51
|
Zhang Y, Liao Y, Chen C, Sun W, Sun X, Liu Y, Xu E, Lai M, Zhang H. p38-regulated FOXC1 stability is required for colorectal cancer metastasis. J Pathol 2019; 250:217-230. [PMID: 31650548 DOI: 10.1002/path.5362] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/17/2019] [Accepted: 10/21/2019] [Indexed: 12/24/2022]
Abstract
Aberrant expression of forkhead box C1 (FOXC1) promotes tumor metastasis in multiple human malignant tumors. However, the upstream modulating mode and downstream molecular mechanism of FOXC1 in metastasis of colorectal cancer (CRC) remain unclear. Herein we describe a systematic analysis of FOXC1 expression and prognosis in CRC performed on our clinical data and public databases, which indicated that FOXC1 upregulation in CRC samples was significantly associated with poor prognosis. FOXC1 knockdown inhibited migration and invasion, whereas FOXC1 overexpression caused the opposite phenotype in vitro and in vivo. Furthermore, MMP10, SOX4 and SOX13 were verified as the target genes of FOXC1 for promoting CRC metastasis. MMP10 was demonstrated as the direct target and mediator of FOXC1. Interestingly, Ser241 and Ser272 of FOXC1 were identified as the key sites to interact with p38 and phosphorylation, which were critically required for maintaining the stability of FOXC1 protein. Moreover, FOXC1 was dephosphorylated by protein phosphatase 2A and phosphorylated by p38, which maintained FOXC1 protein stability through inhibiting ubiquitination. Expression of p38 was correlated with FOXC1 and MMP10 expression, indirectly indicating that FOXC1 was regulated by p38 MAPK. Therefore, FOXC1 is strongly suggested as a pro-metastatic gene in CRC by transcriptionally activating MMP10, SOX4 and SOX13; p38 interacts with and phosphorylates the Ser241 and ser272 sites of FOXC1 to maintain its stability by inhibiting ubiquitination and degradation. In conclusion, the protein stability of FOXC1 mediated by p38 contributes to the metastatic effect in CRC. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China
| | - Yan Liao
- Department of Pharmacology, China Pharmaceutical University, Nanjing, PR China
| | - Chaoyi Chen
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China
| | - Wenjie Sun
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China
| | - Xiaohui Sun
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Zhejiang, PR China
| | - Yuan Liu
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China
| | - Enping Xu
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China
| | - Maode Lai
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China.,Department of Pharmacology, China Pharmaceutical University, Nanjing, PR China
| | - Honghe Zhang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Intelligence Classification of Tumor Pathology and Precision Therapy Research Unit of Chinese Academy of Medical Sciences (2019RU042), Zhejiang University School of Medicine, Zhejiang, PR China
| |
Collapse
|
52
|
Zhao JF, Zha ZA, Xie WH, Wang HB, Li XM, Sun Q, Sun ML. [Effect of long chain non-coding RNA H19 on the migration and invasion of oral cancer cells and its molecular mechanism]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:378-383. [PMID: 31512829 DOI: 10.7518/hxkq.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the effect of the long chain non-coding RNA H19 (lncRNA H19) on the invasion and migration of oral cancer cells and its related molecular mechanism. METHODS The expression levels of lncRNA H19, miR-107, and cyclin-dependent kinase 6 (CDK6) in the immortalized oral epithelial cell line HIOEC and the oral cancer cell line CAL27 were detected by real-time quantitative polymerase chain reaction. CAL27 cells were transfected with siRNA H19, miR-107 mimics, pcDNA H19, or anti-miR-107, and the effects of H19 and miR-107 on the invasion and migration of cells were examined via Transwell assay. The TargetScan database predicted the targeting of H19, miR-107, and CDK6. Double luciferase reporter gene assay was performed to detect interactions among H19, miR-107, and CDK6. Western blot analysis was conducted to examine the effects of H19 and miR-107 on the protein level of the target gene CDK6. RESULTS Compared with that in HIOEC cells, the expression of H19 was significantly increased in CAL27 cells (P<0.05). After transfection with siRNA H19, the expression of H19 decreased, and the invasion and migration ability of CAL27 cells were inhibited (P<0.05). H19 could bind specifically to the 3'-UTR of miR-107 to modulate the expression of miR-107. Compared with that in HIOEC cells, the expression of miR-107 significantly decreased in CAL27 cells (P<0.05). The expression of miR-107 increased after transfection with siRNA H19, and anti-mir-107 co-transfection could promote the invasion and migration ability of siRNA H19 in CAL27 cells (P<0.05). Compared with that in HIOEC cells, CDK6 expression significantly increased in CAL27 cells (P<0.05), and the expression level of the gene was coregulated by H19 and miR-107 (P<0.05). CONCLUSIONS lncRNA H19 plays an important role in the development of oral cancer. It can regulate the invasion and migration of oral cancer cells by targeting the miR-107/CDK6 signaling axis.
