 with membrane versus TIAM1 modulate hyperproliferative and metastatic phenotypes. Disruption of EndoA3-membrane interactions stimulates TIAM1 and small GTPases in vitro, and further promotes pro-metastatic phenotypes in vivo. Together, these results uncover a coupling mechanism, by which EndoA3 promotes growth and migration of colon cancers, by linking membrane dynamics to GTPase regulation.
Collapse
Affiliation(s)
- Kumud R Poudel
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Minna Roh-Johnson
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Allen Su
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Thuong Ho
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Haritha Mathsyaraja
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Sarah Anderson
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - William M Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Cecilia B Moens
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | | | - Robert N Eisenman
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA.
| | - Jihong Bai
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA.
| |
Collapse
|
16
|
Arnst JL, Hein AL, Taylor MA, Palermo NY, Contreras JI, Sonawane YA, Wahl AO, Ouellette MM, Natarajan A, Yan Y. Discovery and characterization of small molecule Rac1 inhibitors. Oncotarget 2018; 8:34586-34600. [PMID: 28410221 PMCID: PMC5470993 DOI: 10.18632/oncotarget.16656] [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: 08/20/2016] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
Aberrant activation of Rho GTPase Rac1 has been observed in various tumor types, including pancreatic cancer. Rac1 activates multiple signaling pathways that lead to uncontrolled proliferation, invasion and metastasis. Thus, inhibition of Rac1 activity is a viable therapeutic strategy for proliferative disorders such as cancer. Here we identified small molecule inhibitors that target the nucleotide-binding site of Rac1 through in silico screening. Follow up in vitro studies demonstrated that two compounds blocked active Rac1 from binding to its effector PAK1. Fluorescence polarization studies indicate that these compounds target the nucleotide-binding site of Rac1. In cells, both compounds blocked Rac1 binding to its effector PAK1 following EGF-induced Rac1 activation in a dose-dependent manner, while showing no inhibition of the closely related Cdc42 and RhoA activity. Furthermore, functional studies indicate that both compounds reduced cell proliferation and migration in a dose-dependent manner in multiple pancreatic cancer cell lines. Additionally, the two compounds suppressed the clonogenic survival of pancreatic cancer cells, while they had no effect on the survival of normal pancreatic ductal cells. These compounds do not share the core structure of the known Rac1 inhibitors and could serve as additional lead compounds to target pancreatic cancers with high Rac1 activity.
Collapse
Affiliation(s)
- Jamie L Arnst
- Department of Radiation Oncology, University of Nebraska Medical Center Omaha, Nebraska, United States of America.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Ashley L Hein
- Department of Radiation Oncology, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Margaret A Taylor
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Nick Y Palermo
- Holland Computing Center University of Nebraska-Lincoln Omaha, Nebraska, United States of America
| | - Jacob I Contreras
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Yogesh A Sonawane
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Andrew O Wahl
- Department of Radiation Oncology, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Michel M Ouellette
- Department of Internal Medicine, University of Nebraska Medical Center Omaha, Nebraska, United States of America.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center Omaha, Nebraska, United States of America.,Department of Pharmaceutical Sciences, University of Nebraska Medical Center Omaha, Nebraska, United States of America.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center Omaha, Nebraska, United States of America.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center Omaha, Nebraska, United States of America
| |
Collapse
|
17
|
Kou W, Xu X, Ji S, Chen M, Liu D, Wang K, Zhuang J, Yu Q, Zhao Q, Xu Y, Zhang H, Peng W. The inhibition of the effect and mechanism of vascular intimal hyperplasia in Tiam1 knockout mice. Biochem Biophys Res Commun 2018; 497:248-255. [DOI: 10.1016/j.bbrc.2018.02.065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/21/2022]
|
18
|
Tiam1 promotes thyroid carcinoma metastasis by modulating EMT via Wnt/β-catenin signaling. Exp Cell Res 2018; 362:532-540. [DOI: 10.1016/j.yexcr.2017.12.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/17/2022]
|
19
|
Melzer C, Hass R, von der Ohe J, Lehnert H, Ungefroren H. The role of TGF-β and its crosstalk with RAC1/RAC1b signaling in breast and pancreas carcinoma. Cell Commun Signal 2017; 15:19. [PMID: 28499439 PMCID: PMC5429551 DOI: 10.1186/s12964-017-0175-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022] Open
Abstract
This article focusses on the role of TGF-β and its signaling crosstalk with the RHO family GTPases RAC1 and RAC1b in the progression of breast and pancreatic carcinoma. The aggressive nature of these tumor types is mainly due to metastatic dissemination. Metastasis is facilitated by desmoplasia, a peculiar tumor microenvironment and the ability of the tumor cells to undergo epithelial-mesenchymal transition (EMT) and to adopt a motile and invasive phenotype. These processes are controlled entirely or in part by TGF-β and the small RHO GTPase RAC1 with both proteins acting as tumor promoters in late-stage cancers. Data from our and other studies point to signaling crosstalk between TGF-β and RAC1 and the related isoform, RAC1b, in pancreatic and mammary carcinoma cells. Based on the exciting observation that RAC1b functions as an endogenous inhibitor of RAC1, we propose a model on how the relative abundance or activity of RAC1 and RAC1b in the tumor cells may determine their responses to TGF-β and, ultimately, the metastatic capacity of the tumor.
