101
|
YAP1 mediates survival of ALK-rearranged lung cancer cells treated with alectinib via pro-apoptotic protein regulation. Nat Commun 2020; 11:74. [PMID: 31900393 PMCID: PMC6941996 DOI: 10.1038/s41467-019-13771-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Despite the promising clinical efficacy of the second-generation anaplastic lymphoma kinase (ALK) inhibitor alectinib in patients with ALK-rearranged lung cancer, some tumor cells survive and eventually relapse, which may be an obstacle to achieving a cure. Limited information is currently available on the mechanisms underlying the initial survival of tumor cells against alectinib. Using patient-derived cell line models, we herein demonstrate that cancer cells survive a treatment with alectinib by activating Yes-associated protein 1 (YAP1), which mediates the expression of the anti-apoptosis factors Mcl-1 and Bcl-xL, and combinatorial inhibition against both YAP1 and ALK provides a longer tumor remission in ALK-rearranged xenografts when compared with alectinib monotherapy. These results suggest that the inhibition of YAP1 is a candidate for combinatorial therapy with ALK inhibitors to achieve complete remission in patients with ALK-rearranged lung cancer.
Collapse
|
102
|
Vargas RE, Duong VT, Han H, Ta AP, Chen Y, Zhao S, Yang B, Seo G, Chuc K, Oh S, El Ali A, Razorenova OV, Chen J, Luo R, Li X, Wang W. Elucidation of WW domain ligand binding specificities in the Hippo pathway reveals STXBP4 as YAP inhibitor. EMBO J 2020; 39:e102406. [PMID: 31782549 PMCID: PMC6939200 DOI: 10.15252/embj.2019102406] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
The Hippo pathway, which plays a critical role in organ size control and cancer, features numerous WW domain-based protein-protein interactions. However, ~100 WW domains and 2,000 PY motif-containing peptide ligands are found in the human proteome, raising a "WW-PY" binding specificity issue in the Hippo pathway. In this study, we have established the WW domain binding specificity for Hippo pathway components and uncovered a unique amino acid sequence required for it. By using this criterion, we have identified a WW domain-containing protein, STXBP4, as a negative regulator of YAP. Mechanistically, STXBP4 assembles a protein complex comprising α-catenin and a group of Hippo PY motif-containing components/regulators to inhibit YAP, a process that is regulated by actin cytoskeleton tension. Interestingly, STXBP4 is a potential tumor suppressor for human kidney cancer, whose downregulation is correlated with YAP activation in clear cell renal cell carcinoma. Taken together, our study not only elucidates the WW domain binding specificity for the Hippo pathway, but also reveals STXBP4 as a player in actin cytoskeleton tension-mediated Hippo pathway regulation.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Hippo Signaling Pathway
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Prognosis
- Protein Binding
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Signal Transduction
- Survival Rate
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Tumor Cells, Cultured
- Vesicular Transport Proteins/genetics
- Vesicular Transport Proteins/metabolism
- WW Domains
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Rebecca E Vargas
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Vy Thuy Duong
- Department of ChemistryUniversity of California, IrvineIrvineCAUSA
| | - Han Han
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Albert Paul Ta
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Yuxuan Chen
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Shiji Zhao
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Bing Yang
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Gayoung Seo
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Kimberly Chuc
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Sunwoo Oh
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Amal El Ali
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCAUSA
| | - Olga V Razorenova
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCAUSA
| | - Junjie Chen
- Department of Experimental Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ray Luo
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of California, IrvineIrvineCAUSA
- Department of Materials Science and EngineeringUniversity of California, IrvineIrvineCAUSA
- Department of Biomedical EngineeringUniversity of California, IrvineIrvineCAUSA
| | - Xu Li
- School of Life SciencesWestlake UniversityHangzhouZhejiangChina
| | - Wenqi Wang
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| |
Collapse
|
103
|
Abstract
The Hippo pathway and its downstream effectors, the transcriptional co-activators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), regulate organ growth and cell plasticity during animal development and regeneration. Remarkably, experimental activation of YAP/TAZ in the mouse can promote regeneration in organs with poor or compromised regenerative capacity, such as the adult heart and the liver and intestine of old or diseased mice. However, therapeutic YAP/TAZ activation may cause serious side effects. Most notably, YAP/TAZ are hyperactivated in human cancers, and prolonged activation of YAP/TAZ triggers cancer development in mice. Thus, can the power of YAP/TAZ to promote regeneration be harnessed in a safe way? Here, we review the role of Hippo signalling in animal regeneration, examine the promises and risks of YAP/TAZ activation for regenerative medicine and discuss strategies to activate YAP/TAZ for regenerative therapy while minimizing adverse side effects.
Collapse
|
104
|
Santos-de-Frutos K, Segrelles C, Lorz C. Hippo Pathway and YAP Signaling Alterations in Squamous Cancer of the Head and Neck. J Clin Med 2019; 8:jcm8122131. [PMID: 31817001 PMCID: PMC6947155 DOI: 10.3390/jcm8122131] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022] Open
Abstract
Head and neck cancer affects the upper aerodigestive tract and is the sixth leading cancer worldwide by incidence and the seventh by cause of death. Despite significant advances in surgery and chemotherapy, molecularly targeted therapeutic options for this type of cancer are scarce and long term survival rates remain low. Recently, comprehensive genomic studies have highlighted the most commonly altered genes and signaling pathways in this cancer. The Hippo-YAP pathway has been identified as a key oncogenic pathway in multiple tumors. Expression of genes controlled by the Hippo downstream transcriptional coactivators YAP (Yes-associated protein 1) and TAZ (WWTR1, WW domain containing transcription regulator 1) is widely deregulated in human cancer including head and neck squamous cell carcinoma (HNSCC). Interestingly, YAP/TAZ signaling might not be as essential for the normal homeostasis of adult tissues as for oncogenic growth, altogether making the pathway an amenable therapeutic target in cancer. Recent advances in the role of Hippo-YAP pathway in HNSCC have provided evidence that genetic alterations frequent in this type of cancer such as PIK3CA (phosphatidylinositide 3-kinase catalytic subunit alpha) overexpression or FAT1 (FAT atypical cadherin 1) functional loss can result in YAP activation. We discuss current therapeutic options targeting this pathway which are currently in use for other tumor types.
Collapse
Affiliation(s)
- Karla Santos-de-Frutos
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
| | - Carmen Segrelles
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ave Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Corina Lorz
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ave Monforte de Lemos 3-5, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-91-4962-521; Fax: +34-91-3466-484
| |
Collapse
|
105
|
Sahu MR, Mondal AC. The emerging role of Hippo signaling in neurodegeneration. J Neurosci Res 2019; 98:796-814. [PMID: 31705587 DOI: 10.1002/jnr.24551] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022]
Abstract
Neurodegeneration refers to the complex process of progressive degeneration or neuronal apoptosis leading to a set of incurable and debilitating conditions. Physiologically, apoptosis is important in proper growth and development. However, aberrant and unrestricted apoptosis can lead to a variety of degenerative conditions including neurodegenerative diseases. Although dysregulated apoptosis has been implicated in various neurodegenerative disorders, the triggers and molecular mechanisms underlying such untimely and faulty apoptosis are still unknown. Hippo signaling pathway is one such apoptosis-regulating mechanism that has remained evolutionarily conserved from Drosophila to mammals. This pathway has gained a lot of attention for its tumor-suppressing task, but recent studies have emphasized the soaring role of this pathway in inflaming neurodegeneration. In addition, strategies promoting inactivation of this pathway have aided in the rescue of neurons from anomalous apoptosis. So, a thorough understanding of the relationship between the Hippo pathway and neurodegeneration may serve as a guide for the development of therapy for various degenerative diseases. The current review focuses on the mechanism of the Hippo signaling pathway, its upstream and downstream regulatory molecules, and its role in the genesis of numerous neurodegenerative diseases. The recent efforts employing the Hippo pathway components as targets for checking neurodegeneration have also been highlighted.
Collapse
Affiliation(s)
- Manas Ranjan Sahu
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
106
|
Raj N, Bam R. Reciprocal Crosstalk Between YAP1/Hippo Pathway and the p53 Family Proteins: Mechanisms and Outcomes in Cancer. Front Cell Dev Biol 2019; 7:159. [PMID: 31448276 PMCID: PMC6695833 DOI: 10.3389/fcell.2019.00159] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
The YAP1/Hippo and p53 pathways are critical protectors of genome integrity in response to DNA damage. Together, these pathways secure cellular adaptation and maintain overall tissue integrity through transcriptional re-programing downstream of various environmental and biological cues generated during normal tissue growth, cell proliferation, and apoptosis. Genetic perturbations in YAP1/Hippo and p53 pathways are known to contribute to the cells’ ability to turn rogue and initiate tumorigenesis. The Hippo and p53 pathways cooperate on many levels and are closely coordinated through multiple molecular components of their signaling pathways. Several functional and physical interactions have been reported to occur between YAP1/Hippo pathway components and the three p53 family members, p53, p63, and p73. Primarily, functional status of p53 family proteins dictates the subcellular localization, protein stability and transcriptional activity of the core component of the Hippo pathway, Yes-associated protein 1 (YAP1). In this review, we dissect the critical points of crosstalk between the YAP1/Hippo pathway components, with a focus on YAP1, and the p53 tumor suppressor protein family. For each p53 family member, we discuss the biological implications of their interaction with Hippo pathway components in determining cell fate under the conditions of tissue homeostasis and cancer pathogenesis.