Collapse
Affiliation(s)
- Jun-Fang Zhao
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhi-An Zha
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei-Hong Xie
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hai-Bin Wang
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin-Ming Li
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qiang Sun
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ming-Lei Sun
- Dept. of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
53
|
Expression and purification of the recombinant full-length retinoblastoma protein and characterisation of its interaction with the oncoprotein HDM2. Protein Expr Purif 2019; 162:62-66. [DOI: 10.1016/j.pep.2019.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/29/2019] [Indexed: 01/11/2023]
|
54
|
Liu K, Zhang C, Li B, Xie W, Zhang J, Nie X, Tan P, Zheng L, Wu S, Qin Y, Cui J, Zhi F. Mutual Stabilization between TRIM9 Short Isoform and MKK6 Potentiates p38 Signaling to Synergistically Suppress Glioblastoma Progression. Cell Rep 2019; 23:838-851. [PMID: 29669288 DOI: 10.1016/j.celrep.2018.03.096] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 03/06/2018] [Accepted: 03/20/2018] [Indexed: 02/02/2023] Open
Abstract
p38 signaling is broadly involved in controlling inflammation and stress-induced cell death; however, the mechanisms controlling its activity have seldom been studied. Here, we report that TRIM9 short isoform (TRIM9s) potentiates p38 signaling by stabilizing MKK6. Mechanistic studies revealed that TRIM9s promotes the K63-linked ubiquitination of MKK6 at Lys82, thus inhibiting the degradative K48-linked ubiquitination of MKK6 at the same lysine. MKK6 could also stabilize TRIM9s by promoting the phosphorylation of TRIM9s at Ser76/80 via p38, thereby blocking the ubiquitin-proteasome pathway. Further functional analyses showed that p38 signaling plays a critical role in suppressing glioblastoma progression. Co-reduction of MKK6 and TRIM9s is significantly associated with overall poor survival of glioblastoma patients. We identify a positive feedback loop in p38 signaling generated by MKK6-TRIM9s, which suppresses glioblastoma progression, and we provide insights into the mechanisms by which TRIM9s and MKK6 potentiate p38 signaling through mutual stabilization.
Collapse
Affiliation(s)
- Kunpeng Liu
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Chuanxia Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Bowen Li
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213000, China; Modern Medical Research Center, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, China
| | - Weihong Xie
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Jindong Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Xichen Nie
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Peng Tan
- Institute of Biosciences and Technology, Texas A&M University, Health Science Center, Houston, TX 77030, USA
| | - Limin Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Song Wu
- Department of Urology Institute of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen 518000, China.
| | - Yunfei Qin
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Liver Disease, Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China.
| | - Jun Cui
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China; Department of Urology Institute of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen 518000, China.
| | - Feng Zhi
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213000, China; Modern Medical Research Center, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, China.
| |
Collapse
|
55
|
Targeting DNA Replication Stress and DNA Double-Strand Break Repair for Optimizing SCLC Treatment. Cancers (Basel) 2019; 11:cancers11091289. [PMID: 31480716 PMCID: PMC6770306 DOI: 10.3390/cancers11091289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Small cell lung cancer (SCLC), accounting for about 15% of all cases of lung cancer worldwide, is the most lethal form of lung cancer. Despite an initially high response rate of SCLC to standard treatment, almost all patients are invariably relapsed within one year. Effective therapeutic strategies are urgently needed to improve clinical outcomes. Replication stress is a hallmark of SCLC due to several intrinsic factors. As a consequence, constitutive activation of the replication stress response (RSR) pathway and DNA damage repair system is involved in counteracting this genotoxic stress. Therefore, therapeutic targeting of such RSR and DNA damage repair pathways will be likely to kill SCLC cells preferentially and may be exploited in improving chemotherapeutic efficiency through interfering with DNA replication to exert their functions. Here, we summarize potentially valuable targets involved in the RSR and DNA damage repair pathways, rationales for targeting them in SCLC treatment and ongoing clinical trials, as well as possible predictive biomarkers for patient selection in the management of SCLC.
