151
|
Abstract
Drosophila melanogaster has been a central player in the discovery of the Hippo pathway and in understanding its in vivo functions. From a technique standpoint, the Flp-FRT system for the generation of genetic mosaics has been a principle tool. It has broadly been used in the discovery of Hippo pathway members in mutagenesis screens, in the analysis of target gene expression, and in genetic epistasis. Here we briefly introduce this tool, summarize its use in the Hippo pathway field, and provide a protocol for the generation of Flp-FRT clones in imaginal discs with dissection and staining for reporter gene expression to characterize candidate Hippo pathway genes.
Collapse
Affiliation(s)
- Mardelle Atkins
- Department of Biological Sciences, Sam Houston State University, Huntsville, TX, USA.
| |
Collapse
|
152
|
Abstract
Cancer is a cumulative manifestation of several complicated disease states that affect multiple organs. Over the last few decades, the fruit fly Drosophila melanogaster, has become a successful model for studying human cancers. The genetic simplicity and vast arsenal of genetic tools available in Drosophila provides a unique opportunity to address questions regarding cancer initiation and progression that would be extremely challenging in other model systems. In this chapter we provide a historical overview of Drosophila as a model organism for cancer research, summarize the multitude of genetic tools available, offer a brief comparison between different model organisms and cell culture platforms used in cancer studies and briefly discuss some of the latest models and concepts in recent Drosophila cancer research.
Collapse
|
153
|
Abstract
The Hippo Pathway comprises a vast network of components that integrate diverse signals including mechanical cues and cell surface or cell-surface-associated molecules to define cellular outputs of growth, proliferation, cell fate, and cell survival on both the cellular and tissue level. Because of the importance of the regulators, core components, and targets of this pathway in human health and disease, individual components were often identified by efforts in mammalian models or for a role in a specific process such as stress response or cell death. However, multiple components were originally discovered in the Drosophila system, and the breakthrough of conceiving that these components worked together in a signaling pathway came from a series of Drosophila genetic screens and fundamental genetic and phenotypic characterization efforts. In this chapter, we will review the original discoveries leading to the conceptual framework of these components as a tumor suppressor network. We will review chronologically the early efforts that established our initial understanding of the core machinery that then launched the growing and vibrant field to be discussed throughout later chapters of this book.
Collapse
Affiliation(s)
- Rewatee Gokhale
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cathie M Pfleger
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
154
|
Two Antagonistic Hippo Signaling Circuits Set the Division Plane at the Medial Position in the Ciliate Tetrahymena. Genetics 2018; 211:651-663. [PMID: 30593491 DOI: 10.1534/genetics.118.301889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/21/2018] [Indexed: 12/17/2022] Open
Abstract
In a single cell, ciliates maintain a complex pattern of cortical organelles that are arranged along the anteroposterior and circumferential axes. The underlying molecular mechanisms of intracellular pattern formation in ciliates are largely unknown. Ciliates divide by tandem duplication, a process that remodels the parental cell into two daughters aligned head-to-tail. In the elo1-1 mutant of Tetrahymena thermophila, the segmentation boundary/division plane forms too close to the posterior end of the parental cell, producing a large anterior and a small posterior daughter cell, respectively. We show that ELO1 encodes a Lats/NDR kinase that marks the posterior segment of the cell cortex, where the division plane does not form in the wild-type. Elo1 acts independently of CdaI, a Hippo/Mst kinase that marks the anterior half of the parental cell, and whose loss shifts the division plane anteriorly. We propose that, in Tetrahymena, two antagonistic Hippo circuits focus the segmentation boundary/division plane at the equatorial position, by excluding divisional morphogenesis from the cortical areas that are too close to cell ends.