Collapse
Affiliation(s)
- Catharina Melzer
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Hendrik Lehnert
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.,First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Hendrik Ungefroren
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany. .,First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany. .,Department of General and Thoracic Surgery, UKSH, Campus Kiel, Kiel, Germany.
| |
Collapse
|
20
|
Guo X, Zheng L, Jiang J, Zhao Y, Wang X, Shen M, Zhu F, Tian R, Shi C, Xu M, Li X, Peng F, Zhang H, Feng Y, Xie Y, Xu X, Jia W, He R, Xie C, Hu J, Ye D, Wang M, Qin R. Blocking NF-κB Is Essential for the Immunotherapeutic Effect of Recombinant IL18 in Pancreatic Cancer. Clin Cancer Res 2016; 22:5939-5950. [PMID: 27297583 DOI: 10.1158/1078-0432.ccr-15-1144] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 04/27/2016] [Accepted: 05/11/2016] [Indexed: 01/16/2023]
Abstract
PURPOSE We sought to find new immune-based treatments for pancreatic cancer. EXPERIMENTAL DESIGN We detected IL18 expression in plasma and specimens from patients with pancreatic cancer. We then investigated whether IL18 had a therapeutic effect for pancreatic cancer in vitro and in vivo and any underlying mechanisms. RESULTS Higher plasma IL18 was associated with longer overall survival (OS), but higher IL18 in pancreatic cancer tissues was associated with shorter OS and increased invasion and metastasis. Recombinant IL18 alone had no antitumor effect in the syngeneic mice with orthotopically transplanted tumors and promoted tumors in immunocompromised mice; it also facilitated immune responses in vitro and in vivo by augmenting the activity of cytotoxic T cells and NK cells in peripheral blood and lymph nodes. However, IL18 promoted the proliferation and invasion of pancreatic cancer cells, in vitro and in vivo, through the NF-κB pathway. Nevertheless, by coadministrating IL18 with BAY11-7082, an NF-κB inhibitor, we were able to prevent the procancerous effects of IL18 and prolong the survival time of the mice. CONCLUSIONS IL18 has both cancer-promoting and cancer-suppressing functions. Although its single-agent treatment has no therapeutic effect on pancreatic cancer, when combined with the NF-κB pathway inhibitor, IL18 improved survival in a murine pancreatic cancer model. Our study implies the possibility of a combinational immunotherapy that uses IL18 and targets NF-κB pathway. Clin Cancer Res; 22(23); 5939-50. ©2016 AACR.
