51
|
Rozengurt E, Eibl G. Central role of Yes-associated protein and WW-domain-containing transcriptional co-activator with PDZ-binding motif in pancreatic cancer development. World J Gastroenterol 2019; 25:1797-1816. [PMID: 31057295 PMCID: PMC6478619 DOI: 10.3748/wjg.v25.i15.1797] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease with no efficacious treatment options. PDAC incidence is projected to increase, which may be caused at least partially by the obesity epidemic. Significantly enhanced efforts to prevent or intercept this cancer are clearly warranted. Oncogenic KRAS mutations are recognized initiating events in PDAC development, however, they are not entirely sufficient for the development of fully invasive PDAC. Additional genetic alterations and/or environmental, nutritional, and metabolic signals, as present in obesity, type-2 diabetes mellitus, and inflammation, are required for full PDAC formation. We hypothesize that oncogenic KRAS increases the intensity and duration of the growth-promoting signaling network. Recent exciting studies from different laboratories indicate that the activity of the transcriptional co-activators Yes-associated protein (YAP) and WW-domain-containing transcriptional co-activator with PDZ-binding motif (TAZ) play a critical role in the promotion and maintenance of PDAC operating as key downstream target of KRAS signaling. While initially thought to be primarily an effector of the tumor-suppressive Hippo pathway, more recent studies revealed that YAP/TAZ subcellular localization and co-transcriptional activity is regulated by multiple upstream signals. Overall, YAP has emerged as a central node of transcriptional convergence in growth-promoting signaling in PDAC cells. Indeed, YAP expression is an independent unfavorable prognostic marker for overall survival of PDAC. In what follows, we will review studies implicating YAP/TAZ in pancreatic cancer development and consider different approaches to target these transcriptional regulators.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
- CURE: Digestive Diseases Research Center, Los Angeles, CA 90095, United States
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
- CURE: Digestive Diseases Research Center, Los Angeles, CA 90095, United States
| |
Collapse
|
52
|
Li Y, He J, Qiu C, Shang Q, Qian G, Fan X, Ge S, Jia R. The oncolytic virus H101 combined with GNAQ siRNA-mediated knockdown reduces uveal melanoma cell viability. J Cell Biochem 2019; 120:5766-5776. [PMID: 30320917 DOI: 10.1002/jcb.27863] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/19/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Uveal melanoma (UM) is a severe human malignancy with a high mortality rate, as well as high metastasis and recurrence potential. The active mutation of G protein subunit alpha q (GNAQ) or G protein subunit alpha 11 (GNA11) is a major trigger for UM. Oncolytic adenovirus H101 (H101) is the first oncolytic virus approved for clinical applications in cancer therapy by the China Food and Drug Administration. We investigated whether combining H101 with the downregulation of GNAQ expression would act synergistically in UM therapy. METHODS Three UM cell lines OMM2.3 and 92.1, harboring GNAQ mutation, and OCM1, harboring B-Raf proto-oncogene mutation, were chosen for our research. The cellular toxicity of adenoviral infection and the cell growth rate were measured with a Cell Counting Kit-8. Western blot analysis was used to detect GNAQ, p-MEK1/2, YAP, and p-YAP expression. The apoptosis and cell-cycle distribution of cells were evaluated with annexin-V and propidium iodide staining. RESULTS Our results revealed that OMM2.3 and 92.1 cells were more sensitive to H101 infection than OCM1 cells. GNAQ expression was markedly reduced by small interfering RNA, siGNAQ. Combined treatment of siGNAQ and H101 inhibited the proliferation and activated the apoptosis of OMM2.3 and 92.1 cells by blocking the phosphorylation of MEK1/2 and increasing the phosphorylation of YAP. CONCLUSIONS In summary, a therapy combining H101 and siGNAQ is feasible, with potential utility as a novel targeted molecular therapy for UM, especially those carrying a GNAQ mutation.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jie He
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Chun Qiu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Qingfeng Shang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Guanxiang Qian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
53
|
Chen YA, Lu CY, Cheng TY, Pan SH, Chen HF, Chang NS. WW Domain-Containing Proteins YAP and TAZ in the Hippo Pathway as Key Regulators in Stemness Maintenance, Tissue Homeostasis, and Tumorigenesis. Front Oncol 2019; 9:60. [PMID: 30805310 PMCID: PMC6378284 DOI: 10.3389/fonc.2019.00060] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/21/2019] [Indexed: 12/29/2022] Open
Abstract
The Hippo pathway is a conserved signaling pathway originally defined in Drosophila melanogaster two decades ago. Deregulation of the Hippo pathway leads to significant overgrowth in phenotypes and ultimately initiation of tumorigenesis in various tissues. The major WW domain proteins in the Hippo pathway are YAP and TAZ, which regulate embryonic development, organ growth, tissue regeneration, stem cell pluripotency, and tumorigenesis. Recent reports reveal the novel roles of YAP/TAZ in establishing the precise balance of stem cell niches, promoting the production of induced pluripotent stem cells (iPSCs), and provoking signals for regeneration and cancer initiation. Activation of YAP/TAZ, for example, results in the expansion of progenitor cells, which promotes regeneration after tissue damage. YAP is highly expressed in self-renewing pluripotent stem cells. Overexpression of YAP halts stem cell differentiation and yet maintains the inherent stem cell properties. A success in reprograming iPSCs by the transfection of cells with Oct3/4, Sox2, and Yap expression constructs has recently been shown. In this review, we update the current knowledge and the latest progress in the WW domain proteins of the Hippo pathway in relevance to stem cell biology, and provide a thorough understanding in the tissue homeostasis and identification of potential targets to block tumor development. We also provide the regulatory role of tumor suppressor WWOX in the upstream of TGF-β, Hyal-2, and Wnt signaling that cross talks with the Hippo pathway.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yu Lu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tian-You Cheng
- Department of Optics and Photonics, National Central University, Chungli, Taiwan
| | - Szu-Hua Pan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University, Taipei, Taiwan
| | - Nan-Shan Chang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, United States.,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
54
|
Abstract
Immunohistochemistry (IHC) analysis of YAP in human tissue samples represents an important means to analyze overall expression levels and subcellular localization of YAP in specimen of interest. As transcriptional coactivator, alterations of YAP levels in the cellular nucleus allow important predictions for YAP activity and transcriptional state of target genes. In the following report, IHC procedures optimized for the detection of YAP in tissue slides of FFPE material are provided. Of note, de-paraffinization and heat-induced antigen retrieval are strictly necessary for successful YAP IHC staining. Further, immunostaining using a labelled polymer-HRP system combined with diaminobenzidine (DAB), as signal-amplifying chromogen, allows strong staining results with minimal unspecific background signal.