Collapse
Affiliation(s)
- Nitin Raj
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States
| | - Rakesh Bam
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
107
|
Xia Y, Pfeifer CR, Zhu K, Irianto J, Liu D, Pannell K, Chen EJ, Dooling LJ, Tobin MP, Wang M, Ivanovska IL, Smith LR, Greenberg RA, Discher DE. Rescue of DNA damage after constricted migration reveals a mechano-regulated threshold for cell cycle. J Cell Biol 2019; 218:2545-2563. [PMID: 31239284 PMCID: PMC6683732 DOI: 10.1083/jcb.201811100] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/19/2019] [Accepted: 06/04/2019] [Indexed: 12/24/2022] Open
Abstract
Migration through 3D constrictions can cause nuclear rupture and mislocalization of nuclear proteins, but damage to DNA remains uncertain, as does any effect on cell cycle. Here, myosin II inhibition rescues rupture and partially rescues the DNA damage marker γH2AX, but an apparent block in cell cycle appears unaffected. Co-overexpression of multiple DNA repair factors or antioxidant inhibition of break formation also exert partial effects, independently of rupture. Combined treatments completely rescue cell cycle suppression by DNA damage, revealing a sigmoidal dependence of cell cycle on excess DNA damage. Migration through custom-etched pores yields the same damage threshold, with ∼4-µm pores causing intermediate levels of both damage and cell cycle suppression. High curvature imposed rapidly by pores or probes or else by small micronuclei consistently associates nuclear rupture with dilution of stiff lamin-B filaments, loss of repair factors, and entry from cytoplasm of chromatin-binding cGAS (cyclic GMP-AMP synthase). The cell cycle block caused by constricted migration is nonetheless reversible, with a potential for DNA misrepair and genome variation.
Collapse
Affiliation(s)
- Yuntao Xia
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Charlotte R. Pfeifer
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA,Department of Physics and Astronomy, Graduate Group, University of Pennsylvania, Philadelphia, PA
| | - Kuangzheng Zhu
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Jerome Irianto
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Dazhen Liu
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Kalia Pannell
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Emily J. Chen
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Lawrence J. Dooling
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Michael P. Tobin
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Mai Wang
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Irena L. Ivanovska
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Lucas R. Smith
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA
| | - Roger A. Greenberg
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dennis E. Discher
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA,Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA,Department of Physics and Astronomy, Graduate Group, University of Pennsylvania, Philadelphia, PA,Correspondence to D.E. Discher:
| |
Collapse
|
108
|
Transformation of normal cells by aberrant activation of YAP via cMyc with TEAD. Sci Rep 2019; 9:10933. [PMID: 31358774 PMCID: PMC6662713 DOI: 10.1038/s41598-019-47301-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/12/2019] [Indexed: 01/09/2023] Open
Abstract
YAP (also known as YAP1 or YAP65) is a transcriptional coactivator that interacts with a number of transcription factors including RUNX and TEAD and plays a pivotal role in controlling cell growth. YAP is classified as a proto-oncogene. However, the mechanism by which activated YAP induces cancerous changes is not well known. Here we demonstrate that overexpression of YAP in NIH3T3 cells was sufficient for inducing tumorigenic transformation of cells. Mechanistically, YAP exerts its function in cooperation with the TEAD transcription factor. Our data also show that cMYC is a critical factor that acts downstream of the YAP/TEAD complex. Furthermore, we also found that aberrant activation of YAP is sufficient to drive tumorigenic transformation of non-immortalized mouse embryonic fibroblasts. Together our data indicate that YAP can be categorized as a new type of proto-oncogene distinct from typical oncogenes, such as H-RAS, whose expression in non-immortalized cells is tightly linked to senescence.
Collapse
|
109
|
Li Y, Kong F, Jin C, Hu E, Shao Q, Liu J, He D, Xiao X. The expression of S100A8/S100A9 is inducible and regulated by the Hippo/YAP pathway in squamous cell carcinomas. BMC Cancer 2019; 19:597. [PMID: 31208368 PMCID: PMC6580480 DOI: 10.1186/s12885-019-5784-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Background S100A8 and S100A9, two heterodimer-forming members of the S100 family, aberrantly express in a variety of cancer types. However, little is known about the mechanism that regulates S100A8/S100A9 co-expression in cancer cells. Methods The expression level of S100A8/S100A9 measured in three squamous cell carcinomas (SCC) cell lines and their corresponding xenografts, as well as in 257 SCC tissues. The correlation between S100A8/S100A9, Hippo pathway and F-actin cytoskeleton were evaluated using western blot, qPCR, ChIP and Immunofluorescence staining tests. IncuCyte ZOOM long time live cell image monitoring system, qPCR and Flow Cytometry measured the effects of S100A8/S100A9 and YAP on cell proliferation, cell differentiation and apoptosis. Results Here, we report that through activation of the Hippo pathway, suspension and dense culture significantly induce S100A8/S100A9 co-expression and co-localization in SCC cells. Furthermore, these expressional characteristics of S100A8/S100A9 also observed in the xenografts derived from the corresponding SCC cells. Importantly, Co-expression of S100A8/S100A9 detected in 257 SCC specimens derived from five types of SCC tissues. Activation of the Hippo pathway by overexpression of Lats1, knockdown of YAP, as well as disruption of F-actin indeed obviously results in S100A8/S100A9 co-expression in attached SCC cells. Conversely, inhibition of the Hippo pathway leads to S100A8/S100A9 co-expression in a manner opposite of cell suspension and dense. In addition, we found that TEAD1 is required for YAP-induced S100A8/S100A9-expressions. The functional studies provide evidence that knockdown of S100A8/S100A9 together significantly inhibit cell proliferation but promote squamous differentiation and apoptosis. Conclusions Our findings demonstrate for the first time that the expression of S100A8/S100A9 is inducible by changes of cell shape and density through activation of the Hippo pathway in SCC cells. Induced S100A8/S100A9 promoted cell proliferation, inhibit cell differentiation and apoptosis. Electronic supplementary material The online version of this article (10.1186/s12885-019-5784-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunguang Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Fei Kong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Chang Jin
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Enze Hu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Qirui Shao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Jin Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Dacheng He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China
| | - Xueyuan Xiao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, 19th, Beijing, 100875, China.
| |
Collapse
|
110
|
Snigdha K, Gangwani KS, Lapalikar GV, Singh A, Kango-Singh M. Hippo Signaling in Cancer: Lessons From Drosophila Models. Front Cell Dev Biol 2019; 7:85. [PMID: 31231648 PMCID: PMC6558396 DOI: 10.3389/fcell.2019.00085] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/03/2019] [Indexed: 12/19/2022] Open
Abstract
Hippo pathway was initially identified through genetic screens for genes regulating organ size in fruitflies. Recent studies have highlighted the role of Hippo signaling as a key regulator of homeostasis, and in tumorigenesis. Hippo pathway is comprised of genes that act as tumor suppressor genes like hippo (hpo) and warts (wts), and oncogenes like yorkie (yki). YAP and TAZ are two related mammalian homologs of Drosophila Yki that act as effectors of the Hippo pathway. Hippo signaling deficiency can cause YAP- or TAZ-dependent oncogene addiction for cancer cells. YAP and TAZ are often activated in human malignant cancers. These transcriptional regulators may initiate tumorigenic changes in solid tumors by inducing cancer stem cells and proliferation, culminating in metastasis and chemo-resistance. Given the complex mechanisms (e.g., of the cancer microenvironment, and the extrinsic and intrinsic cues) that overpower YAP/TAZ inhibition, the molecular roles of the Hippo pathway in tumor growth and progression remain poorly defined. Here we review recent findings from studies in whole animal model organism like Drosophila on the role of Hippo signaling regarding its connection to inflammation, tumor microenvironment, and other oncogenic signaling in cancer growth and progression.
Collapse
Affiliation(s)
- Kirti Snigdha
- Department of Biology, University of Dayton, Dayton, OH, United States
| | | | - Gauri Vijay Lapalikar
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, United States.,Pre-Medical Programs, University of Dayton, Dayton, OH, United States.,Center for Tissue Regeneration and Engineering at Dayton, University of Dayton, Dayton, OH, United States.,Integrated Science and Engineering Center, University of Dayton, Dayton, OH, United States
| | - Madhuri Kango-Singh
- Department of Biology, University of Dayton, Dayton, OH, United States.,Pre-Medical Programs, University of Dayton, Dayton, OH, United States.,Center for Tissue Regeneration and Engineering at Dayton, University of Dayton, Dayton, OH, United States.,Integrated Science and Engineering Center, University of Dayton, Dayton, OH, United States
| |
Collapse
|
111
|
Shreberk-Shaked M, Oren M. New insights into YAP/TAZ nucleo-cytoplasmic shuttling: new cancer therapeutic opportunities? Mol Oncol 2019; 13:1335-1341. [PMID: 31050214 PMCID: PMC6547617 DOI: 10.1002/1878-0261.12498] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Yes‐associated protein (YAP) and transcriptional co‐activator with PDZ‐binding motif (TAZ), the main effectors of the Hippo pathway, are emerging as important players in cancer biology and therapy response. The intracellular localization of YAP/TAZ is a key determinant in the regulation of their activity and their roles in signal transduction. This is particularly relevant for cancer: Aberrant nuclear localization of YAP and TAZ has been observed in numerous human cancers and may therefore represent an attractive target for cancer therapy. In this review, we describe the mechanisms that regulate the nucleo‐cytoplasmic shuttling of YAP/TAZ and their implications for cancer, and discuss how the new insights about this process may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
112
|
Chromatin dynamics underlying the precise regeneration of a vertebrate limb - Epigenetic regulation and cellular memory. Semin Cell Dev Biol 2019; 97:16-25. [PMID: 30991117 DOI: 10.1016/j.semcdb.2019.04.006] [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: 12/14/2018] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
Abstract
Wound healing, tissue regeneration, and organ regrowth are all regeneration phenomena observed in vertebrates after an injury. However, the ability to regenerate differs greatly among species. Mammals can undergo wound healing and tissue regeneration, but cannot regenerate an organ; for example, they cannot regrow an amputated limb. In contrast, amphibians and fish have much higher capabilities for organ-level regeneration. In addition to medical studies and those in conventional mammalian models such as mice, studies in amphibians and fish have revealed essential factors for and mechanisms of regeneration, including the regrowth of a limb, tail, or fin. However, the molecular nature of the cellular memory needed to precisely generate a new appendage from an amputation site is not fully understood. Recent reports have indicated that organ regeneration is closely related to epigenetic regulation. For example, the methylation status of genomic DNA is related to the expression of regeneration-related genes, and histone-modification enzymes are required to control the chromatin dynamics for regeneration. A proposed mechanism of cellular memory involving an inheritable system of epigenetic modification led us to hypothesize that epigenetic regulation forms the basis for cellular memory in organ regeneration. Here we summarize the current understanding of the role of epigenetic regulation in organ regeneration and discuss the relationship between organ regeneration and epigenetic memory.