Collapse
Affiliation(s)
- Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zheng
- Department of Oncology, The Sidney Kimmel Cancer Center, and the Skip Viragh Center for Pancreatic Cancer Research & Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Surgery, The Sidney Kimmel Cancer Center, and the Skip Viragh Center for Pancreatic Cancer Research & Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jianxin Jiang
- Department of hepatic-biliary-pancreatic surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Zhao
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Shen
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Zhu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Tian
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengjian Shi
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Xu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Li
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Peng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Zhang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yechen Feng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xie
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaodong Xu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Jia
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chencheng Xie
- Department of Bioengineering and Therapeutic Sciences, University of Minnesota, Minneapolis, Minnesota
| | - Jun Hu
- Department of Colon Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dawei Ye
- Department of Oncology, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Kurdi AT, Bassil R, Olah M, Wu C, Xiao S, Taga M, Frangieh M, Buttrick T, Orent W, Bradshaw EM, Khoury SJ, Elyaman W. Tiam1/Rac1 complex controls Il17a transcription and autoimmunity. Nat Commun 2016; 7:13048. [PMID: 27725632 PMCID: PMC5062600 DOI: 10.1038/ncomms13048] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022] Open
Abstract
RORγt is a master transcription factor of Th17 cells and considered as a promising drug target for the treatment of autoimmune diseases. Here, we show the guanine nucleotide exchange factor, Tiam1, and its cognate Rho-family G protein, Rac1, regulate interleukin (IL)17A transcription and autoimmunity. Whereas Tiam1 genetic deficiency weakens IL-17A expression partially and inhibits the development of experimental autoimmune encephalomyelitis (EAE), deletion of Rac1 in T cells exhibits more robust effects on Th17 cells and EAE. We demonstrate Tiam1 and Rac1 form a complex with RORγt in the nuclear compartment of Th17 cells, and together bind and activate the Il17 promoter. The clinical relevance of these findings is emphasized by pharmacological targeting of Rac1 that suppresses both murine and human Th17 cells as well as EAE. Thus, our findings highlight a regulatory pathway of Tiam1/Rac1 in Th17 cells and suggest that it may be a therapeutic target in multiple sclerosis. Tiam1 is a guanine nucleotide exchange factor for the Rho-family GTPase Rac1. Here, the authors show that nuclear Tiam1 and Rac1 bind to RORγt on the IL-17 promoter, activating its transcription, and that inhibiting Tiam1/Rac1 is beneficial in a mouse model of autoimmunity.
Collapse
Affiliation(s)
- Ahmed T Kurdi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ribal Bassil
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Marta Olah
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Chuan Wu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sheng Xiao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mariko Taga
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Michael Frangieh
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas Buttrick
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - William Orent
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Elizabeth M Bradshaw
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Samia J Khoury
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Abu Haidar Neuroscience Institute, American University of Beirut Medical Center, Riad El Solh, Beirut 1107 2020, Lebanon
| | - Wassim Elyaman
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| |
Collapse
|
22
|
Li Z, Liu Q, Piao J, Hua F, Wang J, Jin G, Lin Z, Zhang Y. Clinicopathological implications of Tiam1 overexpression in invasive ductal carcinoma of the breast. BMC Cancer 2016; 16:681. [PMID: 27562113 PMCID: PMC4997674 DOI: 10.1186/s12885-016-2724-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 08/04/2016] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND T-lymphoma invasion and metastasis-inducing protein 1 (Tiam1) has been implicated in tumor occurrence and progression. Recent studies have shown that high expression levels of Tiam1 protein appear to be associated with the progression of numerous human tumors. This study attempted to explore the role of Tiam1 protein in tumor progression and the prognostic evaluation of breast cancer. METHODS The localization of the Tiam1 protein was determined in the MDA-MB-231 breast cancer cell line using immunofluorescence (IF) staining. In addition, a total of 283 breast tissue samples, including 153 breast cancer tissues, 67 ductal carcinoma in situ (DCIS) and 63 adjacent non-tumor breast tissues, were analyzed by immunohistochemical (IHC) staining of the Tiam1 protein. The correlation between Tiam1 expression and clinicopathological characteristics was evaluated by Chi-square test and Fisher's exact tests. Disease-free survival (DFS) and 10-year overall survival (OS) rates were calculated by the Kaplan-Meier method. Additionally, univariate and multivariate analyses were performed by the Cox proportional hazards regression models. RESULTS Tiam1 protein showed a mainly cytoplasmic staining pattern in breast cancer cells; however, nuclear staining was also observed. Tiam1 protein expression was significantly higher in breast cancers (42.5 %, 65/153) and DCIS (40.3 %, 27/67) than in adjacent non-tumor tissues (12.7 %, 8/63). In addition, Tiam1 associated with tumor stage and Ki-67 expression, but negatively correlated with receptor tyrosine-protein kinase erbB-2 (Her2) expression. Moreover, survival analyses showed that DFS and 10-year OS rates were significantly lower in breast cancer patients with high Tiam1 expression than those with low Tiam1 expression. Univariate analysis suggested that molecular types, clinical stage, Her2 expression levels and Tiam1 expression levels were also significantly associated with DFS and 10-year OS rates of breast cancer patients. Furthermore, multivariate analysis suggested that Tiam1 expression is a significant independent prognostic factor along with tumor stage in patients with breast cancer. CONCLUSIONS Tiam1 expression is frequently up-regulated in breast cancer. Tiam1 expression correlated with clinicopathological parameters, suggesting that it may be a useful prognostic biomarker and potential therapeutic target for patients with breast cancer.