Collapse
|
56
|
Kim SJ, Asfaha S, Dick FA. CDK4 Inhibitors Thwart Immunity by Inhibiting Phospho-RB-NF-κB Complexes. Mol Cell 2019; 73:1-2. [PMID: 30609388 DOI: 10.1016/j.molcel.2018.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PD-L1 plays a central role in immune recognition of cancer cells. In this issue of Molecular Cell, Jin et al. (2019) report that a phosphorylated retinoblastoma protein contacts the DNA-binding domain of p65 NF-κB, thereby blocking transcription of PD-L1.
Collapse
Affiliation(s)
- Seung J Kim
- London Regional Cancer Program, London, ON, Canada; Children's Health Research Institute, London, ON, Canada; Department of Biochemistry, Western University, London, ON, Canada
| | - Samuel Asfaha
- London Regional Cancer Program, London, ON, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada; Department of Medicine, Western University, London, ON, Canada
| | - Frederick A Dick
- London Regional Cancer Program, London, ON, Canada; Children's Health Research Institute, London, ON, Canada; Department of Biochemistry, Western University, London, ON, Canada.
| |
Collapse
|
57
|
Carbonell C, Ulsamer A, Vivori C, Papasaikas P, Böttcher R, Joaquin M, Miñana B, Tejedor JR, de Nadal E, Valcárcel J, Posas F. Functional Network Analysis Reveals the Relevance of SKIIP in the Regulation of Alternative Splicing by p38 SAPK. Cell Rep 2019; 27:847-859.e6. [PMID: 30995481 PMCID: PMC6484779 DOI: 10.1016/j.celrep.2019.03.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/21/2019] [Accepted: 03/15/2019] [Indexed: 01/03/2023] Open
Abstract
Alternative splicing is a prevalent mechanism of gene regulation that is modulated in response to a wide range of extracellular stimuli. Stress-activated protein kinases (SAPKs) play a key role in controlling several steps of mRNA biogenesis. Here, we show that osmostress has an impact on the regulation of alternative splicing (AS), which is partly mediated through the action of p38 SAPK. Splicing network analysis revealed a functional connection between p38 and the spliceosome component SKIIP, whose depletion abolished a significant fraction of p38-mediated AS changes. Importantly, p38 interacted with and directly phosphorylated SKIIP, thereby altering its activity. SKIIP phosphorylation regulated AS of GADD45α, the upstream activator of the p38 pathway, uncovering a negative feedback loop involving AS regulation. Our data reveal mechanisms and targets of SAPK function in stress adaptation through the regulation of AS.
Collapse
Affiliation(s)
- Caterina Carbonell
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Arnau Ulsamer
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Claudia Vivori
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Panagiotis Papasaikas
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - René Böttcher
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Manel Joaquin
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Belén Miñana
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Juan Ramón Tejedor
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Eulàlia de Nadal
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain.
| | - Juan Valcárcel
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluis Companys 23, 08010 Barcelona, Spain.
| | - Francesc Posas
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain.
| |
Collapse
|
58
|
p38γ is essential for cell cycle progression and liver tumorigenesis. Nature 2019; 568:557-560. [PMID: 30971822 DOI: 10.1038/s41586-019-1112-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/07/2019] [Indexed: 11/08/2022]
Abstract
The cell cycle is a tightly regulated process that is controlled by the conserved cyclin-dependent kinase (CDK)-cyclin protein complex1. However, control of the G0-to-G1 transition is not completely understood. Here we demonstrate that p38 MAPK gamma (p38γ) acts as a CDK-like kinase and thus cooperates with CDKs, regulating entry into the cell cycle. p38γ shares high sequence homology, inhibition sensitivity and substrate specificity with CDK family members. In mouse hepatocytes, p38γ induces proliferation after partial hepatectomy by promoting the phosphorylation of retinoblastoma tumour suppressor protein at known CDK target residues. Lack of p38γ or treatment with the p38γ inhibitor pirfenidone protects against the chemically induced formation of liver tumours. Furthermore, biopsies of human hepatocellular carcinoma show high expression of p38γ, suggesting that p38γ could be a therapeutic target in the treatment of this disease.