Collapse
Affiliation(s)
- Franziska Haderk
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Victor Olivas
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA. .,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA. .,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
55
|
Abstract
In contrast to normal cells, which use the aerobic oxidation of glucose as their main energy production method, cancer cells prefer to use anaerobic glycolysis to maintain their growth and survival, even under normoxic conditions. Such tumor cell metabolic reprogramming is regulated by factors such as hypoxia and the tumor microenvironment. In addition, dysregulation of certain signaling pathways also contributes to cancer metabolic reprogramming. Among them, the Hippo signaling pathway is a highly conserved tumor suppressor pathway. The core oncosuppressive kinase cascade of Hippo pathway inhibits the nuclear transcriptional co-activators YAP and TAZ, which are the downstream effectors of Hippo pathway and oncogenic factors in many solid cancers. YAP/TAZ function as key nodes of multiple signaling pathways and play multiple regulatory roles in cancer cells. However, their roles in cancer metabolic reprograming are less clear. In the present review, we examine progress in research into the regulatory mechanisms of YAP/TAZ on glucose metabolism, fatty acid metabolism, mevalonate metabolism, and glutamine metabolism in cancer cells. Determining the roles of YAP/TAZ in tumor energy metabolism, particularly in relation to the tumor microenvironment, will provide new strategies and targets for the selective therapy of metabolism-related cancers.
Collapse
|
56
|
Mechanoregulation and pathology of YAP/TAZ via Hippo and non-Hippo mechanisms. Clin Transl Med 2018; 7:23. [PMID: 30101371 PMCID: PMC6087706 DOI: 10.1186/s40169-018-0202-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023] Open
Abstract
Yes-associated protein (YAP) and its paralog WW domain containing transcription regulator 1 (TAZ) are important regulators of multiple cellular functions such as proliferation, differentiation, and survival. On the tissue level, YAP/TAZ are essential for embryonic development, organ size control and regeneration, while their deregulation leads to carcinogenesis or other diseases. As an underlying principle for YAP/TAZ-mediated regulation of biological functions, a growing body of research reveals that YAP/TAZ play a central role in delivering information of mechanical environments surrounding cells to the nucleus transcriptional machinery. In this review, we discuss mechanical cue-dependent regulatory mechanisms for YAP/TAZ functions, as well as their clinical significance in cancer progression and treatment.
Collapse
|
57
|
Zyxin promotes colon cancer tumorigenesis in a mitotic phosphorylation-dependent manner and through CDK8-mediated YAP activation. Proc Natl Acad Sci U S A 2018; 115:E6760-E6769. [PMID: 29967145 DOI: 10.1073/pnas.1800621115] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Zyxin is a member of the focal adhesion complex and plays a critical role in actin filament polymerization and cell motility. Several recent studies showed that Zyxin is a positive regulator of Yki/YAP (Yes-associated protein) signaling. However, little is known about the mechanisms by which Zyxin itself is regulated and how Zyxin affects Hippo-YAP activity. We first showed that Zyxin is phosphorylated by CDK1 during mitosis. Depletion of Zyxin resulted in significantly impaired colon cancer cell proliferation, migration, anchorage-independent growth, and tumor formation in xenograft animal models. Mitotic phosphorylation is required for Zyxin activity in promoting growth. Zyxin regulates YAP activity through the colon cancer oncogene CDK8. CDK8 knockout phenocopied Zyxin knockdown in colon cancer cells, while ectopic expression of CDK8 substantially restored the tumorigenic defects of Zyxin-depletion cells. Mechanistically, we showed that CDK8 directly phosphorylated YAP and promoted its activation. Fully activated YAP is required to support the growth in CDK8-knockout colon cancer cells in vitro and in vivo. Together, these observations suggest that Zyxin promotes colon cancer tumorigenesis in a mitotic-phosphorylation-dependent manner and through CDK8-mediated YAP activation.
Collapse
|
58
|
Yes-associated protein (YAP) in pancreatic cancer: at the epicenter of a targetable signaling network associated with patient survival. Signal Transduct Target Ther 2018; 3:11. [PMID: 29682330 PMCID: PMC5908807 DOI: 10.1038/s41392-017-0005-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/27/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is generally a fatal disease with no efficacious treatment modalities. Elucidation of signaling mechanisms that will lead to the identification of novel targets for therapy and chemoprevention is urgently needed. Here, we review the role of Yes-associated protein (YAP) and WW-domain-containing Transcriptional co-Activator with a PDZ-binding motif (TAZ) in the development of PDAC. These oncogenic proteins are at the center of a signaling network that involves multiple upstream signals and downstream YAP-regulated genes. We also discuss the clinical significance of the YAP signaling network in PDAC using a recently published interactive open-access database (www.proteinatlas.org/pathology) that allows genome-wide exploration of the impact of individual proteins on survival outcomes. Multiple YAP/TEAD-regulated genes, including AJUBA, ANLN, AREG, ARHGAP29, AURKA, BUB1, CCND1, CDK6, CXCL5, EDN2, DKK1, FOSL1,FOXM1, HBEGF, IGFBP2, JAG1, NOTCH2, RHAMM, RRM2, SERP1, and ZWILCH, are associated with unfavorable survival of PDAC patients. Similarly, components of AP-1 that synergize with YAP (FOSL1), growth factors (TGFα, EPEG, and HBEGF), a specific integrin (ITGA2), heptahelical receptors (P2Y2R, GPR87) and an inhibitor of the Hippo pathway (MUC1), all of which stimulate YAP activity, are associated with unfavorable survival of PDAC patients. By contrast, YAP inhibitory pathways (STRAD/LKB-1/AMPK, PKA/LATS, and TSC/mTORC1) indicate a favorable prognosis. These associations emphasize that the YAP signaling network correlates with poor survival of pancreatic cancer patients. We conclude that the YAP pathway is a major determinant of clinical aggressiveness in PDAC patients and a target for therapeutic and preventive strategies in this disease. Yes-associated protein (YAP) signaling contributes to pancreatic cancer progression and is associated with poor patient survival. Previous studies have shown that YAP activates genes involved in cell proliferation to incite tumor growth and metastasis. Enrique Rozengurt and colleagues at University of California Los Angeles review the latest knowledge on YAP signaling and used the open access database The Human Protein Atlas to analyze the gene expression profile and prognosis of 176 patients with pancreatic ductal adenocarcinoma. Activation of upstream or downstream elements of the YAP signaling pathway correlated with shorter survival in patients. Conversely, the activation of signaling pathways that oppose YAP signaling were associated with a more favorable prognosis. These findings highlight YAP signaling pathway components as both prognostic markers and potential targets for developing much needed therapeutic and preventative strategies.