Collapse
Affiliation(s)
- Zhenling Li
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.,Department of Breast Surgery, the Second Hospital of Jilin University, Changchun, 130041, China
| | - Qixiang Liu
- Department of Breast Surgery, the Second Hospital of Jilin University, Changchun, 130041, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Fenjian Hua
- Department of Breast Surgery, the Second Hospital of Jilin University, Changchun, 130041, China
| | - Jing Wang
- Department of Breast Surgery, the Second Hospital of Jilin University, Changchun, 130041, China
| | - Guang Jin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Zhenhua Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| | - Yan Zhang
- Department of Breast Surgery, the Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
23
|
Inhibition of RAC1 GTPase sensitizes pancreatic cancer cells to γ-irradiation. Oncotarget 2015; 5:10251-70. [PMID: 25344910 PMCID: PMC4279370 DOI: 10.18632/oncotarget.2500] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/16/2014] [Indexed: 12/31/2022] Open
Abstract
Radiation therapy is a staple treatment for pancreatic cancer. However, owing to the intrinsic radioresistance of pancreatic cancer cells, radiation therapy often fails to increase survival of pancreatic cancer patients. Radiation impedes cancer cells by inducing DNA damage, which can activate cell cycle checkpoints. Normal cells possess both a G1 and G2 checkpoint. However, cancer cells are often defective in G1 checkpoint due to mutations/alterations in key regulators of this checkpoint. Accordingly, our results show that normal pancreatic ductal cells respond to ionizing radiation (IR) with activation of both checkpoints whereas pancreatic cancer cells respond to IR with G2/M arrest only. Overexpression/hyperactivation of Rac1 GTPase is detected in the majority of pancreatic cancers. Rac1 plays important roles in survival and Ras-mediated transformation. Here, we show that Rac1 also plays a critical role in the response of pancreatic cancer cells to IR. Inhibition of Rac1 using specific inhibitor and dominant negative Rac1 mutant not only abrogates IR-induced G2 checkpoint activation, but also increases radiosensitivity of pancreatic cancer cells through induction of apoptosis. These results implicate Rac1 signaling in the survival of pancreatic cancer cells following IR, raising the possibility that this pathway contributes to the intrinsic radioresistance of pancreatic cancer.
Collapse
|
24
|
Pajic M, Herrmann D, Vennin C, Conway JR, Chin VT, Johnsson AKE, Welch HC, Timpson P. The dynamics of Rho GTPase signaling and implications for targeting cancer and the tumor microenvironment. Small GTPases 2015; 6:123-33. [PMID: 26103062 PMCID: PMC4601362 DOI: 10.4161/21541248.2014.973749] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Numerous large scale genomics studies have demonstrated that cancer is a molecularly heterogeneous disease, characterized by acquired changes in the structure and DNA sequence of tumor genomes. More recently, the role of the equally complex tumor microenvironment in driving the aggressiveness of this disease is increasingly being realized. Tumor cells are surrounded by activated stroma, creating a dynamic environment that promotes cancer development, metastasis and chemoresistance. The Rho family of small GTPases plays an essential role in the regulation of cell shape, cytokinesis, cell adhesion, and cell motility. Importantly, these processes need to be considered in the context of a complex 3-dimensional (3D) environment, with reciprocal feedback and cross-talk taking place between the tumor cells and host environment. Here we discuss the role of molecular networks involving Rho GTPases in cancer, and the therapeutic implications of inhibiting Rho signaling in both cancer cells and the emerging concept of targeting the surrounding stroma.
Collapse
Affiliation(s)
- Marina Pajic
- a The Kinghorn Cancer Center; Cancer Division; Garvan Institute of Medical Research ; Sydney , Australia
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Guo X, Wang M, Zhao Y, Wang X, Shen M, Zhu F, Shi C, Xu M, Li X, Peng F, Zhang H, Feng Y, Xie Y, Xu X, Jia W, He R, Jiang J, Hu J, Tian R, Qin R. Par3 regulates invasion of pancreatic cancer cells via interaction with Tiam1. Clin Exp Med 2015; 16:357-65. [PMID: 26084985 DOI: 10.1007/s10238-015-0365-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/03/2015] [Indexed: 12/16/2022]
Abstract
The conserved polarity complex, which comprises partitioning-defective proteins Par3, Par6, and the atypical protein kinase C, affects various cell-polarization events, including assembly of tight junctions. Control of tight junction assembly is closely related to invasion and migration potential. However, as the importance of conserved polarity complexes in regulating pancreatic cancer invasion and metastasis is unclear, we investigated their role and mechanism in pancreatic cancers. We first detect that the key protein of the conserved polarity complex finds that only Par3 is down-regulated in pancreatic cancer tissues while Par6 and aPKC show no difference. What is more, Par3 tissues level was significantly and positively associated with patient overall survival. Knocking-down Par3 promotes pancreatic cancer cells invasion and migration. And Par3 requires interaction with Tiam1 to affect tight junction assembly, and then affect invasion and migration of pancreatic cancer cells. Then, we find that tight junction marker protein ZO-1 and claudin-1 are down-regulated in pancreatic cancer tissues. And the relationship of the expression of Par3 and ZO-1 in pancreatic cancer tissue is linear correlation. We establish liver metastasis model of human pancreatic cancer cells in Balb/c nude mice and find that knocking down Par3 promotes invasion and metastasis and disturbs tight junction assembly in vivo. Taken together, these results suggest that the Par3 regulates invasion and metastasis in pancreatic cancers by controlling tight junction assembly.