Collapse
|
113
|
Rognoni E, Walko G. The Roles of YAP/TAZ and the Hippo Pathway in Healthy and Diseased Skin. Cells 2019; 8:cells8050411. [PMID: 31058846 PMCID: PMC6562585 DOI: 10.3390/cells8050411] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Skin is the largest organ of the human body. Its architecture and physiological functions depend on diverse populations of epidermal cells and dermal fibroblasts. Reciprocal communication between the epidermis and dermis plays a key role in skin development, homeostasis and repair. While several stem cell populations have been identified in the epidermis with distinct locations and functions, there is additional heterogeneity within the mesenchymal cells of the dermis. Here, we discuss the current knowledge of how the Hippo pathway and its downstream effectors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) contribute to the maintenance, activation and coordination of the epidermal and dermal cell populations during development, homeostasis, wound healing and cancer.
Collapse
Affiliation(s)
- Emanuel Rognoni
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Gernot Walko
- Department of Biology and Biochemistry & Centre for Therapeutic Innovation, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
114
|
Gupta R, Bhatt LK, Johnston TP, Prabhavalkar KS. Colon cancer stem cells: Potential target for the treatment of colorectal cancer. Cancer Biol Ther 2019; 20:1068-1082. [PMID: 31050577 DOI: 10.1080/15384047.2019.1599660] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite incessant research, colon cancer still is one of the most common causes of fatalities in both men and women worldwide. Also, nearly 50% of patients with colorectal cancer show tumor recurrence. Recent investigations have highlighted the involvement of colon cancer stem cells (CCSCs) in cancer relapse and chemoresistance. CCSCs deliver a significant protumorigenic niche through persistent overexpression of self-renewal capabilities. Moreover, CSCs cross network with stromal cells, immune infiltrates, and cyotokine-chemokine, which potentiate their aggressive proliferative potential. Targeting CCSCs through small molecule inhibitors, miRNAs, and monoclonal antibodies (mAbs) in in vivo studies has generated compelling evidence for the effectiveness of these various treatments. This review effectively compiles the role of CCSC surface markers and dysregulated and/or upregulated pathways in the pathogenesis of colorectal cancer that can be used to target CCSCs for effective colorectal cancer treatment.
Collapse
Affiliation(s)
- Riya Gupta
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Lokesh Kumar Bhatt
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Thomas P Johnston
- b Division of Pharmacology and Pharmaceutical Sciences , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Kedar S Prabhavalkar
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| |
Collapse
|
115
|
García-Gil A, Galán-Enríquez CS, Pérez-López A, Nava P, Alpuche-Aranda C, Ortiz-Navarrete V. SopB activates the Akt-YAP pathway to promote Salmonella survival within B cells. Virulence 2019; 9:1390-1402. [PMID: 30103648 PMCID: PMC6177241 DOI: 10.1080/21505594.2018.1509664] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
B cells are a target of Salmonella infection, allowing bacteria survival without inducing pyroptosis. This event is due to downregulation of Nlrc4 expression and lack of inflammasome complex activation, which impairs the secretion of IL-1β. YAP phosphorylation is required for downregulation of Nlrc4 in B cells during Salmonella infection; however, the microorganism’s mechanisms underlying the inhibition of the NLRC4 inflammasome in B cells are not fully understood. Our findings demonstrate that the Salmonella effector SopB triggers a signaling cascade involving PI3K, PDK1 and mTORC2 that activates Akt with consequent phosphorylation of YAP. When we deleted sopB in Salmonella, infected B cells that lack Rictor, or inhibited the signaling cascade using a pharmacological approach, we were able to restore the function of the NLRC4 inflammasome in B cells and the ability to control the infection. Furthermore, B cells from infected mice exhibited activation of Akt and YAP phosphorylation, suggesting that Salmonella also triggers this pathway in vivo. In summary, our data demonstrate that the Salmonella effector inositide phosphate phosphatase SopB triggers the PI3K-Akt-YAP pathway to inhibit the NLRC4 inflammasome in B cells. This study provides further evidence that Salmonella triggers cellular mechanisms in B lymphocytes to manipulate the host environment by turning it into a survival niche to establish a successful infection.
Collapse
Affiliation(s)
- Abraham García-Gil
- a Departamento de Biomedicina Molecular , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| | - Carlos Samuel Galán-Enríquez
- a Departamento de Biomedicina Molecular , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| | - Araceli Pérez-López
- b Department of Pediatrics , University of California San Diego , San Diego , CA , USA
| | - Porfirio Nava
- c Departamento de Fisiología , Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| | - Celia Alpuche-Aranda
- d Centro de Investigación Sobre Enfermedades Infecciosa , Instituto Nacional de Salud Pública, SSA , Cuernavaca , México
| | - Vianney Ortiz-Navarrete
- a Departamento de Biomedicina Molecular , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| |
Collapse
|
116
|
Mechanotransduction and Cytoskeleton Remodeling Shaping YAP1 in Gastric Tumorigenesis. Int J Mol Sci 2019; 20:ijms20071576. [PMID: 30934860 PMCID: PMC6480114 DOI: 10.3390/ijms20071576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
The essential role of Hippo signaling pathway in cancer development has been elucidated by recent studies. In the gastrointestinal tissues, deregulation of the Hippo pathway is one of the most important driving events for tumorigenesis. It is widely known that Yes-associated protein 1 (YAP1) and WW domain that contain transcription regulator 1 (TAZ), two transcriptional co-activators with a PDZ-binding motif, function as critical effectors negatively regulated by the Hippo pathway. Previous studies indicate the involvement of YAP1/TAZ in mechanotransduction by crosstalking with the extracellular matrix (ECM) and the F-actin cytoskeleton associated signaling network. In gastric cancer (GC), YAP1/TAZ functions as an oncogene and transcriptionally promotes tumor formation by cooperating with TEAD transcription factors. Apart from the classic role of Hippo-YAP1 cascade, in this review, we summarize the current investigations to highlight the prominent role of YAP1/TAZ as a mechanical sensor and responder under mechanical stress and address its potential prognostic and therapeutic value in GC.
Collapse
|
117
|
Yeung YT, Guerrero-Castilla A, Cano M, Muñoz MF, Ayala A, Argüelles S. Dysregulation of the Hippo pathway signaling in aging and cancer. Pharmacol Res 2019; 143:151-165. [PMID: 30910741 DOI: 10.1016/j.phrs.2019.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/04/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
Human beings are facing emerging degenerative and cancer diseases, in large part, as a consequence of increased life expectancy. In the near future, researchers will have to put even more effort into fighting these new challenges, one of which will be prevention of cancer while continuing to improve the aging process through this increased life expectancy. In the last few decades, relevance of the Hippo pathway on cancer has become an important study since it is a major regulator of organ size control and proliferation. However, its deregulation can induce tumors throughout the body by regulating cell proliferation, disrupting cell polarity, releasing YAP and TAZ from the Scribble complexes and facilitating survival gene expression via activation of TEAD transcription factors. This pathway is also involved in some of the most important mechanisms that control the aging processes, such as the AMP-activated protein kinase and sirtuin pathways, along with autophagy and oxidative stress response/antioxidant defense. This could be the link between two tightly connected processes that could open a broader range of targeted molecular therapies to fight aging and cancer. Therefore, available knowledge of the processes involved in the Hippo pathway during aging and cancer must necessarily be well understood.
Collapse
Affiliation(s)
- Yiu To Yeung
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | | | - Mercedes Cano
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mario F Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Antonio Ayala
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
118
|
Callus BA, Finch-Edmondson ML, Fletcher S, Wilton SD. YAPping about and not forgetting TAZ. FEBS Lett 2019; 593:253-276. [PMID: 30570758 DOI: 10.1002/1873-3468.13318] [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] [Received: 10/24/2018] [Revised: 12/05/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022]
Abstract
The Hippo pathway has emerged as a major eukaryotic signalling pathway and is increasingly the subject of intense interest, as are the key effectors of canonical Hippo signalling, YES-associated protein (YAP) and TAZ. The Hippo pathway has key roles in diverse biological processes, including network signalling regulation, development, organ growth, tissue repair and regeneration, cancer, stem cell regulation and mechanotransduction. YAP and TAZ are multidomain proteins and function as transcriptional coactivators of key genes to evoke their biological effects. YAP and TAZ interact with numerous partners and their activities are controlled by a complex set of processes. This review provides an overview of Hippo signalling and its role in growth. In particular, the functional domains of YAP and TAZ and the complex mechanisms that regulate their protein stability and activity are discussed. Notably, the similarities and key differences are highlighted between the two paralogues including which partner proteins interact with which functional domains to regulate their activity.