Collapse
|
59
|
Barley β-glucan accelerates wound healing by favoring migration versus proliferation of human dermal fibroblasts. Carbohydr Polym 2019; 210:389-398. [DOI: 10.1016/j.carbpol.2019.01.090] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 02/05/2023]
|
60
|
Sanidas I, Morris R, Fella KA, Rumde PH, Boukhali M, Tai EC, Ting DT, Lawrence MS, Haas W, Dyson NJ. A Code of Mono-phosphorylation Modulates the Function of RB. Mol Cell 2019; 73:985-1000.e6. [PMID: 30711375 DOI: 10.1016/j.molcel.2019.01.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/26/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022]
Abstract
Hyper-phosphorylation of RB controls its interaction with E2F and inhibits its tumor suppressor properties. However, during G1 active RB can be mono-phosphorylated on any one of 14 CDK phosphorylation sites. Here, we used quantitative proteomics to profile protein complexes formed by each mono-phosphorylated RB isoform (mP-RB) and identified the associated transcriptional outputs. The results show that the 14 sites of mono-phosphorylation co-ordinate RB's interactions and confer functional specificity. All 14 mP-RBs interact with E2F/DP proteins, but they provide different shades of E2F regulation. RB mono-phosphorylation at S811, for example, alters RB transcriptional activity by promoting its association with NuRD complexes. The greatest functional differences between mP-RBs are evident beyond the cell cycle machinery. RB mono-phosphorylation at S811 or T826 stimulates the expression of oxidative phosphorylation genes, increasing cellular oxygen consumption. These results indicate that RB activation signals are integrated in a phosphorylation code that determines the diversity of RB activity.
Collapse
Affiliation(s)
- Ioannis Sanidas
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Robert Morris
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Katerina A Fella
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Purva H Rumde
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Myriam Boukhali
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Eric C Tai
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - David T Ting
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Michael S Lawrence
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA; Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA 02142, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA
| | - Nicholas J Dyson
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Building 149 13th Street, Charlestown, MA 02129, USA.
| |
Collapse
|
61
|
Phosphorylated RB Promotes Cancer Immunity by Inhibiting NF-κB Activation and PD-L1 Expression. Mol Cell 2018; 73:22-35.e6. [PMID: 30527665 DOI: 10.1016/j.molcel.2018.10.034] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/29/2018] [Accepted: 10/19/2018] [Indexed: 12/22/2022]
Abstract
Aberrant expression of programmed death ligand-1 (PD-L1) in tumor cells promotes cancer progression by suppressing cancer immunity. The retinoblastoma protein RB is a tumor suppressor known to regulate the cell cycle, DNA damage response, and differentiation. Here, we demonstrate that RB interacts with nuclear factor κB (NF-κB) protein p65 and that their interaction is primarily dependent on CDK4/6-mediated serine-249/threonine-252 (S249/T252) phosphorylation of RB. RNA-seq analysis shows a subset of NF-κB pathway genes including PD-L1 are selectively upregulated by RB knockdown or CDK4/6 inhibitor. S249/T252-phosphorylated RB inversely correlates with PD-L1 expression in patient samples. Expression of a RB-derived S249/T252 phosphorylation-mimetic peptide suppresses radiotherapy-induced upregulation of PD-L1 and augments therapeutic efficacy of radiation in vivo. Our findings reveal a previously unrecognized tumor suppressor function of hyperphosphorylated RB in suppressing NF-κB activity and PD-L1 expression and suggest that the RB-NF-κB axis can be exploited to overcome cancer immune evasion triggered by conventional or targeted therapies.
Collapse
|
62
|
Chen X, Liu C, Zhao R, Zhao P, Wu J, Zhou N, Ying M. Synergetic and Antagonistic Molecular Effects Mediated by the Feedback Loop of p53 and JNK between Saikosaponin D and SP600125 on Lung Cancer A549 Cells. Mol Pharm 2018; 15:4974-4984. [PMID: 30207732 DOI: 10.1021/acs.molpharmaceut.8b00595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We jointly analyzed the changes in cell cycle arrest and distribution, the accumulation of subphase cells, apoptosis, and proliferation in A549 cells treated with Saikosaponin D (Ssd) and JNK inhibitor SP600125 alone or in combination. Our results indicated that cell cycle arrest at G0/G1, S, and G2/M phases was coupled with the accumulation of subG1, subS, and subG2 cells, corresponding to early apoptosis, DNA endoreplication, and later inhibitory proliferation, respectively. Analyzing the expression of 18 cell cycle regulatory genes and JNK and phosphorylated JNK (pJNK) levels revealed an enhancement in these factors by Ssd. Additional SP600125 weakened or eliminated the Ssd-induced increase of these factors except that p53/p21 and Rassfia levels were further improved. Ingenuity Pathway Analysis (IPA) of the interactions of these factors revealed a negative synergistic effect on apoptosis while a positive synergistic effect on proliferative inhibition of the two drugs: (1) Ssd induced