Collapse
Affiliation(s)
- Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Yan Zhao
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Xin Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Ming Shen
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Feng Zhu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Chengjian Shi
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Meng Xu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Xu Li
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Feng Peng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Hang Zhang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Yechen Feng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Yu Xie
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Xiaodong Xu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Wei Jia
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China
| | - Jianxin Jiang
- Department of Hepatic-Biliary-Pancreatic Surgery, Hubei Cancer Hospital, Wuhan City, People's Republic of China
| | - Jun Hu
- Department of Colon Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Rui Tian
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan City, 430030, Hubei Province, People's Republic of China.
| |
Collapse
|
26
|
Gonzalez-Villasana V, Fuentes-Mattei E, Ivan C, Dalton HJ, Rodriguez-Aguayo C, Fernandez-de Thomas RJ, Aslan B, Del C Monroig P, Velazquez-Torres G, Previs RA, Pradeep S, Kahraman N, Wang H, Kanlikilicer P, Ozpolat B, Calin G, Sood AK, Lopez-Berestein G. Rac1/Pak1/p38/MMP-2 Axis Regulates Angiogenesis in Ovarian Cancer. Clin Cancer Res 2015; 21:2127-37. [PMID: 25595279 DOI: 10.1158/1078-0432.ccr-14-2279] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/22/2014] [Indexed: 11/16/2022]
Abstract
PURPOSE Zoledronic acid is being increasingly recognized for its antitumor properties, but the underlying functions are not well understood. In this study, we hypothesized that zoledronic acid inhibits ovarian cancer angiogenesis preventing Rac1 activation. EXPERIMENTAL DESIGN The biologic effects of zoledronic acid were examined using a series of in vitro [cell invasion, cytokine production, Rac1 activation, reverse-phase protein array, and in vivo (orthotopic mouse models)] experiments. RESULTS There was significant inhibition of ovarian cancer (HeyA8-MDR and OVCAR-5) cell invasion as well as reduced production of proangiogenic cytokines in response to zoledronic acid treatment. Furthermore, zoledronic acid inactivated Rac1 and decreased the levels of Pak1/p38/matrix metalloproteinase-2 in ovarian cancer cells. In vivo, zoledronic acid reduced tumor growth, angiogenesis, and cell proliferation and inactivated Rac1 in both HeyA8-MDR and OVCAR-5 models. These in vivo antitumor effects were enhanced in both models when zoledronic acid was combined with nab-paclitaxel. CONCLUSIONS Zoledronic acid has robust antitumor and antiangiogenic activity and merits further clinical development as ovarian cancer treatment.