Collapse
Affiliation(s)
| | - Megan L Finch-Edmondson
- Discipline of Child and Adolescent Health, Children's Hospital at Westmead Clinical School, University of Sydney Medical School, Australia.,Cerebral Palsy Alliance Research Institute, University of Sydney, Australia
| | - Sue Fletcher
- Centre for Comparative Genomics, Murdoch University, Australia.,Perron Institute for Neurological and Translational Research, Nedlands, Australia
| | - Steve D Wilton
- Centre for Comparative Genomics, Murdoch University, Australia.,Perron Institute for Neurological and Translational Research, Nedlands, Australia
| |
Collapse
|
119
|
Zhang S, Wei Q, Yang Y, Qin H, Li X, Cai S, Ma Y. Loss of Yes-associated Protein Represents an Aggressive Subtype of Colorectal Cancer. J Cancer 2019; 10:689-696. [PMID: 30719167 PMCID: PMC6360423 DOI: 10.7150/jca.28333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/27/2018] [Indexed: 01/15/2023] Open
Abstract
Background: Yes-associated protein (YAP) is a downstream effecter of Hippo signaling pathway, and has been linked to the initiation and development of colorectal cancer (CRC). However, the clinical significance of YAP in CRC remains controversial. This study was designed to investigate the clinical significance of YAP in CRC. Methods: We selected 206 eligible patients diagnosed with CRC from 2003 to 2007. Tissue microarray (TMA) blocks were made from 206 formalin-fixed paraffin-embedded CRC tissues and 158 corresponding normal colonic tissues. Using the TMA blocks, we performed immunohistochemical staining of YAP and assessed its expression status in different subcellular locations. The patients were divided into four groups according to the expression status of YAP in the cytoplasm and nucleus. Statistical analysis was performed to explore the correlation between YAP expression and clinicopathological features and overall survival (OS) in CRC patients. Results: Our results showed that both cytoplasmic YAP and nuclear YAP were overexpressed in CRC tissues compared to normal colonic tissues. Complete loss of YAP expression in CRC was significantly correlated with larger tumor size (p=0.023), proximal tumor location (p=0.038), higher tumor grade (p=0.022) and worse OS (p<0.001). Univariate and multivariate Cox regression analyses revealed that complete loss of YAP expression was an independent indicator of poor prognosis in CRC (p<0.001). Conclusions: Loss of YAP expression correlates with poor prognosis and may represent a subgroup with more aggressive biological features in CRC.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Huanlong Qin
- Department of GI Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
120
|
Deng LJ, Qi M, Peng QL, Chen MF, Qi Q, Zhang JY, Yao N, Huang MH, Li XB, Peng YH, Liu JS, Fu DR, Chen JX, Ye WC, Zhang DM. Arenobufagin induces MCF-7 cell apoptosis by promoting JNK-mediated multisite phosphorylation of Yes-associated protein. Cancer Cell Int 2018; 18:209. [PMID: 30574018 PMCID: PMC6299615 DOI: 10.1186/s12935-018-0706-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
Background It has been demonstrated that bufadienolides exert potent anti-cancer activity in various tumor types. However, the mechanisms that underlie their anti-cancer properties remain unclear. Yes-associated protein, a key effector of Hippo signaling, functions as a transcription coactivator, plays oncogenic and tumor suppressor roles under different conditions. Here, we report that arenobufagin (ABF), a representative bufadienolide, induced breast cancer MCF-7 cells to undergo apoptosis, which occurred through the JNK-mediated multisite phosphorylation of YAP. Methods Cytotoxicity was examined using an MTT assay. ABF-induced apoptosis was measured with a TUNEL assay and Annexin V-FITC/PI double staining assay. Western blotting, immunofluorescence, qRT-PCR and coimmunoprecipitation were employed to assess the expression levels of the indicated molecules. Lose-of-function experiments were carried out with siRNA transfection and pharmacological inhibitors. ABF-induced phosphopeptides were enriched with Ti4+-IMAC chromatography and further subjected to reverse-phase nano-LC–MS/MS analysis. Results ABF significantly reduced the viability of MCF-7 cells and increased the percentage of early and late apoptotic cells in a concentration- and time-dependent manner. Following ABF treatment, YAP accumulated in the nucleus and bound to p73, which enhanced the transcription of the pro-apoptotic genes Bax and p53AIP1. YAP knock-down significantly attenuated ABF-induced apoptotic cell death. Importantly, we found that the mobility shift of YAP was derived from its phosphorylation at multiple sites, including Tyr357. Moreover, mass spectrometry analysis identified 19 potential phosphorylation sites in YAP, with a distribution of 14 phosphoserine and 5 phosphothreonine residues. Furthermore, we found that the JNK inhibitor SP600125 completely diminished the mobility shift of YAP and its phosphorylation at Tyr357, the binding of YAP and p73, the transcription of Bax and p53AIP1 as well as the apoptosis induced by ABF. These data indicate that ABF induced YAP multisite phosphorylation, which was associated with p73 binding, and that apoptosis was mediated by the JNK signaling pathway. Conclusions Our data demonstrate that ABF suppresses MCF-7 breast cancer proliferation by triggering the pro-apoptotic activity of YAP, which is mediated by JNK signaling-induced YAP multisite phosphorylation as well as its association with p73. The present work not only provides additional information on the use of ABF as an anti-breast cancer drug, but also offers evidence that the induction of the tumor suppressor role of YAP may be a therapeutic strategy. Electronic supplementary material The online version of this article (10.1186/s12935-018-0706-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Juan Deng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,2Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Ming Qi
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Qun-Long Peng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Min-Feng Chen
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Qi Qi
- 4Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Jia-Yan Zhang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Nan Yao
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Mao-Hua Huang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Xiao-Bo Li
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Yin-Hui Peng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Jun-Shan Liu
- 5School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515 People's Republic of China
| | - Deng-Rui Fu
- Guangzhou Yucai Middle School, Fujin Road 2#, Dongshan District, Guangzhou, China
| | - Jia-Xu Chen
- 2Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Wen-Cai Ye
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Dong-Mei Zhang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| |
Collapse
|
121
|
Jamous A, Salah Z. WW-Domain Containing Protein Roles in Breast Tumorigenesis. Front Oncol 2018; 8:580. [PMID: 30619734 PMCID: PMC6300493 DOI: 10.3389/fonc.2018.00580] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Protein-protein interactions are key factors in executing protein function. These interactions are mediated through different protein domains or modules. An important domain found in many different types of proteins is WW domain. WW domain-containing proteins were shown to be involved in many human diseases including cancer. Some of these proteins function as either tumor suppressor genes or oncogenes, while others show dual identity. Some of these proteins act on their own and alter the function(s) of specific or multiple proteins implicated in cancer, others interact with their partners to compose WW domain modular pathway. In this review, we discuss the role of WW domain-containing proteins in breast tumorigenesis. We give examples of specific WW domain containing proteins that play roles in breast tumorigenesis and explain the mechanisms through which these proteins lead to breast cancer initiation and progression. We discuss also the possibility of using these proteins as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Abrar Jamous
- Al Quds-Bard College for Arts and Sciences, Al Quds University, Abu Dis, Palestine
| | - Zaidoun Salah
- Al Quds-Bard College for Arts and Sciences, Al Quds University, Abu Dis, Palestine
| |
Collapse
|
122
|
The Ambivalent Function of YAP in Apoptosis and Cancer. Int J Mol Sci 2018; 19:ijms19123770. [PMID: 30486435 PMCID: PMC6321280 DOI: 10.3390/ijms19123770] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/16/2018] [Accepted: 11/23/2018] [Indexed: 02/07/2023] Open
Abstract
Yes-associated protein, a core regulator of the Hippo-YAP signaling pathway, plays a vital role in inhibiting apoptosis. Thus, several studies and reviews suggest that yes-associated protein is a good target for treating cancer. Unfortunately, more and more evidence demonstrates that this protein is also an essential contributor of p73-mediated apoptosis. This questions the concept that yes-associated protein is always a good target for developing novel anti-cancer drugs. Thus, the aim of this review was to evaluate the clinical relevance of yes-associated protein for cancer pathophysiology. This review also summarized the molecules, processes and drugs, which regulate Hippo-YAP signaling and discusses their effect on apoptosis. In addition, issues are defined, which should be addressed in the future in order to provide a solid basis for targeting the Hippo-YAP signaling pathway in clinical trials.
Collapse
|
123
|
Targeting the Hippo Pathway for Breast Cancer Therapy. Cancers (Basel) 2018; 10:cancers10110422. [PMID: 30400599 PMCID: PMC6266939 DOI: 10.3390/cancers10110422] [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: 10/17/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) is one of the most prominent diseases in the world, and the treatments for BC have many limitations, such as resistance and a lack of reliable biomarkers. Currently the Hippo pathway is emerging as a tumor suppressor pathway with its four core components that regulate downstream transcriptional targets. In this review, we introduce the present targeted therapies of BC, and then discuss the roles of the Hippo pathway in BC. Finally, we summarize the evidence of the small molecule inhibitors that target the Hippo pathway, and then discuss the possibilities and future direction of the Hippo-targeted drugs for BC therapy.