Collapse
|
59
|
Warren JSA, Xiao Y, Lamar JM. YAP/TAZ Activation as a Target for Treating Metastatic Cancer. Cancers (Basel) 2018; 10:cancers10040115. [PMID: 29642615 PMCID: PMC5923370 DOI: 10.3390/cancers10040115] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ) have both emerged as important drivers of cancer progression and metastasis. YAP and TAZ are often upregulated or nuclear localized in aggressive human cancers. There is abundant experimental evidence demonstrating that YAP or TAZ activation promotes cancer formation, tumor progression, and metastasis. In this review we summarize the evidence linking YAP/TAZ activation to metastasis, and discuss the roles of YAP and TAZ during each step of the metastatic cascade. Collectively, this evidence strongly suggests that inappropriate YAP or TAZ activity plays a causal role in cancer, and that targeting aberrant YAP/TAZ activation is a promising strategy for the treatment of metastatic disease. To this end, we also discuss several potential strategies for inhibiting YAP/TAZ activation in cancer and the challenges each strategy poses.
Collapse
Affiliation(s)
- Janine S A Warren
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
60
|
Jiang Z, Zhou C, Cheng L, Yan B, Chen K, Chen X, Zong L, Lei J, Duan W, Xu Q, Li X, Wang Z, Ma Q, Ma J. Inhibiting YAP expression suppresses pancreatic cancer progression by disrupting tumor-stromal interactions. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:69. [PMID: 29587800 PMCID: PMC5870346 DOI: 10.1186/s13046-018-0740-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/16/2018] [Indexed: 12/21/2022]
Abstract
Background Hippo/YAP pathway is known to be important for development, growth and organogenesis, and dysregulation of this pathway leads to tumor progression. We and others find that YAP is up-regulated in pancreatic ductal adenocarcinoma (PDAC) and associated with worse prognosis of patients. Activated pancreatic stellate cells (PSCs) forming the components of microenvironment that enhance pancreatic cancer cells (PCs) invasiveness and malignance. However, the role and mechanism of YAP in PDAC tumor-stromal interaction is largely unknown. Methods The expression of YAP in Pancreatic cancer cell lines and PDAC samples was examined by Western blot and IHC. The biological role of YAP on cancer cell proliferation, epithelial-mesenchymal transition (EMT) and invasion were evaluated by MTT, Quantitative real-time PCR analysis, Western blot analysis and invasion assay. The effect of YAP on PSC activation was evaluated by PC-PSC co-culture conditions and xenograft PDAC mouse model. Results Firstly, knockdown of YAP inhibits PDAC cell proliferation and invasion in vitro. In addition, YAP modulates the PC and PSC interaction via reducing the production of connective tissue growth factor (CTGF) from PCs, inhibits paracrine-mediated PSC activation under PC-PSC co-culture conditions and in turn disrupts TGF-β1-mediated tumor-stromal interactions. Lastly, inhibiting YAP expression prevents tumor growth and suppresses desmoplastic reaction in vivo. Conclusions These results demonstrate that YAP contributes to the proliferation and invasion of PC and the activation of PSC via tumor-stromal interactions and that targeting YAP may be a promising therapeutic strategy for PDAC treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-0740-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liang Cheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Bin Yan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liang Zong
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| | - Jiguang Ma
- Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
61
|
Greenwood E, Maisel S, Ebertz D, Russ A, Pandey R, Schroeder J. Llgl1 prevents metaplastic survival driven by epidermal growth factor dependent migration. Oncotarget 2018; 7:60776-60792. [PMID: 27542214 PMCID: PMC5308616 DOI: 10.18632/oncotarget.11320] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/02/2016] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that Llgl1 loss results in a gain of mesenchymal phenotypes and a loss of apicobasal and planar polarity. We now demonstrate that these changes represent a fundamental shift in cellular phenotype. Llgl1 regulates the expression of multiple cell identity markers, including CD44, CD49f, and CD24, and the nuclear translocation of TAZ and Slug. Cells lacking Llgl1 form mammospheres, where survival and transplantability is dependent upon the Epidermal Growth Factor Receptor (EGFR). Additionally, Llgl1 loss allows cells to grow in soft-agar and maintain prolonged survival as orthotopic transplants in NOD-SCIDmice. Lineage tracing and wound healing experiments demonstrate that mammosphere survival is due to enhanced EGF-dependent migration. The loss of Llgl1 drives EGFR mislocalization and an EGFR mislocalization point mutation (P667A) drives these same phenotypes, including activation of AKT and TAZ nuclear translocation. Together, these data indicate that the loss of Llgl1 results in EGFR mislocalization, promoting pre-neoplastic changes.
Collapse
Affiliation(s)
- Erin Greenwood
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| | - Sabrina Maisel
- Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Cancer Biology Program, University of Arizona, Tucson, Arizona
| | - David Ebertz
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| | - Atlantis Russ
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona.,Genetics Program, University of Arizona, Tucson, Arizona
| | - Ritu Pandey
- Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Joyce Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona.,Arizona Cancer Center, University of Arizona, Tucson, Arizona.,BIO5 Institute, University of Arizona, Tucson, Arizona.,Genetics Program, University of Arizona, Tucson, Arizona.,Cancer Biology Program, University of Arizona, Tucson, Arizona
| |
Collapse
|
62
|
Zhou Y, Shan T, Ding W, Hua Z, Shen Y, Lu Z, Chen B, Dai T. Study on mechanism about long noncoding RNA MALAT1 affecting pancreatic cancer by regulating Hippo-YAP signaling. J Cell Physiol 2018; 233:5805-5814. [PMID: 29215734 DOI: 10.1002/jcp.26357] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022]
Abstract
By investigating the migration and invasion ability in pancreatic cancer, this study probed into the lncRNA MALAT1 molecular mechanism on Hippo-YAP signaling. The expression of lncRNA MALAT1 in PC tissues and cells was detected by qRT-PCR and Western blot. The effect of si-MALAT1 on proliferation was determined by CCK-8 assay. Cell apoptosis, migration, and invasion were respectively detected by flow cytometry assay, wound healing assay, and transwell assay. Western blot and immunohistochemistry were successively used for detecting LATS1 and YAP1 expression in pancreatic cancer tissues. The microarray analysis determined that lncRNA MALAT1 in pancreatic cancer was highly expressed. LncRNA MALAT1 presented an extremely high expression level in pancreatic cancer tissues and cells. After transfected with si-MALAT1, the proliferation of AsPC-1 cells decreased, induce apoptosis of AsPC-1 cells, and migration and invasion ability were reduced. The tendency of LATS1 expression level was down-regulated and YAP1 show the opposite trend in the Hippo-YAP signaling. The in vivo assay was found that the tumor to be small in size and volume, and the expression of Ki-67 was decreased. High expression of lncRNA MALAT1 in PC disorder the proliferation, apoptosis, and migration and invasion ability via influence Hippo-YAP signaling pathway.