Collapse
Affiliation(s)
- Vianey Gonzalez-Villasana
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Enrique Fuentes-Mattei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristina Ivan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heather J Dalton
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paloma Del C Monroig
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Guermarie Velazquez-Torres
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rebecca A Previs
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunila Pradeep
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nermin Kahraman
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pinar Kanlikilicer
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNAi and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNAi and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNAi and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
27
|
Tang SC, Chen YC. Novel therapeutic targets for pancreatic cancer. World J Gastroenterol 2014; 20:10825-10844. [PMID: 25152585 PMCID: PMC4138462 DOI: 10.3748/wjg.v20.i31.10825] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/13/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer has become the fourth leading cause of cancer death in the last two decades. Only 3%-15% of patients diagnosed with pancreatic cancer had 5 year survival rate. Drug resistance, high metastasis, poor prognosis and tumour relapse contributed to the malignancies and difficulties in treating pancreatic cancer. The current standard chemotherapy for pancreatic cancer is gemcitabine, however its efficacy is far from satisfactory, one of the reasons is due to the complex tumour microenvironment which decreases effective drug delivery to target cancer cell. Studies of the molecular pathology of pancreatic cancer have revealed that activation of KRAS, overexpression of cyclooxygenase-2, inactivation of p16INK4A and loss of p53 activities occurred in pancreatic cancer. Co-administration of gemcitabine and targeting the molecular pathological events happened in pancreatic cancer has brought an enhanced therapeutic effectiveness of gemcitabine. Therefore, studies looking for novel targets in hindering pancreatic tumour growth are emerging rapidly. In order to give a better understanding of the current findings and to seek the direction in future pancreatic cancer research; in this review we will focus on targets suppressing tumour metastatsis and progression, KRAS activated downstream effectors, the relationship of Notch signaling and Nodal/Activin signaling with pancreatic cancer cells, the current findings of non-coding RNAs in inhibiting pancreatic cancer cell proliferation, brief discussion in transcription remodeling by epigenetic modifiers (e.g., HDAC, BMI1, EZH2) and the plausible therapeutic applications of cancer stem cell and hyaluronan in tumour environment.
Collapse
|
28
|
Hegde SM, Srivastava K, Tiwary E, Srivastava OP. Molecular mechanism of formation of cortical opacity in CRYAAN101D transgenic mice. Invest Ophthalmol Vis Sci 2014; 55:6398-408. [PMID: 25146988 DOI: 10.1167/iovs.14-14623] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE The CRYAAN101D transgenic mouse model expressing deamidated αA-crystallin (deamidation at N101 position to D) develops cortical cataract at the age of 7 to 9 months. The present study was carried out to explore the molecular mechanism that leads to the development of cortical opacity in CRYAAN101D lenses. METHODS RNA sequence analysis was carried out on 2- and 4-month-old αA-N101D and wild type (WT) lenses. To understand the biologic relevance and function of significantly altered genes, Ingenuity Pathway Analysis (IPA) was done. To elucidate terminal differentiation defects, immunohistochemical, and Western blot analyses were carried out. RESULTS RNA sequence and IPA data suggested that the genes belonging to gene expression, cellular assembly and organization, and cell cycle and apoptosis networks were altered in N101D lenses. In addition, the tight junction signaling and Rho A signaling were among the top three canonical pathways that were affected in N101D mutant. Immunohistochemical analysis identified a series of terminal differentiation defects in N101D lenses, specifically, increased proliferation and decreased differentiation of lens epithelial cells (LEC) and decreased denucleation of lens fiber cells (LFC). The expression of Rho A was reduced in different-aged N101D lenses, and, conversely, Cdc42 and Rac1 expressions were increased in the N101D mutants. Moreover, earlier in development, the expression of major membrane-bound molecular transporter Na,K-ATPase was drastically reduced in N101D lenses. CONCLUSIONS The results suggest that the terminal differentiation defects, specifically, increased proliferation and decreased denucleation are responsible for the development of lens opacity in N101D lenses.
Collapse
Affiliation(s)
- Shylaja M Hegde
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kiran Srivastava
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ekta Tiwary
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Om P Srivastava
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
29
|
Asad M, Wong MK, Tan TZ, Choolani M, Low J, Mori S, Virshup D, Thiery JP, Huang RYJ. FZD7 drives in vitro aggressiveness in Stem-A subtype of ovarian cancer via regulation of non-canonical Wnt/PCP pathway. Cell Death Dis 2014; 5:e1346. [PMID: 25032869 PMCID: PMC4123093 DOI: 10.1038/cddis.2014.302] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/27/2014] [Accepted: 06/09/2014] [Indexed: 01/02/2023]