Collapse
|
124
|
Affiliation(s)
- Eekhoon Jho
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| |
Collapse
|
125
|
Segrelles C, Paramio JM, Lorz C. The transcriptional co-activator YAP: A new player in head and neck cancer. Oral Oncol 2018; 86:25-32. [PMID: 30409308 DOI: 10.1016/j.oraloncology.2018.08.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/26/2018] [Indexed: 12/14/2022]
Abstract
The Hippo-YAP (Yes-associated protein) pathway is a key regulator of tissue growth, organ size and stem cell function. More recently, a fundamental role for this pathway has emerged in stem cell function and tumorigenesis. Activation of the transcriptional co-activator YAP promotes cell-contact independent proliferation, epithelial to mesenchymal transition (EMT), cancer stem cell features and drug resistance. In this review, we describe the main components of the pathway, the microenvironment and the cell-intrinsic cues governing its activation, the downstream players of the pathway and the biological implications of their activation in the context of cancer. We will focus on the existing knowledge of this pathway in head and neck squamous carcinoma (HNSCC), its clinical value in this type of cancer as a marker of poor prognosis and resistance to therapy, as well as the most encouraging therapeutic strategies targeting the pathway.
Collapse
Affiliation(s)
- Carmen Segrelles
- Molecular Oncology Unit, CIEMAT (ed 70A), Av. Complutense 40, 28040 Madrid, Spain; Molecular Oncology, University Hospital 12 de Octubre, Research Institute 12 de Octubre i+12, Av. Córdoba s/n, 28041 Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Jesús M Paramio
- Molecular Oncology Unit, CIEMAT (ed 70A), Av. Complutense 40, 28040 Madrid, Spain; Molecular Oncology, University Hospital 12 de Octubre, Research Institute 12 de Octubre i+12, Av. Córdoba s/n, 28041 Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Corina Lorz
- Molecular Oncology Unit, CIEMAT (ed 70A), Av. Complutense 40, 28040 Madrid, Spain; Molecular Oncology, University Hospital 12 de Octubre, Research Institute 12 de Octubre i+12, Av. Córdoba s/n, 28041 Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
126
|
Abstract
Hippo signaling plays critical roles in regulation of tissue homeostasis, organ size, and tumorigenesis by inhibiting YES-associated protein (YAP) and PDZ-binding protein TAZ through MST1/2 and LATS1/2 pathway. It is also engaged in cross-talk with various other signaling pathways, including WNT, BMPs, Notch, GPCRs, and Hedgehog to further modulate activities of YAP/TAZ. Because YAP and TAZ are transcriptional coactivators that lack DNA-binding activity, both proteins must interact with DNA-binding transcription factors to regulate target gene’s expression. To activate target genes involved in cell proliferation, TEAD family members are major DNA-binding partners of YAP/TAZ. Accordingly, YAP/TAZ were originally classified as oncogenes. However, YAP might also play tumor-suppressing role. For example, YAP can bind to DNA-binding tumor suppressors including RUNXs and p73. Thus, YAP might act either as an oncogene or tumor suppressor depending on its binding partners. Here, we summarize roles of YAP depending on its DNA-binding partners and discuss context-dependent functions of YAP/TAZ.
Collapse
Affiliation(s)
- Min-Kyu Kim
- Department of Biochemistry, College of Medicine, and Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Ju-Won Jang
- Department of Biochemistry, College of Medicine, and Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Suk-Chul Bae
- Department of Biochemistry, College of Medicine, and Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
127
|
Zhu JY, Lin S, Ye J. YAP and TAZ, the conductors that orchestrate eye development, homeostasis, and disease. J Cell Physiol 2018; 234:246-258. [PMID: 30094836 DOI: 10.1002/jcp.26870] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 12/25/2022]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators established as a nexus in numerous signaling pathways, notably in Hippo signaling. Previous research revealed multifarious function of YAP and TAZ in oncology and cardiovasology. Recently, the focus has been laid on their pivotal role in eye morphogenesis and homeostasis. In this review, we synthesize advances of YAP and TAZ function during eye development in different model organisms, introduce their function in different ocular tissues and eye diseases, and highlight the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Jing-Yi Zhu
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
128
|
YAP/TAZ upstream signals and downstream responses. Nat Cell Biol 2018; 20:888-899. [PMID: 30050119 DOI: 10.1038/s41556-018-0142-z] [Citation(s) in RCA: 679] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
Abstract
Cell behaviour is strongly influenced by physical, mechanical contacts between cells and their extracellular matrix. We review how the transcriptional regulators YAP and TAZ integrate mechanical cues with the response to soluble signals and metabolic pathways to control multiple aspects of cell behaviour, including proliferation, cell plasticity and stemness essential for tissue regeneration. Corruption of cell-environment interplay leads to aberrant YAP and TAZ activation that is instrumental for multiple diseases, including cancer.
Collapse
|
129
|
Lee M, Goraya N, Kim S, Cho SH. Hippo-yap signaling in ocular development and disease. Dev Dyn 2018; 247:794-806. [PMID: 29532607 PMCID: PMC5980750 DOI: 10.1002/dvdy.24628] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 12/17/2022] Open
Abstract
The Hippo-Yes associated protein (Yap) pathway plays an important role in organ size control by regulating cell proliferation, apoptosis, and stem cell renewal. Hippo-Yap signaling also functions at the level of cellular development in a variety of organs through its effects on cell cycle control, cell survival, cell polarity, and cell fate. Because of its important roles in normal development and homeostasis, abnormal regulation of this pathway has been shown to lead to pathological outcomes such as tissue overgrowth, tumor formation, and abnormal organogenesis, including ocular-specific disorders. In this review, we summarize how normal and perturbed control of Yap signaling is implicated in ocular development and disease Developmental Dynamics 247:794-806, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Matthew Lee
- Temple University Lewis Katz School of Medicine, Temple University Lewis Katz School of Medicine 3500 N. Broad Street, Philadelphia, PA 19140
| | - Navneet Goraya
- Temple University Lewis Katz School of Medicine, Temple University Lewis Katz School of Medicine 3500 N. Broad Street, Philadelphia, PA 19140
| | - Seonhee Kim
- Shriners Hospitals Pediatric Research Center and Department of Anatomy and Cell Biology, Temple University Lewis Katz School of Medicine 3500 N. Broad Street, Philadelphia, PA 19140
| | - Seo-Hee Cho
- Shriners Hospitals Pediatric Research Center and Department of Anatomy and Cell Biology, Temple University Lewis Katz School of Medicine 3500 N. Broad Street, Philadelphia, PA 19140
| |
Collapse
|
130
|
|
131
|
Abstract
Alternative splicing is a well-studied gene regulatory mechanism that produces biological diversity by allowing the production of multiple protein isoforms from a single gene. An involvement of alternative splicing in the key biological signalling Hippo pathway is emerging and offers new therapeutic avenues. This review discusses examples of alternative splicing in the Hippo pathway, how deregulation of these processes may contribute to disease and whether these processes offer new potential therapeutic targets.
Collapse
|
132
|
Sun D, Li X, He Y, Li W, Wang Y, Wang H, Jiang S, Xin Y. YAP1 enhances cell proliferation, migration, and invasion of gastric cancer in vitro and in vivo. Oncotarget 2018; 7:81062-81076. [PMID: 27835600 PMCID: PMC5348376 DOI: 10.18632/oncotarget.13188] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022] Open
Abstract
Yes-associated protein 1 (YAP1) plays an important role in the development of carcinomas such as breast, colorectal, and gastric (GC) cancers, but the role of YAP1 in GC has not been investigated comprehensively. The present study strongly suggests that YAP1 and P62 were significantly up-regulated in GC specimens, compared with normal gastric mucosa. In addition, the YAP1high P62high expression was independently associated with poor prognosis in GC (hazard ratio: 1.334, 95% confidence interval: 1.045–1.704, P = 0.021). Stable YAP1 silencing inhibited the proliferation, migration, and invasion of BGC-823 GC cells in vitro and inhibited the growth of xenograft tumor and hematogenous metastasis of BGC-823 GC cells in vivo. The mechanism was associated with inhibited extracellular signal-regulated kinases (ERK)1/2 phosphorylation, elevated E-cadherin protein expression and decreased vimentin protein expression, down-regulated β-catenin protein expression and elevated α-catenin protein expression, and down-regulated long non-coding RNA (lncRNA) expressions including HOX transcript antisense RNA (HOTAIR), H19, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), human large tumor suppressor-2 (LATS2)-AS1-001, and LATS2. YAP1 over-expression promoted the proliferation, migration, and invasion of human immortalized normal gastric mucosa GES-1 cells in vitro by reversing the above signal molecules. Subcutaneous inoculation of GES-1 cells and YAP1-over-expressing GES-1 cells into nude mice did not generate tumors. We successfully established the xenograft tumor models using MKN-45 GC cells, but immunochemistry showed that there was no YAP1 expression in MKN-45 cells. These results suggest that YAP1 is not a direct factor affecting tumor formation, but could accelerate tumor growth and metastasis. Collectively, this study highlights an important role for YAP1 as a promoter of GC growth and metastasis, and suggests that YAP1 could possibly be a potential treatment target for GC.