Collapse
Affiliation(s)
- Yongping Zhou
- Department of Hepatobiliary, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ting Shan
- Department of General Surgery, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Wenzhou Ding
- Department of Hepatobiliary, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhiyuan Hua
- Department of Hepatobiliary, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yijun Shen
- Department of Hepatobiliary, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhihua Lu
- Department of Hepatobiliary, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Bo Chen
- Department of Hepatobiliary, East Hospital Affiliated to Tongji University, Shanghai, China
| | - Tu Dai
- Department of Hepatobiliary, Wuxi Second Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
63
|
Lin KC, Park HW, Guan KL. Deregulation and Therapeutic Potential of the Hippo Pathway in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kimberly C. Lin
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
64
|
Zhang W, Shen J, Gu F, Zhang Y, Wu W, Weng J, Liao Y, Deng Z, Yuan Q, Zheng L, Zhang Y, Shen W. Monopolar spindle-one-binder protein 2 regulates the activity of large tumor suppressor/yes-associated protein to inhibit the motility of SMMC-7721 hepatocellular carcinoma cells. Oncol Lett 2018; 15:5375-5383. [PMID: 29552181 DOI: 10.3892/ol.2018.7952] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 01/03/2018] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence implicates monopolar spindle-one-binder protein (MOB)2 as an inhibitor of nuclear-Dbf2-related kinase (NDR) by competing with MOB1 for interaction with NDR1/2. NDR/large tumor suppressor (LATS) kinases may function similarly to yes-associated protein (YAP) kinases and be considered as members of the Hippo core cassette. MOB2 appears to serve roles in cell survival, cell cycle progression, responses to DNA damage and cell motility. However, the underlying mechanisms involved remain unclarified. In the present study, it was demonstrated that the knockout of MOB2 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 promoted migration and invasion, induced phosphorylation of NDR1/2 and decreased phosphorylation of YAP in SMMC-7721 cells when compared with the blank vector-transduced cells. By contrast, the overexpression of MOB2 resulted in the opposite results. Mechanistically, MOB2 regulated the alternative interaction of MOB1 with NDR1/2 and LATS1, which resulted in increased phosphorylation of LATS1 and MOB1 and thereby led to the inactivation of YAP and consequently inhibition of cell motility. The results of the present study provide evidence of MOB2 serving a positive role in LATS/YAP activation by activating the Hippo signaling pathway.
Collapse
Affiliation(s)
- Weicheng Zhang
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jingyuan Shen
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Fengming Gu
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Wenjuan Wu
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China.,Department of Medical Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jiachun Weng
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yuexia Liao
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Zijing Deng
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Qing Yuan
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Lu Zheng
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yu Zhang
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, P.R. China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Weigan Shen
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, P.R. China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
65
|
Ciamporcero E, Daga M, Pizzimenti S, Roetto A, Dianzani C, Compagnone A, Palmieri A, Ullio C, Cangemi L, Pili R, Barrera G. Crosstalk between Nrf2 and YAP contributes to maintaining the antioxidant potential and chemoresistance in bladder cancer. Free Radic Biol Med 2018; 115:447-457. [PMID: 29248722 DOI: 10.1016/j.freeradbiomed.2017.12.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/25/2022]
Abstract
Redox adaptation plays an important role in cancer cells drug resistance. The antioxidant response is principally mediated by the transcription factor Nrf2, that induces the transcriptional activation of several genes involved in GSH synthesis, chemoresistance, and cytoprotection. YAP is emerging as a key mediator of chemoresistance in a variety of cancers, but its role in controlling the antioxidant status of the cells is yet elusive. Here, we show that impairing YAP protein expression reduced GSH content and Nrf2 protein and mRNA expression in bladder cancer cells. Moreover, in YAP knocked down cells the expression of FOXM1, a transcription factor involved in Nrf2 transcription, was down-regulated and the silencing of FOXM1 reduced Nrf2 expression. On the other hand, the silencing of Nrf2, as well as the depletion of GSH by BSO treatment, inhibited YAP expression, suggesting that cross-talk exists between YAP and Nrf2 proteins. Importantly, we found that silencing either YAP or Nrf2 enhanced sensitivity of bladder cancer cells to cytotoxic agents and reduced their migration. Furthermore, the inhibition of both YAP and Nrf2 expressions significantly increased cytotoxic drug sensitivity and synergistically reduced the migration of chemoresistant bladder cancer cells. These findings provide a rationale for targeting these transcriptional regulators in patients with chemoresistant bladder cancer, expressing high YAP and bearing a proficient antioxidant system.
Collapse
Affiliation(s)
- Eric Ciamporcero
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Martina Daga
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy.
| | - Antonella Roetto
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Alessandra Compagnone
- Department of Oncology, University of Turin, Via Michelangelo 27, 10125 Turin, Italy
| | - Antonietta Palmieri
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Chiara Ullio
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Luigi Cangemi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Roberto Pili
- Department of Medicine, Genitourinary Program, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA; Genitourinary Program, Indiana University-Simon Cancer Center, Hematology/Oncology980 W. Walnut Street R3 C516, Indianapolis, IN 46202, USA
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| |
Collapse
|
66
|
Zhang Y, Xie P, Wang X, Pan P, Wang Y, Zhang H, Dong Y, Shi Y, Jiang Y, Yu R, Zhou X. YAP Promotes Migration and Invasion of Human Glioma Cells. J Mol Neurosci 2018; 64:262-272. [PMID: 29306996 DOI: 10.1007/s12031-017-1018-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Previously, we have reported that Yes-associated protein (YAP) is upregulated in human glioma tissues and its level is positively correlated with patient prognosis. However, the role and mechanism of YAP in the highly invasive nature of human gliomas were largely unknown. In this study, examined by wound healing assay, transwell assay, or live-imaging, we found that YAP downregulation inhibited glioma cell migration and invasion, while YAP over-expression promoted them. Interestingly, the above effect of YAP on immortalized glioma cells was recapitulated in cultured primary glioma cells. In addition, the protein level of N-cadherin and Twist, two important proteins involved in tumor invasion, increased after YAP over-expression. Meanwhile, YAP over-expression significantly increased the F-actin level and changed the distribution of F-actin, leading to cytoskeletal reorganization, which plays an important role in cell motility. Furthermore, the promotion effect of YAP over-expression on glioma cell migration and invasion was partially abolished by Twist downregulation. Taken together, our findings show that YAP contributes to glioma cell migration and invasion by regulating N-cadherin and Twist, as well as cytoskeletal reorganization.