Abstract
Ovarian cancer (OC) can be classified into five biologically distinct molecular subgroups: epithelial-A (Epi-A), Epi-B, mesenchymal (Mes), Stem-A and Stem-B. Among them, Stem-A expresses genes relating to stemness and is correlated with poor clinical prognosis. In this study, we show that frizzled family receptor 7 (FZD7), a receptor for Wnt signalling, is overexpressed in the Stem-A subgroup. To elucidate the functional roles of FZD7, we used an RNA interference gene knockdown approach in three Stem-A cell lines: CH1, PA1 and OV-17R. Si-FZD7 OC cells showed reduced cell proliferation with an increase in the G0/G1 sub-population, with no effect on apoptosis. The cells also displayed a distinctive morphologic change by colony compaction to become more epithelial-like and polarised with smaller internuclear distances and increased z-axis height. Immunofluorescence (IF) staining patterns of pan-cadherin and β-catenin suggested an increase in cadherin-based cell–cell adhesion in si-FZD7 cells. We also observed a significant rearrangement in the actin cytoskeleton and an increase in tensile contractility in si-FZD7 OC cells, as evident by the loss of stress fibres and the redistribution of phospho-myosin light chain (pMLC) from the sites of cell–cell contacts to the periphery of cell colonies. Furthermore, there was reciprocal regulation of RhoA (Ras homolog family member A) and Rac1 (Ras-related C3 botulinum toxin substrate 1 (Rho family, small GTP-binding protein Rac1)) activities upon FZD7 knockdown, with a significant reduction in RhoA activity and a concomitant upregulation in Rac1 activity. These changes in pMLC and RhoA, as well as the increased TopFlash reporter activities in si-FZD7 cells, suggested involvement of the non-canonical Wnt/planar cell polarity (PCP) pathway. Selected PCP pathway genes (cadherin EGF LAG seven-pass G-type receptor 3 (CELSR3), prickle homolog 4 (Drosophila) (PRICKLE4), dishevelled-associated activator of morphogenesis 1 (DAAM1), profilin 2 (PFN2), protocadherin 9 (PCDH9), protocadherin α1 (PCDHA1), protocadherin β17 pseudogene (PCDHB17), protocadherin β3 (PCDHB3), sprouty homolog 1 (SPRY1) and protein tyrosine kinase 7 (PTK7)) were found to be more highly expressed in Stem-A than non Stem-A subgroup of OC. Taken together, our results suggest that FZD7 might drive aggressiveness in Stem-A OC by regulating cell proliferation, cell cycle progression, maintenance of the Mes phenotype and cell migration via casein kinase 1ɛ-mediated non-canonical Wnt/PCP pathway.
Collapse
Affiliation(s)
- M Asad
- Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore
| | - M K Wong
- Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore
| | - T Z Tan
- Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore
| | - M Choolani
- Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore, Singapore
| | - J Low
- 1] Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore, Singapore [2] National University Cancer Institute of Singapore, Singapore, Singapore
| | - S Mori
- Division of Cancer Genomics, Cancer Institute of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, Japan
| | - D Virshup
- Duke NUS Graduate Medical School, Singapore, Singapore
| | - J P Thiery
- 1] Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore [2] National University Cancer Institute of Singapore, Singapore, Singapore [3] Department of Biochemistry, National University of Singapore, Singapore, Singapore [4] Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - R Y-J Huang
- 1] Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore [2] Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore, Singapore [3] National University Cancer Institute of Singapore, Singapore, Singapore
| |
Collapse
|
30
|
Hofbauer SW, Krenn PW, Ganghammer S, Asslaber D, Pichler U, Oberascher K, Henschler R, Wallner M, Kerschbaum H, Greil R, Hartmann TN. Tiam1/Rac1 signals contribute to the proliferation and chemoresistance, but not motility, of chronic lymphocytic leukemia cells. Blood 2014; 123:2181-8. [PMID: 24501217 DOI: 10.1182/blood-2013-08-523563] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Signals from the tumor microenvironment promote the migration, survival, and proliferation of chronic lymphocytic leukemia (CLL) cells. Rho GTPases control various signaling pathways downstream of microenvironmental cues. Here, we analyze the function of Rac1 in the motility and proliferation of CLL cells. We found decreased transcription of the Rac guanine nucleotide exchange factors Tiam1 and Vav1 in unstimulated peripheral blood CLL cells with almost complete loss of Tiam1 but increased transcription of the potential Rac antagonist RhoH. Consistently, stimulation of CLL cells with the chemokine CXCL12 induced RhoA but not Rac1 activation, whereas chemokine-induced CLL cell motility was Rac1-independent. Coculture of CLL cells with activated T cells induced their activation and subsequent proliferation. Here, Tiam1 expression was induced in the malignant cells in line with increased Ki-67 and c-Myc expression. Rac1 or Tiam1 knockdown using siRNA or treatment with the Tiam1/Rac inhibitor NSC-23766 attenuated c-Myc transcription. Furthermore, treatment of CLL cells with NSC-23766 reduced their proliferation. Rac inhibition also antagonized the chemoresistance of activated CLL cells toward fludarabine. Collectively, our data suggest a dynamic regulation of Rac1 function in the CLL microenvironment. Rac inhibition could be of clinical use by selectively interfering with CLL cell proliferation and chemoresistance.