Collapse
Affiliation(s)
- Dan Sun
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Xiaoting Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yingjian He
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Wenhui Li
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Ying Wang
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Huan Wang
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Shanshan Jiang
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yan Xin
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
133
|
Kulkarni M, Tan TZ, Syed Sulaiman NB, Lamar JM, Bansal P, Cui J, Qiao Y, Ito Y. RUNX1 and RUNX3 protect against YAP-mediated EMT, stem-ness and shorter survival outcomes in breast cancer. Oncotarget 2018; 9:14175-14192. [PMID: 29581836 PMCID: PMC5865662 DOI: 10.18632/oncotarget.24419] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Hippo pathway target, YAP has emerged as an important player in solid tumor progression. Here, we identify RUNX1 and RUNX3 as novel negative regulators of oncogenic function of YAP in the context of breast cancer. RUNX proteins are one of the first transcription factors identified to interact with YAP. RUNX1 or RUNX3 expression abrogates YAP-mediated pro-tumorigenic properties of mammary epithelial cell lines in an interaction dependent manner. RUNX1 and RUNX3 inhibit YAP-mediated migration and stem-ness properties of mammary epithelial cell lines by co-regulating YAP-mediated gene expression. Analysis of whole genome expression profiles of breast cancer samples revealed significant co-relation between YAP-RUNX1/RUNX3 expression levels and survival outcomes of breast cancer patients. High RUNX1/RUNX3 expression proved protective towards YAP-dependent patient survival outcomes. High YAP in breast cancer patients' expression profiles co-related with EMT and stem-ness gene signature enrichment. High RUNX1/RUNX3 expression along with high YAP reflected lower enrichment of EMT and stem-ness signatures. This antagonistic activity of RUNX1 and RUNX3 towards oncogenic function of YAP identified in mammary epithelial cells as well as in breast cancer expression profiles gives a novel mechanistic insight into oncogene-tumor suppressor interplay in the context of breast cancer progression. The novel interplay between YAP, RUNX1 and RUNX3 and its significance in breast cancer progression can serve as a prognostic tool to predict cancer recurrence.
Collapse
Affiliation(s)
- Madhura Kulkarni
- Cancer Science Institute, NUS, Singapore.,Current address: Transnational Cancer Research Centre, Prashanti Cancer Care Mission and Indian Institute of Science Education and Research, Pune, India
| | | | | | - John M Lamar
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Current address: Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Prashali Bansal
- Cancer Science Institute, NUS, Singapore.,Current address: Max Planck Institute for Developmental Biology, Tübingen, Germany
| | | | | | - Yoshiaki Ito
- Cancer Science Institute, NUS, Singapore.,Yong Loo Lin Professor in Medical Oncology, NUS, Singapore
| |
Collapse
|
134
|
Sugimoto W, Itoh K, Mitsui Y, Ebata T, Fujita H, Hirata H, Kawauchi K. Substrate rigidity-dependent positive feedback regulation between YAP and ROCK2. Cell Adh Migr 2018; 12:101-108. [PMID: 28686514 DOI: 10.1080/19336918.2017.1338233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Extracellular matrix (ECM) stiffness influences gene expression, leading to modulation of various cellular functions. While ROCK2 regulates actomyosin activity as well as cell migration and proliferation, expression of ROCK2 is increased in response to stiffening ECM. However, the mechanism underlying rigidity-dependent ROCK2 expression remains elusive. Here, we show that YAP, a mechanically regulated transcription coactivator, upregulates ROCK2 expression in an ECM rigidity-dependent manner. YAP interacted with the ROCK2 promoter region in an actomyosin activity-dependent manner. Knockdown of YAP decreased ROCK2 expression while activity of the ROCK2 promoter was upregulated by expressing constitutively active YAP. Furthermore, we found that ROCK2 expression promotes transcriptional activation by YAP. Our results reveal a novel positive feedback loop between YAP and ROCK2, which is modulated by ECM stiffness.
Collapse
Affiliation(s)
- Wataru Sugimoto
- a Frontiers of Innovative Research in Science and Technology , Konan University , Kobe, Hyogo , Japan
| | - Katsuhiko Itoh
- a Frontiers of Innovative Research in Science and Technology , Konan University , Kobe, Hyogo , Japan
| | - Yasumasa Mitsui
- a Frontiers of Innovative Research in Science and Technology , Konan University , Kobe, Hyogo , Japan
| | - Takahiro Ebata
- a Frontiers of Innovative Research in Science and Technology , Konan University , Kobe, Hyogo , Japan
| | - Hideaki Fujita
- b Laboratory for Comprehensive Bioimaging , Riken Qbic , Osaka , Japan.,c Waseda Bioscience Research Institute in Singapore , Singapore , Republic of Singapore
| | - Hiroaki Hirata
- d Mechanobiology Laboratory , Nagoya University Graduate School of Medicine , Nagoya, Aichi , Japan
| | - Keiko Kawauchi
- a Frontiers of Innovative Research in Science and Technology , Konan University , Kobe, Hyogo , Japan.,e Department of Molecular Oncology , Institute for Advanced Medical Sciences, Nippon Medical School , Kawasaki, Kanagawa , Japan
| |
Collapse
|
135
|
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: 5.0] [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
|
136
|
Thongon N, Castiglioni I, Zucal C, Latorre E, D'Agostino V, Bauer I, Pancher M, Ballestrero A, Feldmann G, Nencioni A, Provenzani A. The GSK3β inhibitor BIS I reverts YAP-dependent EMT signature in PDAC cell lines by decreasing SMADs expression level. Oncotarget 2018; 7:26551-66. [PMID: 27034169 PMCID: PMC5041998 DOI: 10.18632/oncotarget.8437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/06/2016] [Indexed: 12/16/2022] Open
Abstract
The Yes-associated protein, YAP, is a transcriptional co-activator, mediating the Epithelial to Mesenchymal Transition program in pancreatic ductal adenocarcinoma (PDAC). With the aim to identify compounds that can specifically modulate YAP functionality in PDAC cell lines, we performed a small scale, drug-based screening experiment using YAP cell localization as the read-out. We identified erlotinib as an inducer of YAP cytoplasmic localization, an inhibitor of the TEA luciferase reporter system and the expression of the bona fide YAP target gene, Connective Tissue Growth Factor CTGF. On the other hand, BIS I, an inhibitor of PKCδ and GSK3β, caused YAP accumulation into the nucleus. Activation of β-catenin reporter and interfering experiments show that inhibition of the PKCδ/GSK3β pathway triggers YAP nuclear accumulation inducing YAP/TEAD transcriptional response. Inhibition of GSK3β by BIS I reduced the expression levels of SMADs protein and reduced YAP contribution to EMT. Notably, BIS I reduced proliferation, migration and clonogenicity of PDAC cells in vitro, phenocopying YAP genetic down-regulation. As shown by chromatin immunoprecipitation experiments and YAP over-expressing rescue experiments, BIS I reverted YAP-dependent EMT program by modulating the expression of the YAP target genes E-cadherin, vimentin, CTGF and of the newly identified target, CD133. In conclusion, we identified two different molecules, erlotinib and BIS I, modulating YAP functionality although via different mechanisms of action, with the second one specifically inhibiting the YAP-dependent EMT program in PDAC cell lines.
Collapse
Affiliation(s)
- Natthakan Thongon
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Ilaria Castiglioni
- Laboratory of Gene Expression and Muscular Dystrophy, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Zucal
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Elisa Latorre
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Vito D'Agostino
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Inga Bauer
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Michael Pancher
- High Throughput Screening Facility, Centre for Integrative Biology, University of Trento, Trento, Italy
| | | | - Georg Feldmann
- Laboratory of Pancreatic Cancer Translational Research, Clinic University of Bonn, Bonn, Germany
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Alessandro Provenzani
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
137
|
Furth N, Aylon Y, Oren M. p53 shades of Hippo. Cell Death Differ 2018; 25:81-92. [PMID: 28984872 PMCID: PMC5729527 DOI: 10.1038/cdd.2017.163] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/15/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022] Open
Abstract
The three p53 family members, p53, p63 and p73, are structurally similar and share many biochemical activities. Yet, along with their common fundamental role in protecting genomic fidelity, each has acquired distinct functions related to diverse cell autonomous and non-autonomous processes. Similar to the p53 family, the Hippo signaling pathway impacts a multitude of cellular processes, spanning from cell cycle and metabolism to development and tumor suppression. The core Hippo module consists of the tumor-suppressive MST-LATS kinases and oncogenic transcriptional co-effectors YAP and TAZ. A wealth of accumulated data suggests a complex and delicate regulatory network connecting the p53 and Hippo pathways, in a highly context-specific manner. This generates multiple layers of interaction, ranging from interdependent and collaborative signaling to apparent antagonistic activity. Furthermore, genetic and epigenetic alterations can disrupt this homeostatic network, paving the way to genomic instability and cancer. This strengthens the need to better understand the nuances that control the molecular function of each component and the cross-talk between the different components. Here, we review interactions between the p53 and Hippo pathways within a subset of physiological contexts, focusing on normal stem cells and development, as well as regulation of apoptosis, senescence and metabolism in transformed cells.