Collapse
Affiliation(s)
- Yu Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng Xie
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurosurgery, Huaian Second People's Hospital, Huaian, 223002, China
| | - Xu Wang
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Peng Pan
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan Wang
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hao Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Dong
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yi Shi
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Jiang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Xiuping Zhou
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
67
|
Netrin-1 promotes metastasis of gastric cancer by regulating YAP activity. Biochem Biophys Res Commun 2018; 496:76-82. [PMID: 29305865 DOI: 10.1016/j.bbrc.2017.12.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 12/31/2017] [Indexed: 11/23/2022]
Abstract
Yes-associated protein (YAP) is a major downstream molecular of the Hippo pathway, which plays important role in cancer development. Netrin-1 conveys oncogenic activity in many types of malignant tumors. However, the downstream signaling of netrin-1 mediating its oncogenic effects in gastric cancer (GC) is not well defined. Here, we aim to investigate the role of netrin-1 in metastasis potential of GC by regulating YAP. In this study, we showed that netrin-1 inhibition significantly decreased migration and invasion abilities of GC cells, while netrin-1 overexpression effectively reversed this effect. We also demonstrated that netrin-1 upregulated YAP expression via its transmembrane receptor neogenin. Furthermore, our in vitro and in vivo results showed that the effect of netrin-1 on GC cells migration and invasion abilities was regulated by YAP. Collectively, our results defined netrin-1 as a positive regulator of malignant tumor metastasis in GC by activating the YAP signaling, with potential implications for new approaches to GC therapy.
Collapse
|
68
|
Amyloid precursor protein and amyloid precursor-like protein 2 in cancer. Oncotarget 2017; 7:19430-44. [PMID: 26840089 PMCID: PMC4991393 DOI: 10.18632/oncotarget.7103] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/23/2016] [Indexed: 12/22/2022] Open
Abstract
Amyloid precursor protein (APP) and its family members amyloid precursor-like protein 1 (APLP1) and amyloid precursor-like protein 2 (APLP2) are type 1 transmembrane glycoproteins that are highly conserved across species. The transcriptional regulation of APP and APLP2 is similar but not identical, and the cleavage of both proteins is regulated by phosphorylation. APP has been implicated in Alzheimer's disease causation, and in addition to its importance in neurology, APP is deregulated in cancer cells. APLP2 is likewise overexpressed in cancer cells, and APLP2 and APP are linked to increased tumor cell proliferation, migration, and invasion. In this present review, we discuss the unfolding account of these APP family members’ roles in cancer progression and metastasis.
Collapse
|
69
|
Eibl G, Rozengurt E. KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol 2017; 54:50-62. [PMID: 29079305 DOI: 10.1016/j.semcancer.2017.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/22/2017] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be a lethal disease with no efficacious treatment modalities. The incidence of PDAC is expected to increase, at least partially because of the obesity epidemic. Increased efforts to prevent or intercept this disease are clearly needed. Mutations in KRAS are initiating events in pancreatic carcinogenesis supported by genetically engineered mouse models of the disease. However, oncogenic KRAS is not entirely sufficient for the development of fully invasive PDAC. Additional genetic mutations and/or environmental, nutritional, and metabolic stressors, e.g. inflammation and obesity, are required for efficient PDAC formation with activation of KRAS downstream effectors. Multiple factors "upstream" of KRAS associated with obesity, including insulin resistance, inflammation, changes in gut microbiota and GI peptides, can enhance/modulate downstream signals. Multiple signaling networks and feedback loops "downstream" of KRAS have been described that respond to obesogenic diets. We propose that KRAS mutations potentiate a signaling network that is promoted by environmental factors. Specifically, we envisage that KRAS mutations increase the intensity and duration of the growth-promoting signaling network. As the transcriptional activator YAP plays a critical role in the network, we conclude that the rationale for targeting the network (at different points), e.g. with FDA approved drugs such as statins and metformin, is therefore compelling.
Collapse
Affiliation(s)
- Guido Eibl
- Departments of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, CA, United States.
| | - Enrique Rozengurt
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
70
|
Tan Y, Li QM, Huang N, Cheng S, Zhao GJ, Chen H, Chen S, Tang ZH, Zhang WQ, Huang Q, Cheng Y. Upregulation of DACT2 suppresses proliferation and enhances apoptosis of glioma cell via inactivation of YAP signaling pathway. Cell Death Dis 2017; 8:e2981. [PMID: 28796248 PMCID: PMC5596571 DOI: 10.1038/cddis.2017.385] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/02/2017] [Accepted: 07/07/2017] [Indexed: 01/01/2023]
Abstract
DACT2, one of the Dact gene family members, was shown to function as a tumor suppressor. However, its function in gliomas remains largely unknown. In this study, we investigated the role of DACT2, underlying molecular mechanisms and its clinical significance in glioma patients. Downexpression of DACT2 in gliomas compared with adjacent normal brain tissues was correlated with glioma grade and poor survival. Cox regression analysis revealed that the DACT2 is an independent prognostic indicator for glioma patients. Overexpression of DACT2 in glioma cells inhibited proliferation, cell cycle and enhanced apoptosis, sensitivity to temozolomide in vitro and suppressed tumor growth in vivo. Whereas knockdown of DACT2 induce opposite reaction. Mechanistically, overexpression of DACT2 resulted in upregulation of important signaling molecules such as p-YAP and p-β-catenin, and prevent YAP translocating into nucleus and sequestering in the cytoplasm to degrade. The study further proved that DACT2 can suppress YAP through Wnt/β-catenin signaling pathway. Collectively, these data indicate that DACT2 has a tumor suppressor function via inactivation of YAP pathway, providing a promising target for the treatment of gliomas.
Collapse
Affiliation(s)
- Ying Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiu-Meng Li
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si Cheng
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guan-Jian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhao-Hua Tang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen-Qian Zhang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
71
|
Xia J, Zeng M, Zhu H, Chen X, Weng Z, Li S. Emerging role of Hippo signalling pathway in bladder cancer. J Cell Mol Med 2017; 22:4-15. [PMID: 28782275 PMCID: PMC5742740 DOI: 10.1111/jcmm.13293] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/31/2017] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer (BC) is one of the most common cancers worldwide with a high progression rate and poor prognosis. The Hippo signalling pathway is a conserved pathway that plays a crucial role in cellular proliferation, differentiation and apoptosis. Furthermore, dysregulation and/or malfunction of the Hippo pathway is common in various human tumours, including BC. In this review, an overview of the Hippo pathway in BC and other cancers is presented. We focus on recent data regarding the Hippo pathway, its network and the regulation of the downstream co-effectors YAP1/TAZ. The core components of the Hippo pathway, which induce BC stemness acquisition, metastasis and chemoresistance, will be emphasized. Additional research on the Hippo pathway will advance our understanding of the mechanism of BC as well as the development and progression of other cancers and may be exploited therapeutically.