Collapse
MESH Headings
- Aminoquinolines/pharmacology
- Animals
- Antineoplastic Agents/pharmacology
- Cell Movement/genetics
- Cell Proliferation
- Cells, Cultured
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Guanine Nucleotide Exchange Factors/antagonists & inhibitors
- Guanine Nucleotide Exchange Factors/physiology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- NIH 3T3 Cells
- Pyrimidines/pharmacology
- RNA, Small Interfering/genetics
- Signal Transduction/physiology
- T-Lymphoma Invasion and Metastasis-inducing Protein 1
- rac1 GTP-Binding Protein/antagonists & inhibitors
- rac1 GTP-Binding Protein/physiology
Collapse
Affiliation(s)
- Sebastian W Hofbauer
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department with Hematology, Oncology, Hemostaseology, Infectiology, and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
p104 binds to Rac1 and reduces its activity during myotube differentiation of C2C12 cell. ScientificWorldJournal 2014; 2014:592450. [PMID: 24600331 PMCID: PMC3926281 DOI: 10.1155/2014/592450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/12/2013] [Indexed: 11/17/2022] Open
Abstract
The p104 protein inhibits cellular proliferation when overexpressed in NIH3T3 cells and has been shown to associate with p85α, Grb2, and PLCγ1. In order to isolate other proteins that interact with p104, yeast two-hybrid screening was performed. Rac1 was identified as a binding partner of p104 and the interaction between p104 and Rac1 was confirmed by immunoprecipitation. Using a glutathione S-transferase (GST) pull-down assay with various p104 fragments, the 814–848 amino acid residue at the carboxyl-terminal region of p104 was identified as the key component to interact with Rac1. The CrkII which is involved in the Rac1-mediated cellular response was also found to interact with p104 protein. NIH3T3 cells which overexpressed p104 showed a decrease of Rac1 activity. However, neither the proline-rich domain mutant, which is unable to interact with CrkII, nor the carboxy-terminal deletion mutant could attenuate Rac1 activity. During the differentiation of myoblasts, the amount of p104 protein as well as transcript level was increased. The overexpression of p104 enhanced myotube differentiation, whereas siRNA of p104 reversed this process. In this process, more Rac1 and CrkII were bound to increased p104. Based on these results, we conclude that p104 is involved in muscle cell differentiation by modulating the Rac1 activity.
Collapse
|
32
|
The guanine nucleotide exchange factor Tiam1: A Janus-faced molecule in cellular signaling. Cell Signal 2014; 26:483-91. [DOI: 10.1016/j.cellsig.2013.11.034] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 11/22/2022]
|
33
|
Exploring the Wnt pathway-associated LncRNAs and genes involved in pancreatic carcinogenesis driven by Tp53 mutation. Pharm Res 2014; 32:793-805. [PMID: 24469904 DOI: 10.1007/s11095-013-1269-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/19/2013] [Indexed: 01/05/2023]
Abstract
PURPOSE Study the contribution of long non-coding RNAs (lncRNAs) to progression of pancreatic intraepithelial neoplasia (PanIN) to pancreatic ductal adenocarcinoma (PDAC). METHODS We explored lncRNAs profilings in PanIN cell line (SH-PAN) isolated from Pdx-1-Cre; LSL-Kras (G12D/+) mice and PDAC cell line (DT-PCa) isolated from Pdx-1-Cre; LSL- Kras (G12D/+) ; LSL- Tp53 (R172H/+) mice by lncRNAs microarray, and detected expression of lncRNAs and genes in PDAC by Real-time PCR, Western blot, ChIP and immunohistochemistry. RESULTS Eight lncRNAs and five protein-coding genes, associated with Wnt pathway, were identified with more than five-fold changes between DT-PCa cells and SH-PAN cells. Of them, lincRNA1611 and Ppp3ca were validated significantly high expression in DT-PCa cells and in 22 of 26 fresh resected human PDAC tissues, compared to SH-PAN cells and normal pancreatic tissues, respectively. Moreover, Tp53 mutation status displayed a positive correlation with lincRNA1611 or Ppp3ca level. Immunohistochemical staining for Ppp3ca was weak or lack in 91 of 107 normal pancreatic tissues, 24 of 29 PanIN-I and 13 of 16 PanIN-II tissues, however, was strong in 10 of 27 PanIN-III and 62 of 107 PDAC tissues post operation. CONCLUSIONS LincRNA1611 and Ppp3ca were high expression in PDAC and may serve as new potential targets for intervention of the disease.
Collapse
|