Collapse
Affiliation(s)
- Noa Furth
- Department of Molecular Cell Biology, The Weizmann Institute, Rehovot, Israel
| | - Yael Aylon
- Department of Molecular Cell Biology, The Weizmann Institute, Rehovot, Israel
- Department of Molecular Cell Biology, The Weizmann Institute, POB 26, 234 Herzl Street, Rehovot 7610001, Israel. Tel: +972 89342358; Fax: +972 89346004; E-mail: or
| | - Moshe Oren
- Department of Molecular Cell Biology, The Weizmann Institute, Rehovot, Israel
- Department of Molecular Cell Biology, The Weizmann Institute, POB 26, 234 Herzl Street, Rehovot 7610001, Israel. Tel: +972 89342358; Fax: +972 89346004; E-mail: or
| |
Collapse
|
138
|
Wang SP, Wang LH. Disease implication of hyper-Hippo signalling. Open Biol 2017; 6:rsob.160119. [PMID: 27805903 PMCID: PMC5090056 DOI: 10.1098/rsob.160119] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
The Hippo signalling pathway regulates cellular proliferation, apoptosis and differentiation, thus exerting profound effects on cellular homeostasis. Inhibition of Hippo signalling has been frequently implicated in human cancers, indicating a well-known tumour suppressor function of the Hippo pathway. However, it is less certain whether and how hyperactivation of the Hippo pathway affects biological outcome in living cells. This review describes current knowledge of the regulatory mechanisms of the Hippo pathway, mainly focusing on hyperactivation of the Hippo signalling nexus. The disease implications of hyperactivated Hippo signalling have also been discussed, including arrhythmogenic cardiomyopathy, Sveinsson's chorioretinal atrophy, Alzheimer's disease, amyotrophic lateral sclerosis and diabetes. By highlighting the significance of disease-relevant Hippo signalling activation, this review can offer exciting prospects to address the onset and potential reversal of Hippo-related disorders.
Collapse
Affiliation(s)
- Shu-Ping Wang
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 114, Taiwan
| |
Collapse
|
139
|
Yang W, Ma J, Zhou W, Zhou X, Cao B, Zhang H, Zhao Q, Fan D, Hong L. Molecular mechanisms and clinical implications of miRNAs in drug resistance of esophageal cancer. Expert Rev Gastroenterol Hepatol 2017; 11:1151-1163. [PMID: 28838272 DOI: 10.1080/17474124.2017.1372189] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
With the increasing incidence of esophageal cancer, drug resistance is becoming a major obstacle to successful cancer therapy since chemotherapy is regarded as a curative approach to inhibit cancer cell proliferation. Despite the great progress in anticancer treatment achieved during the last decades, the mechanisms of multidrug resistance have not been completely elucidated. Recently, accumulating studies and pre-clinical reports highlighted the role of miRNAs in the drug resistance of esophageal cancer. Areas covered: In this review, we mainly summarized the current advances of miRNAs in esophageal cancer and the mechanisms underlying drug resistance. We also reviewed the potential role of miRNAs as biomarkers for predicting drug response and prognosis. Finally, we envisaged the future orientation and challenges in translating the existing knowledge of drug resistance related miRNAs into clinical applications. Expert commentary: Based on the current knowledge of certain miRNAs, we believe that miRNAs would be helpful to overcome the drug resistance and provide personalized treatment for patients with esophageal cancer. The aims of this study were to provide a comprehensive summary on the emerging role of miRNAs in the drug resistance of esophageal cancer and attract broad attention of more researchers on this field.
Collapse
Affiliation(s)
- Wanli Yang
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jiaojiao Ma
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Wei Zhou
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Xin Zhou
- b The First Brigade of Student , Fourth Military Medical University , Xi'an , China
| | - Bo Cao
- b The First Brigade of Student , Fourth Military Medical University , Xi'an , China
| | - Hongwei Zhang
- c Department of Digestive Surgery , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Qingchuan Zhao
- c Department of Digestive Surgery , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Daiming Fan
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Liu Hong
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| |
Collapse
|
140
|
Andrade D, Mehta M, Griffith J, Panneerselvam J, Srivastava A, Kim TD, Janknecht R, Herman T, Ramesh R, Munshi A. YAP1 inhibition radiosensitizes triple negative breast cancer cells by targeting the DNA damage response and cell survival pathways. Oncotarget 2017; 8:98495-98508. [PMID: 29228705 PMCID: PMC5716745 DOI: 10.18632/oncotarget.21913] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/29/2017] [Indexed: 11/25/2022] Open
Abstract
The Hippo pathway is an evolutionarily conserved signaling pathway that regulates proliferation and apoptosis to control organ size during developmental growth. Yes-associated protein 1 (YAP1), the terminal effector of the Hippo pathway, is a transcriptional co-activator and a potent growth promoter that has emerged as a critical oncogene. Overexpression of YAP1 has been implicated in promoting resistance to chemo-, radiation and targeted therapy in various cancers. However, the role of YAP1 in radioresistance in triple-negative breast cancer (TNBC) is currently unknown. We evaluated the role of YAP1 in radioresistance in TNBC in vitro, using two approaches to inhibit YAP1: 1) genetic inhibition by YAP1 specific shRNA or siRNA, and 2) pharmacological inhibition by using the small molecule inhibitor, verteporfin that prevents YAP1 transcriptional activity. Our findings demonstrate that both genetic and pharmacological inhibition of YAP1 sensitizes TNBC cells to radiation by inhibiting the EGFR/PI3K/AKT signaling axis and causing an increased accumulation of DNA damage. Our results reveal that YAP1 activation exerts a protective role for TNBC cells in radiotherapy and represents a pharmacological target to enhance the anti-tumor effects of DNA damaging modalities in the treatment of TNBC.
Collapse
Affiliation(s)
- Daniel Andrade
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Meghna Mehta
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - James Griffith
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Janani Panneerselvam
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Akhil Srivastava
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Tae-Dong Kim
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ralf Janknecht
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Terence Herman
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Anupama Munshi
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
141
|
Ou C, Sun Z, Li S, Li G, Li X, Ma J. Dual roles of yes-associated protein (YAP) in colorectal cancer. Oncotarget 2017; 8:75727-75741. [PMID: 29088905 PMCID: PMC5650460 DOI: 10.18632/oncotarget.20155] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 07/30/2017] [Indexed: 02/07/2023] Open
Abstract
Yes-associated protein (YAP) is a downstream effector molecule of a newly emerging tumour suppressor pathway called the Hippo pathway. YAP is a transcriptional co-activator and mis-expressed in various cancers, including colorectal cancer (CRC). Accumulating studies show that the high expression of nuclear YAP is linked with tumour progression and decreased survival. Nuclear YAP can interact with other transcription factors to promote cancer cell proliferation, apoptosis, metastasis and maintenance of stemness. Therefore, YAP has the potential to be a tumour biomarker or therapeutic target for CRC. However, recently, a number of studies have supported a contradictory role for YAP as a tumour suppressor, demonstrating inhibition of the tumorigenesis of CRC, involvement in promoting cell apoptosis, and inhibiting the maintenance of intestinal stem cells and inflammatory activity. In these studies, high expression of YAP was highly correlated with worse survival in CRC. In this review, we will comprehensively summarize and analyse these paradoxical reports, and discuss both the oncogenic and tumour suppressor functions of YAP in the differential status of CRC progression. Further investigation into the mechanisms responsible for the dual function of YAP will be of great value in the prevention, early diagnosis, and therapy of CRC.
Collapse
Affiliation(s)
- Chunlin Ou
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zhenqiang Sun
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Gastrointestinal Surgery, Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Shen Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
| | - Guiyuan Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
142
|
Tissue inhibitor of metalloproteinase-1 promotes cell proliferation through YAP/TAZ activation in cancer. Oncogene 2017; 37:263-270. [DOI: 10.1038/onc.2017.321] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/25/2017] [Accepted: 08/07/2017] [Indexed: 02/06/2023]
|
143
|
Li Y, Kong F, Shao Q, Wang R, Hu E, Liu J, Jin C, He D, Xiao X. YAP Expression and Activity Are Suppressed by S100A7 via p65/NFκB-mediated Repression of ΔNp63. Mol Cancer Res 2017; 15:1752-1763. [DOI: 10.1158/1541-7786.mcr-17-0349] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/14/2017] [Accepted: 09/08/2017] [Indexed: 11/16/2022]
|
144
|
Hicks-Berthet J, Varelas X. Integrin-FAK-CDC42-PP1A signaling gnaws at YAP/TAZ activity to control incisor stem cells. Bioessays 2017; 39. [PMID: 28891248 DOI: 10.1002/bies.201700116] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
How epithelial tissues are able to self-renew to maintain homeostasis and regenerate in response to injury remains a persistent question. The transcriptional effectors YAP and TAZ are increasingly being recognized as central mediators of epithelial stem cell biology, and a wealth of recent studies have been directed at understanding the control and activity of these factors. Recent work by Hu et al. has added to this knowledge, as they identify an Integrin-FAK-CDC42-PP1A signaling cascade that directs nuclear YAP/TAZ activity in stem cell populations of the mouse incisor, and define convergence on mTORC1 signaling as an important mediator of the proliferation of these cells. Here, we review recent studies on YAP/TAZ function and regulation in epithelial tissue-specific stem cells, merging the Hu et al. study together with our current knowledge of YAP/TAZ.
Collapse
Affiliation(s)
- Julia Hicks-Berthet
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
145
|
Abstract
Proper cellular functionality and homeostasis are maintained by the convergent integration of various signaling cascades, which enable cells to respond to internal and external changes. The Dbf2-related kinases LATS1 and LATS2 (LATS) have emerged as central regulators of cell fate, by modulating the functions of numerous oncogenic or tumor suppressive effectors, including the canonical Hippo effectors YAP/TAZ, the Aurora mitotic kinase family, estrogen signaling and the tumor suppressive transcription factor p53. While the basic functions of the LATS kinase module are strongly conserved over evolution, the genomic duplication event leading to the emergence of two closely related kinases in higher organisms has increased the complexity of this signaling network. Here, we review the LATS1 and LATS2 intrinsic features as well as their reported cellular activities, emphasizing unique characteristics of each kinase. While differential activities between the two paralogous kinases have been reported, many converge to similar pathways and outcomes. Interestingly, the regulatory networks controlling the mRNA expression pattern of LATS1 and LATS2 differ strongly, and may contribute to the differences in protein binding partners of each kinase and in the subcellular locations in which each kinase exerts its functions.