apoptosis via the activation of two axes, TGFα-JNK-p53 and TGFα-Rassfia-Mst1. By eliminating the Ssd-induced increase of JNK/pJNK, additional SP600125 weakened the Ssd-induced apoptotic axis of TGFα-JNK-p53 and simultaneously abolished Ssd-induced apoptosis; (2) Ssd inhibited proliferation by the activation of two axes, TGFβ-p53/p21/p27/p15/p16 and TGFα-Rassfia-cyclin D1. By improving the Ssd-induced increase of p53/p21 and Rassfia, additional SP600125 enhanced the two axes of Ssd-induced inhibitory proliferation. Analyzing JNK/pJNK, p53, phospho-p53, and TNF-α levels revealed an opposite association of JNK/pJNK with p53 while consistent with phospho-p53 and TNF-α, which supported the proposals that JNK/pJNK negatively regulated p53 level, while it mediated p53 phosphorylation to transcriptionally activate TNF-α expression of apoptotic gene and trigger apoptosis. With the multiple roles, JNK/pJNK forms a synergetic and antagonistic feedback loop with phospho-p53/p53. Within the feedback loop, (1) Ssd-induced apoptosis depended on JNK/pJNK activities mediating phospho-p53 that activated TNF-α expression; (2) by weakening the negative regulation of JNK/pJNK in p53, SP600125 enhanced p53 level and the Ssd-induced inhibitory proliferation axes of TGFβ-p53/p21/p27/p15/p16. The results indicated the central coordinating roles of the feedback loop in the synergistic and antagonistic effects of the two drugs in A549 cells and provided a rationale for the combination of Ssd with SP600125 in the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiaoman Chen
- Department of Molecular Biology and Biochemistry , Basic Medical College of Nanchang University , Nanchang , P. R. China
| | - Chenglin Liu
- Department of Molecular Biology and Biochemistry , Basic Medical College of Nanchang University , Nanchang , P. R. China
| | - Ruilin Zhao
- Department of Molecular Biology and Biochemistry , Basic Medical College of Nanchang University , Nanchang , P. R. China
| | - Ping Zhao
- Department of Molecular Biology and Biochemistry , Basic Medical College of Nanchang University , Nanchang , P. R. China
| | - Ju Wu
- Department of Molecular Biology and Biochemistry , Basic Medical College of Nanchang University , Nanchang , P. R. China
| | - Nanjin Zhou
- Institute of Molecular Medicine , Jiangxi Academy of Medical Sciences , Nanchang , P. R. China
| | - Muying Ying
- Department of Molecular Biology and Biochemistry , Basic Medical College of Nanchang University , Nanchang , P. R. China.,Institute of Molecular Medicine , Jiangxi Academy of Medical Sciences , Nanchang , P. R. China
| |
Collapse
|
63
|
Jiang X, Liu Y, Ma L, Ji R, Qu Y, Xin Y, Lv G. Chemopreventive activity of sulforaphane. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2905-2913. [PMID: 30254420 PMCID: PMC6141106 DOI: 10.2147/dddt.s100534] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancer is one of the major causes of morbidity and mortality in the world. Carcinogenesis is a multistep process induced by genetic and epigenetic changes that disrupt pathways controlling cell proliferation, apoptosis, differentiation, and senescence. In this context, many bioactive dietary compounds from vegetables and fruits have been demonstrated to be effective in cancer prevention and intervention. Over the years, sulforaphane (SFN), found in cruciferous vegetables, has been shown to have chemopreventive activity in vitro and in vivo. SFN protects cells from environmental carcinogens and also induces growth arrest and/or apoptosis in various cancer cells. In this review, we will discuss several potential mechanisms of the chemopreventive activity of SFN, including regulation of Phase I and Phase II drug-metabolizing enzymes, cell cycle arrest, and induction of apoptosis, especially via regulation of signaling pathways such as Nrf2-Keap1 and NF-κB. Recent studies suggest that SFN can also affect the epigenetic control of key genes and greatly influence the initiation and progression of cancer. This research may provide a basis for the clinical use of SFN for cancer chemoprevention and enable us to design preventive strategies for cancer management, reduce cancer development and recurrence, and thus improve patient survival.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Ye Liu
- Department of Pathobiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, China
| | - Lixin Ma
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Rui Ji
- Department of Internal Medicine, Florida Hospital, Orlando, FL, USA
| | - Yaqin Qu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China,
| | - Guoyue Lv
- Department of General Surgery, The First Hospital of Jilin University, Changchun 130021, China,
| |
Collapse
|
64
|
Abstract
The canonical model of RB-mediated tumour suppression developed over the past 30 years is based on the regulation of E2F transcription factors to restrict cell cycle progression. Several additional functions have been proposed for RB, on the basis of which a non-canonical RB pathway can be described. Mechanistically, the non-canonical RB pathway promotes histone modification and regulates chromosome structure in a manner distinct from cell cycle regulation. These functions have implications for chemotherapy response and resistance to targeted anticancer agents. This Opinion offers a framework to guide future studies of RB in basic and clinical research.