Collapse
Affiliation(s)
- Jianling Xia
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Hospital of the University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ming Zeng
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Hospital of the University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hua Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangjian Chen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiliang Weng
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shi Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
72
|
Zeng Y, Stauffer S, Zhou J, Chen X, Chen Y, Dong J. Cyclin-dependent kinase 1 (CDK1)-mediated mitotic phosphorylation of the transcriptional co-repressor Vgll4 inhibits its tumor-suppressing activity. J Biol Chem 2017; 292:15028-15038. [PMID: 28739871 DOI: 10.1074/jbc.m117.796284] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/12/2017] [Indexed: 01/07/2023] Open
Abstract
The Hippo pathway is an evolutionarily conserved signaling pathway that plays important roles in stem cell biology, tissue homeostasis, and cancer development. Vestigial-like 4 (Vgll4) functions as a transcriptional co-repressor in the Hippo-Yes-associated protein (YAP) pathway. Vgll4 inhibits cell proliferation and tumor growth by competing with YAP for binding to TEA-domain proteins (TEADs). However, the mechanisms by which Vgll4 itself is regulated are unclear. Here we identified a mechanism that regulates Vgll4's tumor-suppressing function. We found that Vgll4 is phosphorylated in vitro and in vivo by cyclin-dependent kinase 1 (CDK1) during antimitotic drug-induced mitotic arrest and also in normal mitosis. We further identified Ser-58, Ser-155, Thr-159, and Ser-280 as the main mitotic phosphorylation sites in Vgll4. We also noted that the nonphosphorylatable mutant Vgll4-4A (S58A/S155A/T159A/S280A) suppressed tumorigenesis in pancreatic cancer cells in vitro and in vivo to a greater extent than did wild-type Vgll4, suggesting that mitotic phosphorylation inhibits Vgll4's tumor-suppressive activity. Consistent with these observations, the Vgll4-4A mutant possessed higher-binding affinity to TEAD1 than wild-type Vgll4. Interestingly, Vgll4 and Vgll4-4A markedly suppressed YAP and β-catenin signaling activity. Together, these findings reveal a previously unrecognized mechanism for Vgll4 regulation in mitosis and its role in tumorigenesis.
Collapse
Affiliation(s)
- Yongji Zeng
- From the Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center.,Department of Pathology and Microbiology, and
| | - Seth Stauffer
- From the Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center.,Department of Pathology and Microbiology, and
| | - Jiuli Zhou
- From the Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center.,Department of Pathology and Microbiology, and
| | - Xingcheng Chen
- From the Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center.,Department of Pathology and Microbiology, and
| | - Yuanhong Chen
- From the Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center
| | - Jixin Dong
- From the Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, .,Department of Pathology and Microbiology, and.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|
73
|
Wei H, Xu Z, Liu F, Wang F, Wang X, Sun X, Li J. Hypoxia induces oncogene yes-associated protein 1 nuclear translocation to promote pancreatic ductal adenocarcinoma invasion via epithelial-mesenchymal transition. Tumour Biol 2017; 39:1010428317691684. [PMID: 28475017 DOI: 10.1177/1010428317691684] [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] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most lethal cancers. The Hippo pathway is involved in tumorigenesis and remodeling of tumor microenvironments. Hypoxia exists in the microenvironment of solid tumors, including pancreatic ductal adenocarcinoma and plays a vital role in tumor progression and metastasis. However, it remains unclear how hypoxia interacts with the Hippo pathway to regulate these events. In this study, expressions of yes-associated protein 1 and hypoxia-inducible factor-1α were found to be elevated in pancreatic ductal adenocarcinoma samples compared with those in matched adjacent non-tumor samples. Moreover, hypoxia-inducible factor-1α expression was positively correlated with yes-associated protein 1 level in pancreatic ductal adenocarcinoma tissues. The higher expression of nuclear yes-associated protein 1 was associated with poor histological grade and prognosis for pancreatic ductal adenocarcinoma patients. In vitro, yes-associated protein 1 was highly expressed in pancreatic ductal adenocarcinoma cells. Depletion of yes-associated protein 1 inhibited the invasion of pancreatic ductal adenocarcinoma cells via downregulation of Vimentin, matrix metalloproteinase-2, and matrix metalloproteinase-13, and upregulation of E-cadherin. In addition, hypoxia promoted the invasion of pancreatic ductal adenocarcinoma cells via regulating the targeted genes. Hypoxia also deactivated the Hippo pathway and induced yes-associated protein 1 nuclear translocation. Furthermore, depletion of yes-associated protein 1 or hypoxia-inducible factor-1α suppressed the invasion of pancreatic ductal adenocarcinoma cells under hypoxia. Mechanism studies showed that nuclear yes-associated protein 1 interacted with hypoxia-inducible factor-1α and activated Snail transcription to participate in epithelial-mesenchymal transition-mediated and matrix metalloproteinase-mediated remodeling of tumor microenvironments. Collectively, yes-associated protein 1 is an independent prognostic predictor that interacts with hypoxia-inducible factor-1α to enhance the invasion of pancreatic cancer cells and remodeling of tumor microenvironments. Therefore, yes-associated protein 1 may serve as a novel promising target to enhance therapeutic effects for treating pancreatic cancer.
Collapse
Affiliation(s)
- Honglong Wei
- 1 Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zongzhen Xu
- 1 Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Feng Liu
- 1 Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Fuhai Wang
- 1 Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xin Wang
- 1 Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xueying Sun
- 2 Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jie Li
- 1 Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
74
|
A combinatorial strategy using YAP and pan-RAF inhibitors for treating KRAS-mutant pancreatic cancer. Cancer Lett 2017; 402:61-70. [PMID: 28576749 DOI: 10.1016/j.canlet.2017.05.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/13/2017] [Accepted: 05/19/2017] [Indexed: 12/13/2022]
Abstract
KRAS mutation is the most common genetic event in pancreatic cancer. Whereas KRAS itself has proven difficult to inhibit, agents that target key downstream signals of KRAS, such as RAF, are possibly effective for pancreatic cancer treatment. Because selective BRAF inhibitors paradoxically induce downstream signaling activation, a pan-RAF inhibitor, LY3009120 is a better alternate for KRAS-mutant tumor treatment. Here we explored a new combinational strategy using a YAP inhibitor and LY3009120 in pancreatic cancer treatment. We found that reduced YAP expression closely correlates with longer relapse-free and overall survival of patients. Stable knockdown of YAP significantly inhibited pancreatic cancer cell proliferation and tumor growth. In addition, LY3009120 exhibited a dramatically enhanced antitumor effect in combination with YAP knockdown. YAP depletion blocks the activation of a parallel AKT signal pathway after LY3009120 treatment. Finally, combination with a YAP inhibitor, verteporfin, significantly enhanced the antitumor efficacy of LY3009120. Collectively, our results demonstrate that genetic or pharmacological inhibition of YAP can increase sensitivity to LY3009120 in pancreatic cancer through blocking compensatory activation of a parallel AKT signal pathway, thereby validating a combinatorial approach for treating KRAS-mutant pancreatic cancer.