Collapse
Affiliation(s)
- Noa Furth
- Department of Molecular Cell Biology, The Weizmann Institute of Science, POB 26, 234 Herzl St., Rehovot 7610001, Israel
| | - Yael Aylon
- Department of Molecular Cell Biology, The Weizmann Institute of Science, POB 26, 234 Herzl St., Rehovot 7610001, Israel
| |
Collapse
|
146
|
Mutant p53 Protein and the Hippo Transducers YAP and TAZ: A Critical Oncogenic Node in Human Cancers. Int J Mol Sci 2017; 18:ijms18050961. [PMID: 28467351 PMCID: PMC5454874 DOI: 10.3390/ijms18050961] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/11/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
p53 protein is a well-known tumor suppressor factor that regulates cellular homeostasis. As it has several and key functions exerted, p53 is known as “the guardian of the genome” and either loss of function or gain of function mutations in the TP53 coding protein sequence are involved in cancer onset and progression. The Hippo pathway is a key regulator of developmental and regenerative physiological processes but if deregulated can induce cell transformation and cancer progression. The p53 and Hippo pathways exert a plethora of fine-tuned functions that can apparently be in contrast with each other. In this review, we propose that the p53 status can affect the Hippo pathway function by switching its outputs from tumor suppressor to oncogenic activities. In detail, we discuss: (a) the oncogenic role of the protein complex mutant p53/YAP; (b) TAZ oncogenic activation mediated by mutant p53; (c) the therapeutic potential of targeting mutant p53 to impair YAP and TAZ oncogenic functions in human cancers.
Collapse
|
147
|
Cho SY, Kim K, Park MS, Jang MY, Choi YH, Han S, Shin HM, Chung C, Han HY, Yang JB, Ko YB, Yoo HJ. Expression of Yes-associated protein 1 and its clinical significance in ovarian serous cystadenocarcinoma. Oncol Rep 2017; 37:2620-2632. [PMID: 28339095 PMCID: PMC5428545 DOI: 10.3892/or.2017.5517] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/03/2016] [Indexed: 02/04/2023] Open
Abstract
Yes-associated protein 1 (YAP1) is a key transcriptional regulator in the Hippo signaling pathway that plays a critical role in the development and progression of several types of malignancies, including ovarian cancer. Herein, we investigated the expression of YAP1 and its clinical significance in a large population of patients with ovarian serous cystadenocarcinoma (OSC), which is the most common form of epithelial ovarian neoplasm, using the TCGA database. Surprisingly, cross-cancer mRNA expression and alterations in YAP1 were higher in OSC than in those of other types of cancers in the TCGA database. YAP1 mRNA expression was significantly higher in OSC compared with normal ovarian samples, and was higher in stages III and IV, than stages I and II. The level of YAP1 protein, which is mainly localized to the nucleus, was also higher in stage IV as compared with stages I, II and III. However, the protein level of pYAP1, which is inactive and is localized to the cytoplasm, was not significantly different between stages. The ratio of pYAP/YAP, which shows higher activity at a low ratio, was lower in stage III than in stages I and II. High YAP and low pYAP levels were significantly correlated with a poor prognosis in patients with OSC. The mRNA and protein expression of YAP1 were significantly increased in the proliferative subtype as compared to the differentiated, immunoreactive and mesenchymal subtypes. According to bioinformatics analysis, YAP1 is most highly correlated with the cell cycle. TGF-β signaling and WNT signaling were significantly increased in the high YAP1 group according to gene set enrichment analysis. Taken together, our results suggest that not only high YAP1 expression but also its subcellular distribution may be associated with poor overall survival in patients with OSC.
Collapse
Affiliation(s)
- Sang Yeon Cho
- Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Kwanghun Kim
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Soo Park
- Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Mi Young Jang
- Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Young Hwan Choi
- Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Suyeon Han
- Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyun Mo Shin
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chaeuk Chung
- Department of Pulmonary and Critical Care Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Hye Young Han
- Department of Pediatrics, Chungnam National University, Daejeon, Republic of Korea
| | - Jung Bo Yang
- Department of Obstetrics and Gynecology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Young Bok Ko
- Department of Obstetrics and Gynecology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Heon Jong Yoo
- Department of Obstetrics and Gynecology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
148
|
Liu Y, He K, Hu Y, Guo X, Wang D, Shi W, Li J, Song J. YAP modulates TGF-β1-induced simultaneous apoptosis and EMT through upregulation of the EGF receptor. Sci Rep 2017; 7:45523. [PMID: 28425446 PMCID: PMC5397873 DOI: 10.1038/srep45523] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/28/2017] [Indexed: 02/08/2023] Open
Abstract
YAP is a transcriptional co-regulator that plays important roles in various patho-physiological processes, including the survival and death of cells. However, the effect of YAP on apoptosis and EMT, simultaneously mediated by TGF-β1, is not known. In this study, we demonstrate that YAP can modulate cell fate of apoptosis versus EMT by acting as a surviving factor. Overexpression of YAP in mouse mammary epithelial (NMuMG) cells suppressed TGF-β1-induced apoptosis, which shifted the cellular response predominantly toward EMT. In contrast, knockdown of YAP induced spontaneous apoptosis and enhanced TGF-β1-induced apoptosis, leading to a sharp decrease in the proportion of surviving cells that underwent EMT. These data suggest that YAP is an essential factor for modulating cellular responses to TGF-β1. Further investigation showed that YAP could regulate the expression level and activation of EGFR. Knockdown or inhibition of EGFR abolished the suppressive effect of YAP on apoptosis, whereas activation of EGFR by EGF significantly reduced apoptosis caused by the knockdown of YAP. The results indicate that EGFR and its activation are critical for YAP-mediated suppression of TGF-β1-induced apoptosis. This study provides a new understanding of the regulatory mechanism underlying the determination of cell fate in response to TGF-β1-mediated simultaneous apoptosis and EMT.
Collapse
Affiliation(s)
- Yi Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Kai He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaojie Guo
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Dongmei Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Weiwei Shi
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jingsong Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jianguo Song
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
149
|
Pfleger CM. The Hippo Pathway: A Master Regulatory Network Important in Development and Dysregulated in Disease. Curr Top Dev Biol 2017; 123:181-228. [PMID: 28236967 DOI: 10.1016/bs.ctdb.2016.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Hippo Pathway is a master regulatory network that regulates proliferation, cell growth, stemness, differentiation, and cell death. Coordination of these processes by the Hippo Pathway throughout development and in mature organisms in response to diverse external and internal cues plays a role in morphogenesis, in controlling organ size, and in maintaining organ homeostasis. Given the importance of these processes, the Hippo Pathway also plays an important role in organismal health and has been implicated in a variety of diseases including eye disease, cardiovascular disease, neurodegeneration, and cancer. This review will focus on Drosophila reports that identified the core components of the Hippo Pathway revealing specific downstream biological outputs of this complicated network. A brief description of mammalian reports will complement review of the Drosophila studies. This review will also survey upstream regulation of the core components with a focus on feedback mechanisms.
Collapse
Affiliation(s)
- Cathie M Pfleger
- The Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
150
|
Xiao Q, Qian Z, Zhang W, Liu J, Hu E, Zhang J, Li M, Wang J, Kong F, Li Y, Wang R, Tan X, He D, Xiao X. Depletion of CABYR-a/b sensitizes lung cancer cells to TRAIL-induced apoptosis through YAP/p73-mediated DR5 upregulation. Oncotarget 2017; 7:9513-24. [PMID: 26843620 PMCID: PMC4891056 DOI: 10.18632/oncotarget.7069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/13/2016] [Indexed: 12/24/2022] Open
Abstract
Our previous study revealed that knockdown of CABYR-a/b increases the chemosensitivity of lung cancer cells through inactivation of Akt. Here, we demonstrated that depletion of CABYR-a/b significantly increased DR5 expression and sensitized lung cancer cells to TRAIL-induced apoptosis in vitro and/or in vivo. Importantly, treatment with AD5-10, a DR5-specific agonistic monoclonal antibody, was able to mimic TRAIL-induced apoptosis in CABYR-a/b-silenced cells. Strikingly, we identified that depletion of CABYR-a/b not only increased the expressions of p73 and DR5 but also decreased the phosphorylation of YAP S127. Loss- or gain-of-function studies of YAP and p73 revealed that double deletions of YAP and p73 effectively decreased the expression of DR5 and abolished TRAIL-induced apoptosis in CABYR-a/b knockdown cells. Conversely, the co-overexpression of YAP and p73 promoted the expression of DR5 and sensitized cells to TRAIL-induced apoptosis. Taken together, our results demonstrate that depletion of CABYR-a/b sensitizes lung cancer cells to TRAIL-induced apoptosis through YAP/p73-mediated DR5 upregulation.
Collapse
Affiliation(s)
- Qianqian Xiao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Zunlei Qian
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China.,College of Forensic Sciences, People's Public Security University of China, Beijing, China
| | - Weiqing Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Jin Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Enze Hu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Jinsan Zhang
- School of Pharmaceutical Sciences and Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mingying Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China.,Center of Reproduction and Genetics, First People's Hospital of Yunnan Province, Kunming, China
| | - Junhao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Fei Kong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Yunguang Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Rui Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Xiaohua Tan
- School of Pharmaceutical Sciences and Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dacheng He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| | - Xueyuan Xiao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, China
| |
Collapse
|