Collapse
Affiliation(s)
- Frederick A Dick
- London Regional Cancer Program, Children's Health Research Institute, Western University, London, Ontario, Canada.
- London Regional Cancer Program, Department of Biochemistry, Western University, London, Ontario, Canada.
| | - David W Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Julien Sage
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, USA
| | - Nicholas J Dyson
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
65
|
Miller RE, Uwamahoro N, Park JH. PPM1B depletion in U2OS cells supresses cell growth through RB1-E2F1 pathway and stimulates bleomycin-induced cell death. Biochem Biophys Res Commun 2018; 500:391-397. [DOI: 10.1016/j.bbrc.2018.04.084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/28/2022]
|
66
|
Yu-Lee LY, Yu G, Lee YC, Lin SC, Pan J, Pan T, Yu KJ, Liu B, Creighton CJ, Rodriguez-Canales J, Villalobos PA, Wistuba II, de Nadal E, Posas F, Gallick GE, Lin SH. Osteoblast-Secreted Factors Mediate Dormancy of Metastatic Prostate Cancer in the Bone via Activation of the TGFβRIII-p38MAPK-pS249/T252RB Pathway. Cancer Res 2018. [PMID: 29514796 DOI: 10.1158/0008-5472.can-17-1051] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bone metastasis from prostate cancer can occur years after prostatectomy, due to reactivation of dormant disseminated tumor cells (DTC) in the bone, yet the mechanism by which DTCs are initially induced into a dormant state in the bone remains to be elucidated. We show here that the bone microenvironment confers dormancy to C4-2B4 prostate cancer cells, as they become dormant when injected into mouse femurs but not under the skin. Live-cell imaging of dormant cells at the single-cell level revealed that conditioned medium from differentiated, but not undifferentiated, osteoblasts induced C4-2B4 cellular quiescence, suggesting that differentiated osteoblasts present locally around the tumor cells in the bone conferred dormancy to prostate cancer cells. Gene array analyses identified GDF10 and TGFβ2 among osteoblast-secreted proteins that induced quiescence of C4-2B4, C4-2b, and PC3-mm2, but not 22RV1 or BPH-1 cells, indicating prostate cancer tumor cells differ in their dormancy response. TGFβ2 and GDF10 induced dormancy through TGFβRIII to activate phospho-p38MAPK, which phosphorylates retinoblastoma (RB) at the novel N-terminal S249/T252 sites to block prostate cancer cell proliferation. Consistently, expression of dominant-negative p38MAPK in C4-2b and C4-2B4 prostate cancer cell lines abolished tumor cell dormancy both in vitro and in vivo Lower TGFβRIII expression in patients with prostate cancer correlated with increased metastatic potential and decreased survival rates. Together, our results identify a dormancy mechanism by which DTCs are induced into a dormant state through TGFβRIII-p38MAPK-pS249/pT252-RB signaling and offer a rationale for developing strategies to prevent prostate cancer recurrence in the bone.Significance: These findings provide mechanistic insights into the dormancy of metastatic prostate cancer in the bone and offer a rationale for developing strategies to prevent prostate cancer recurrence in the bone. Cancer Res; 78(11); 2911-24. ©2018 AACR.
Collapse
Affiliation(s)
- Li-Yuan Yu-Lee
- Departments of Medicine and Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| | - Guoyu Yu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yu-Chen Lee
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Song-Chang Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Pan
- Departments of Medicine and Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Tianhong Pan
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kai-Jie Yu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bin Liu
- Center for Cancer Genetics and Genomics and Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pamela A Villalobos
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eulalia de Nadal
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Francesc Posas
- Departament de Ciencies Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
67
|
Meyer RD, Zou X, Ali M, Ersoy E, Bondzie PA, Lavaei M, Alexandrov I, Henderson J, Rahimi N. TMIGD1 acts as a tumor suppressor through regulation of p21Cip1/p27Kip1 in renal cancer. Oncotarget 2017. [PMID: 29515762 PMCID: PMC5839393 DOI: 10.18632/oncotarget.23822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinoma (RCC) is a high-risk metastasizing tumor with a poor prognosis and poorly understood mechanism. In this study, we demonstrate that transmembrane and immunoglobulin domain-containing 1 (TMIGD1) is a novel tumor suppressor that is highly expressed in normal renal tubular epithelial cells, but it is downregulated in human renal cancer. We have identified CCAAT/enhancer-binding proteinβ (C/EBPβ, also called LAP) as a key transcriptional regulator of TMIGD1, whose loss of expression is responsible for downregulation of TMIGD1 in RCC. Transcriptionally active C/EBPβ/LAP physically interacted with and increased TMIGD1 promoter activity and expression of TMIGD1. Re-introduction of TMIGD1 into renal tumor cells significantly inhibited tumor growth and metastatic behaviors such as morphogenic branching and cell migration. Restoring TMIGD1 expression in renal tumor cells stimulated phosphorylation of p38MAK, induced expression of p21CIP1 (cyclin-dependent kinase inhibitor 1), and p27KIP1 (cyclin-dependent kinase inhibitor 1B) expression, key cell cycle inhibitor proteins involved in regulation of the cell cycle. The present study identifies TMIGD1 as a novel candidate tumor suppressor gene and provides important insight into pathobiology of RCC that could lead to a better diagnosis and possible novel therapy for RCC.