Collapse
|
75
|
Quan M, Cui JJ, Feng X, Huang Q. The critical role and potential target of the autotaxin/lysophosphatidate axis in pancreatic cancer. Tumour Biol 2017; 39:1010428317694544. [PMID: 28347252 DOI: 10.1177/1010428317694544] [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] [Indexed: 01/27/2023] Open
Abstract
Autotaxin, an ecto-lysophospholipase D encoded by the human ENNP2 gene, is expressed in multiple tissues, and participates in numerous critical physiologic and pathologic processes including inflammation, pain, obesity, embryo development, and cancer via the generation of the bioactive lipid lysophosphatidate. Overwhelming evidences indicate that the autotaxin/lysophosphatidate signaling axis serves key roles in the numerous processes central to tumorigenesis and progression, including proliferation, survival, migration, invasion, metastasis, cancer stem cell, tumor microenvironment, and treatment resistance by interacting with a series of at least six G-protein-coupled receptors (LPAR1-6). This review provides an overview of the autotaxin/lysophosphatidate axis and collates current knowledge regarding its specific role in pancreatic cancer. With a deeper understanding of the critical role of the autotaxin/lysophosphatidate axis in pancreatic cancer, targeting autotaxin or lysophosphatidate receptor may be a potential and promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Ming Quan
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jiu-Jie Cui
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiao Feng
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Qian Huang
- Cancer Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
76
|
Hao F, Xu Q, Zhao Y, Stevens JV, Young SH, Sinnett-Smith J, Rozengurt E. Insulin Receptor and GPCR Crosstalk Stimulates YAP via PI3K and PKD in Pancreatic Cancer Cells. Mol Cancer Res 2017; 15:929-941. [PMID: 28360038 DOI: 10.1158/1541-7786.mcr-17-0023] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 01/30/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
We examined the impact of crosstalk between the insulin receptor and G protein-coupled receptor (GPCR) signaling pathways on the regulation of Yes-associated protein (YAP) localization, phosphorylation, and transcriptional activity in the context of human pancreatic ductal adenocarcinoma (PDAC). Stimulation of PANC-1 or MiaPaCa-2 cells with insulin and neurotensin, a potent mitogenic combination of agonists for these cells, promoted striking YAP nuclear localization and decreased YAP phosphorylation at Ser127 and Ser397 Challenging PDAC cells with either insulin or neurotensin alone modestly induced the expression of YAP/TEAD-regulated genes, including connective tissue growth factor (CTGF), cysteine-rich angiogenic inducer 61 (CYR61), and CXCL5, whereas the combination of neurotensin and insulin induced a marked increase in the level of expression of these genes. In addition, siRNA-mediated knockdown of YAP/TAZ prevented the increase in the expression of these genes. A small-molecule inhibitor (A66), selective for the p110α subunit of PI3K, abrogated the increase in phosphatidylinositol 3,4,5-trisphosphate production and the expression of CTGF, CYR61, and CXCL5 induced by neurotensin and insulin. Furthermore, treatment of PDAC cells with protein kinase D (PKD) family inhibitors (CRT0066101 or kb NB 142-70) or with siRNAs targeting the PKD family prevented the increase of CTGF, CYR61, and CXCL5 mRNA levels in response to insulin and neurotensin stimulation. Thus, PI3K and PKD mediate YAP activation in response to insulin and neurotensin in pancreatic cancer cells.Implications: Inhibitors of PI3K or PKD disrupt crosstalk between insulin receptor and GPCR signaling systems by blocking YAP/TEAD-regulated gene expression in pancreatic cancer cells. Mol Cancer Res; 15(7); 929-41. ©2017 AACR.
Collapse
Affiliation(s)
- Fang Hao
- Tianjin Medical University, Tianjin, China.,Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Qinhong Xu
- Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California.,Xi'an Jiaotong University, Xi'an, China
| | - Yinglan Zhao
- Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California.,Sichuan University, Chengdu, China
| | - Jan V Stevens
- Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Steven H Young
- Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California.,CURE: Digestive Disease Research Center, Los Angeles, California.,VA Greater Los Angeles Health Care System, Los Angeles, California
| | - James Sinnett-Smith
- Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California.,CURE: Digestive Disease Research Center, Los Angeles, California.,VA Greater Los Angeles Health Care System, Los Angeles, California
| | - Enrique Rozengurt
- Division of Digestive Disease, Department of Medicine, David Geffen School of Medicine, Los Angeles, California. .,CURE: Digestive Disease Research Center, Los Angeles, California.,VA Greater Los Angeles Health Care System, Los Angeles, California.,Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
77
|
Landrith TA, Sureshchandra S, Rivera A, Jang JC, Rais M, Nair MG, Messaoudi I, Wilson EH. CD103 + CD8 T Cells in the Toxoplasma-Infected Brain Exhibit a Tissue-Resident Memory Transcriptional Profile. Front Immunol 2017; 8:335. [PMID: 28424687 PMCID: PMC5372813 DOI: 10.3389/fimmu.2017.00335] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
During chronic infection, memory T cells acquire a unique phenotype and become dependent on different survival signals than those needed for memory T cells generated during an acute infection. The distinction between the role of effector and memory T cells in an environment of persistent antigen remains unclear. Here, in the context of chronic Toxoplasma gondii infection, we demonstrate that a population of CD8 T cells exhibiting a tissue-resident memory (TRM) phenotype accumulates within the brain. We show that this population is distributed throughout the brain in both parenchymal and extraparenchymal spaces. Furthermore, this population is transcriptionally distinct and exhibits a transcriptional signature consistent with the TRM observed in acute viral infections. Finally, we establish that the CD103+ TRM population has an intrinsic capacity to produce both IFN-γ and TNF-α, cytokines critical for parasite control within the central nervous system (CNS). The contribution of this population to pro-inflammatory cytokine production suggests an important role for TRM in protective and ongoing immune responses in the infected CNS. Accession number: GSE95105
Collapse
Affiliation(s)
- Tyler A Landrith
- School of Medicine, University of California, Riverside, CA, USA
| | | | - Andrea Rivera
- School of Medicine, University of California, Riverside, CA, USA
| | - Jessica C Jang
- School of Medicine, University of California, Riverside, CA, USA
| | - Maham Rais
- School of Medicine, University of California, Riverside, CA, USA
| | - Meera G Nair
- School of Medicine, University of California, Riverside, CA, USA
| | - Ilhem Messaoudi
- School of Medicine, University of California, Riverside, CA, USA
| | - Emma H Wilson
- School of Medicine, University of California, Riverside, CA, USA
| |
Collapse
|
78
|
Wei H, Wang F, Wang Y, Li T, Xiu P, Zhong J, Sun X, Li J. Verteporfin suppresses cell survival, angiogenesis and vasculogenic mimicry of pancreatic ductal adenocarcinoma via disrupting the YAP-TEAD complex. Cancer Sci 2017; 108:478-487. [PMID: 28002618 PMCID: PMC5378285 DOI: 10.1111/cas.13138] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 12/07/2016] [Accepted: 12/17/2016] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human malignancies. The Yes-associated protein-1 (YAP) plays a critical role in cell proliferation, apoptosis and angiogenesis. Verteporfin is a photosensitizer used in photodynamic therapy and also a small molecular inhibitor of the Hippo-YAP pathway. However, little is known about whether verteporfin could inhibit YAP activity in PDAC cells. Our present results showed that verteporfin suppressed the proliferation of human PDAC PANC-1 and SW1990 cells by arresting cells at the G1 phase, and inducing apoptosis in dose- and time-dependent manners. Verteporfin also inhibited the tumor growth on the PDAC xenograft model. Treatment with verteporfin led to downregulation of cyclinD1 and cyclinE1, modulation of Bcl-2 family proteins and activation of PARP. In addition, verteporfin exhibited an inhibitory effect on angiogenesis and vasculogenic mimicry via suppressing Ang2, MMP2, VE-cadherin, and α-SMA expression in vitro and in vivo. Mechanism studies demonstrated that verteporfin impaired YAP and TEAD interaction to suppress the expression of targeted genes. Our results provide a foundation for repurposing verteporfin as a promising anti-tumor drug in the treatment of pancreatic cancer by targeting the Hippo pathway.