Collapse
Affiliation(s)
- Rosana D Meyer
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xueqing Zou
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Marwa Ali
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Esma Ersoy
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Philip Apraku Bondzie
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mehrdad Lavaei
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | - Joel Henderson
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nader Rahimi
- Department of Pathology, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
68
|
Ji L, Zhong B, Jiang X, Mao F, Liu G, Song B, Wang CY, Jiao Y, Wang JP, Xu ZB, Li X, Zhan B. Actein induces autophagy and apoptosis in human bladder cancer by potentiating ROS/JNK and inhibiting AKT pathways. Oncotarget 2017; 8:112498-112515. [PMID: 29348843 PMCID: PMC5762528 DOI: 10.18632/oncotarget.22274] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023] Open
Abstract
Human bladder cancer is a common genitourinary malignant cancer worldwide. However, new therapeutic strategies are required to overcome its stagnated survival rate. Triterpene glycoside Actein (ACT), extracted from the herb black cohosh, suppresses the growth of human breast cancer cells. Our study attempted to explore the role of ACT in human bladder cancer cell growth and to reveal the underlying molecular mechanisms. We found that ACT significantly impeded the bladder cancer cell proliferation via induction of G2/M cycle arrest. Additionally, ACT administration triggered autophagy and apoptosis in bladder cancer cells, proved by the autophagosome formation, LC3B-II accumulation, improved cleavage of Caspases/poly (ADP-ribose) polymerase (PARP). Furthermore, reduction of reactive oxygen species (ROS) and p-c-Jun N-terminal kinase (JNK) could markedly reverse ACT-induced autophagy and apoptosis. In contrast, AKT and mammalian target of rapamycin (mTOR) were greatly de-phosphorylated by ACT, while suppressing AKT and mTOR activity could enhance the effects of ACT on apoptosis and autophagy induction. In vivo, ACT reduced the tumor growth with little toxicity. Taken together, our findings indicated that ACT suppressed cell proliferation, induced autophagy and apoptosis through promoting ROS/JNK activation, and blunting AKT pathway in human bladder cancer, which indicated that ACT might be an effective candidate against human bladder cancer in future.
Collapse
Affiliation(s)
- Lu Ji
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bing Zhong
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Xi Jiang
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Fei Mao
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Gang Liu
- Department of Orthopaedics, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bin Song
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Cheng-Yuan Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Yong Jiao
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Jiang-Ping Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Zhi-Bin Xu
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Xing Li
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Bo Zhan
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| |
Collapse
|
69
|
Ma Y, Fei L, Zhang M, Zhang W, Liu X, Wang C, Luo Y, Zhang H, Han Y. Lamin B2 binding to minichromosome maintenance complex component 7 promotes non-small cell lung carcinogenesis. Oncotarget 2017; 8:104813-104830. [PMID: 29285216 PMCID: PMC5739603 DOI: 10.18632/oncotarget.20338] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/17/2017] [Indexed: 12/13/2022] Open
Abstract
We investigated the role of lamin B2 in non-small cell lung cancer (NSCLC). We detected higher lamin B2 expression in 20 NSCLC tumor tissues obtained from The Cancer Genome Atlas than in adjacent normal lung tissues. LMNB2-RNAi knockdown in A549 and H1299 NSCLC cells inhibited colony formation, cell proliferation and G1-S cell cycle progression while increasing apoptosis. LMNB2 overexpression had the opposite effects. Tumor xenograft experiments showed diminished tumor growth with LMNB2 knockdown H1299 cells than with controls. Yeast two-hybrid studies revealed minichromosome maintenance complex component 7 (MCM7) to be a binding partner of lamin B2, which was confirmed by co-immunoprecipitation and co-localization studies. Lamin B2 binding enhanced DNA binding and helicase activities of MCM7. Deletion analysis with MCM7-N, MCM7-M or MCM7-C mutant proteins showed that lamin B2 binds to the C-terminus of MCM7, and competes with the binding of the tumor suppressor retinoblastoma (RB) protein. Immunohistochemical analysis of 150 NSCLC patient samples revealed that both lamin B2 and MCM7 levels positively correlated with histological grade and tumor TNM stage. Moreover, high lamin B2 and MCM7 levels correlated with shorter overall survival of NSCLC patients. In sum, these results show that lamin B2 interaction with MCM7 promotes NSCLC progression.