Collapse
Affiliation(s)
- Honglong Wei
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Fuhai Wang
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Yong Wang
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Tao Li
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Peng Xiu
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Jingtao Zhong
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| | - Xueying Sun
- Department of Molecular Medicine and PathologyFaculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Jie Li
- Department of General SurgeryQianfoshan HospitalShandong UniversityJinanChina
| |
Collapse
|
79
|
Elevation of YAP promotes the epithelial-mesenchymal transition and tumor aggressiveness in colorectal cancer. Exp Cell Res 2017; 350:218-225. [DOI: 10.1016/j.yexcr.2016.11.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/23/2016] [Accepted: 11/29/2016] [Indexed: 02/08/2023]
|
80
|
Janse van Rensburg HJ, Yang X. The roles of the Hippo pathway in cancer metastasis. Cell Signal 2016; 28:1761-72. [DOI: 10.1016/j.cellsig.2016.08.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/07/2016] [Accepted: 08/08/2016] [Indexed: 01/08/2023]
|
81
|
Zanconato F, Cordenonsi M, Piccolo S. YAP/TAZ at the Roots of Cancer. Cancer Cell 2016; 29:783-803. [PMID: 27300434 PMCID: PMC6186419 DOI: 10.1016/j.ccell.2016.05.005] [Citation(s) in RCA: 1321] [Impact Index Per Article: 165.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/26/2016] [Accepted: 05/16/2016] [Indexed: 02/06/2023]
Abstract
YAP and TAZ are highly related transcriptional regulators pervasively activated in human malignancies. Recent work indicates that, remarkably, YAP/TAZ are essential for cancer initiation or growth of most solid tumors. Their activation induces cancer stem cell attributes, proliferation, chemoresistance, and metastasis. YAP/TAZ are sensors of the structural and mechanical features of the cell microenvironment. A number of cancer-associated extrinsic and intrinsic cues conspire to overrule the YAP-inhibiting microenvironment of normal tissues, including changes in mechanotransduction, inflammation, oncogenic signaling, and regulation of the Hippo pathway. Addiction to YAP/TAZ thus potentially represents a central cancer vulnerability that may be exploited therapeutically.
Collapse
Affiliation(s)
- Francesca Zanconato
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Michelangelo Cordenonsi
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy.
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy.
| |
Collapse
|
82
|
Abstract
In this review, Meng et al. focus on recent developments in our understanding of the molecular actions of the core Hippo kinase cascade and discuss key open questions in Hippo pathway regulation and function. The Hippo pathway was initially identified in Drosophila melanogaster screens for tissue growth two decades ago and has been a subject extensively studied in both Drosophila and mammals in the last several years. The core of the Hippo pathway consists of a kinase cascade, transcription coactivators, and DNA-binding partners. Recent studies have expanded the Hippo pathway as a complex signaling network with >30 components. This pathway is regulated by intrinsic cell machineries, such as cell–cell contact, cell polarity, and actin cytoskeleton, as well as a wide range of signals, including cellular energy status, mechanical cues, and hormonal signals that act through G-protein-coupled receptors. The major functions of the Hippo pathway have been defined to restrict tissue growth in adults and modulate cell proliferation, differentiation, and migration in developing organs. Furthermore, dysregulation of the Hippo pathway leads to aberrant cell growth and neoplasia. In this review, we focus on recent developments in our understanding of the molecular actions of the core Hippo kinase cascade and discuss key open questions in the regulation and function of the Hippo pathway.
Collapse
Affiliation(s)
- Zhipeng Meng
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Toshiro Moroishi
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
83
|
Lei L, Wu J, Gu D, Liu H, Wang S. CIZ1 interacts with YAP and activates its transcriptional activity in hepatocellular carcinoma cells. Tumour Biol 2016; 37:11073-9. [PMID: 26906552 DOI: 10.1007/s13277-016-4866-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/13/2016] [Indexed: 10/22/2022] Open
Abstract
Dysregulation of Hippo-Yes-associate protein (YAP) signaling has important roles in the tumorigenesis of hepatocellular carcinoma (HCC). Our previous studies have shown that Cip1 interacting zinc finger protein 1 (CIZ1) activated YAP signaling in the HCC cells and promoted the growth and migration of cancer cells. However, the mechanisms for the activation of YAP signaling by CIZ1 are unknown. In this study, it was found that CIZ1 interacted with the transcriptional factor YAP in HCC cells. The nuclear matrix anchor domain of CIZ1 is responsible for its interaction with YAP. Moreover, CIZ1 enhanced the interaction between YAP and TEAD. Knocking down the expression of CIZ1 impaired the transcriptional activity as well as the biological functions of YAP. Taken together, our study demonstrated that CIZ1 is a positive regulator of YAP signaling, and CIZ1 might be a therapeutic target for HCC.
Collapse
Affiliation(s)
- Liu Lei
- Department of Hepatobiliary & Pancreatic Surgery, Huai'an First People's Hospital, Nanjing Medical University, 6th of West Beijing Road, Huai'an, Jiangsu Province, 223300, People's Republic of China
| | - Jinsheng Wu
- Department of Hepatobiliary & Pancreatic Surgery, Huai'an First People's Hospital, Nanjing Medical University, 6th of West Beijing Road, Huai'an, Jiangsu Province, 223300, People's Republic of China
| | - Dianhua Gu
- Department of Hepatobiliary & Pancreatic Surgery, Huai'an First People's Hospital, Nanjing Medical University, 6th of West Beijing Road, Huai'an, Jiangsu Province, 223300, People's Republic of China
| | - Hui Liu
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| | - Shaochuang Wang
- Department of Hepatobiliary & Pancreatic Surgery, Huai'an First People's Hospital, Nanjing Medical University, 6th of West Beijing Road, Huai'an, Jiangsu Province, 223300, People's Republic of China.
| |
Collapse
|