Collapse
Affiliation(s)
- Yinan Ma
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Liangru Fei
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Meiyu Zhang
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Wenzhu Zhang
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Xiaofang Liu
- Department of Pathology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Congcong Wang
- Department of Pathology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Yuan Luo
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Haiyan Zhang
- Department of Pathology, The First People's Hospital of Jining, Shandong, China
| | - Yuchen Han
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China.,Department of Pathology, The First Affiliated Hospital of China Medical University, Liaoning, China.,Department of Pathology, Shanghai Chest Hospital, Shanghai, China
| |
Collapse
|
70
|
Chang YL, Tseng SF, Huang YC, Shen ZJ, Hsu PH, Hsieh MH, Yang CW, Tognetti S, Canal B, Subirana L, Wang CW, Chen HT, Lin CY, Posas F, Teng SC. Yeast Cip1 is activated by environmental stress to inhibit Cdk1-G1 cyclins via Mcm1 and Msn2/4. Nat Commun 2017; 8:56. [PMID: 28676626 PMCID: PMC5496861 DOI: 10.1038/s41467-017-00080-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/01/2017] [Indexed: 12/20/2022] Open
Abstract
Upon environmental changes, proliferating cells delay cell cycle to prevent further damage accumulation. Yeast Cip1 is a Cdk1 and Cln2-associated protein. However, the function and regulation of Cip1 are still poorly understood. Here we report that Cip1 expression is co-regulated by the cell-cycle-mediated factor Mcm1 and the stress-mediated factors Msn2/4. Overexpression of Cip1 arrests cell cycle through inhibition of Cdk1–G1 cyclin complexes at G1 stage and the stress-activated protein kinase-dependent Cip1 T65, T69, and T73 phosphorylation may strengthen the Cip1and Cdk1–G1 cyclin interaction. Cip1 accumulation mainly targets Cdk1–Cln3 complex to prevent Whi5 phosphorylation and inhibit early G1 progression. Under osmotic stress, Cip1 expression triggers transient G1 delay which plays a functionally redundant role with another hyperosmolar activated CKI, Sic1. These findings indicate that Cip1 functions similarly to mammalian p21 as a stress-induced CDK inhibitor to decelerate cell cycle through G1 cyclins to cope with environmental stresses. A G1 cell cycle regulatory kinase Cip1 has been identified in budding yeast but how this is regulated is unclear. Here the authors identify cell cycle (Mcm1) and stress-mediated (Msn 2/4) transcription factors as regulating Cip1, causing stress induced CDK inhibition and delay in cell cycle progression.
Collapse
Affiliation(s)
- Ya-Lan Chang
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Shun-Fu Tseng
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.,Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Yu-Ching Huang
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Zih-Jie Shen
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Pang-Hung Hsu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 20224, Taiwan
| | - Meng-Hsun Hsieh
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chia-Wei Yang
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Silvia Tognetti
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Berta Canal
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Laia Subirana
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Chien-Wei Wang
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Hsiao-Tan Chen
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chi-Ying Lin
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Francesc Posas
- Cell Signaling Research Group, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Shu-Chun Teng
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
| |
Collapse
|
71
|
Joaquin M, de Nadal E, Posas F. An RB insensitive to CDK regulation. Mol Cell Oncol 2016; 4:e1268242. [PMID: 28197540 DOI: 10.1080/23723556.2016.1268242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
Abstract
The N-term phosphorylation of Retinoblastoma (RB) by the p38 stress-activated protein kinase (SAPK) makes RB insensitive to cyclin-dependent kinase (CDK)-Cyclin inhibition, which enhances the transcriptional repression of E2F-driven promoters and delays tumor cell growth. This novel mechanism of RB regulation opens up a window for developing new cancer drug treatments for tumors harboring high CDK-Cyclin activity and a wild-type RB gene.
Collapse
Affiliation(s)
- Manel Joaquin
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF) , Barcelona, Spain
| | - Eulàlia de Nadal
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF) , Barcelona, Spain
| | - Francesc Posas
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF) , Barcelona, Spain
| |
Collapse
|