Collapse
|
155
|
Sollazzo M, Genchi C, Paglia S, Di Giacomo S, Pession A, de Biase D, Grifoni D. High MYC Levels Favour Multifocal Carcinogenesis. Front Genet 2018; 9:612. [PMID: 30619451 PMCID: PMC6297171 DOI: 10.3389/fgene.2018.00612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/20/2018] [Indexed: 02/05/2023] Open
Abstract
The term "field cancerisation" describes the formation of tissue sub-areas highly susceptible to multifocal tumourigenesis. In the earlier stages of cancer, cells may indeed display a series of molecular alterations that allow them to proliferate faster, eventually occupying discrete tissue regions with irrelevant morphological anomalies. This behaviour recalls cell competition, a process based on a reciprocal fitness comparison: when cells with a growth advantage arise in a tissue, they are able to commit wild-type neighbours to death and to proliferate at their expense. It is known that cells expressing high MYC levels behave as super-competitors, able to kill and replace less performant adjacent cells; given MYC upregulation in most human cancers, MYC-mediated cell competition is likely to pioneer field cancerisation. Here we show that MYC overexpression in a sub-territory of the larval wing epithelium of Drosophila is sufficient to trigger a number of cellular responses specific to mammalian pre-malignant tissues. Moreover, following induction of different second mutations, high MYC-expressing epithelia were found to be susceptible to multifocal growth, a hallmark of mammalian pre-cancerous fields. In summary, our study identified an early molecular alteration implicated in field cancerisation and established a genetically amenable model which may help study the molecular basis of early carcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniela Grifoni
- Cancer Evolution Laboratory, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
156
|
Xie H, Wu L, Deng Z, Huo Y, Cheng Y. Emerging roles of YAP/TAZ in lung physiology and diseases. Life Sci 2018; 214:176-183. [PMID: 30385178 DOI: 10.1016/j.lfs.2018.10.062] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/22/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022]
Abstract
The YAP and TAZ, as the downstream effectors of Hippo pathway, have emerged as important translational co-activators of a wide variety of biological processes. YAP/TAZ plays a crucial role in the lung development and physiology. Dysregulation of YAP/TAZ signaling pathway contributes to the development and progression of chronic lung diseases, including lung cancer, pulmonary fibrosis, pulmonary hypertension, COPD, asthma, and lung infection. Therefore, owing to its critical functions, delineation of the signaling mechanisms of YAP/TAZ in pathological conditions will shed light on developing strategies for its therapeutic targeting. Currently, the complex regulation of this pathway is under extensive investigation. In this review, we summarize and present recent findings of molecular mechanisms of YAP/TAZ in the lung physiological and pathological conditions, as well as the implications of YAP/TAZ for lung diseases treatment and regeneration.
Collapse
Affiliation(s)
- Haojun Xie
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Liquan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenan Deng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yating Huo
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yuanxiong Cheng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
157
|
Xu X, Bi HL, Zhang ZJ, Yang Y, Li K, Huang YP, Zhang Y, He L. BmHpo mutation induces smaller body size and late stage larval lethality in the silkworm, Bombyx mori. INSECT SCIENCE 2018; 25:1006-1016. [PMID: 29808962 DOI: 10.1111/1744-7917.12607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/19/2018] [Accepted: 05/24/2018] [Indexed: 06/08/2023]
Abstract
As a core member of the Hippo signaling pathway, Hpo plays a critical role in regulating growth and development. Previous studies reported that loss of function of Hpo results in increased proliferation, reduced apoptosis and induction of tissue overgrowth in Drosophila. In this study, we used CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/Cas9) to study Hpo gene (BmHpo) function in the lepidopteran insect Bombyx mori, known commonly as the silkworm. Sequence analysis of BmHpo revealed an array of deletions in mutants. We found that BmHpo knockout resulted in defects in body size regulation, in developmental defects and pigment accumulation and early death. Our data show that BmHpo is essential for regulation of insect growth and development and that CRISPR/Cas9 technology can serve as a basis for functional analysis of target genes in lepidopteran insects.
Collapse
Affiliation(s)
- Xia Xu
- School of Life Science, East China Normal University, Shanghai, China
| | - Hong-Lun Bi
- School of Life Science, East China Normal University, Shanghai, China
| | - Zhong-Jie Zhang
- School of Life Science, East China Normal University, Shanghai, China
| | - Yang Yang
- School of Life Science, East China Normal University, Shanghai, China
| | - Kai Li
- School of Life Science, East China Normal University, Shanghai, China
| | - Yong-Ping Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yong Zhang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lin He
- School of Life Science, East China Normal University, Shanghai, China
| |
Collapse
|
158
|
Xu X, Zhang Z, Yang Y, Huang S, Li K, He L, Zhou X. Genome editing reveals the function of Yorkie during the embryonic and early larval development in silkworm, Bombyx mori. INSECT MOLECULAR BIOLOGY 2018; 27:675-685. [PMID: 29797485 DOI: 10.1111/imb.12502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
As a transcriptional coactivator, Yorkie (Yki) is a major downstream target of the Hippo signalling pathway to regulate the organ size during animal development and regeneration. Previous microarray analysis in the silkworm, Bombyx mori, has shown that genes associated with the Hippo pathway were primarily expressed in gonads and imaginal discs. The RNA-interference-mediated silencing of Yki at the early wandering stage delayed B. mori development and ovary maturation, whereas baculovirus-mediated overexpression at the late larval instar facilitated organ growth and accelerated metamorphosis. Here, we employed CRISPR/Cas9-mediated mutagenesis to investigate the function of Yki in B. mori (BmYki) at the embryonic and early larval stages. Knocking out of BmYki led to reduced body size, moulting defects and, eventually, larval lethality. Sequence analysis of CRISPR/Cas9 mutants exhibited an array of deletions in BmYki. As a critical downstream effector of the Hippo kinase cassette, silencing of BmYki at the embryonic stage is indispensable and the consequence is lethal. Given that the Hippo signalling pathway is evolutionarily conserved, Yki has the potential to be a novel molecular target for genetic-based pest management practices.
Collapse
Affiliation(s)
- X Xu
- School of Life Science, East China Normal University, Shanghai, China
| | - Z Zhang
- School of Life Science, East China Normal University, Shanghai, China
| | - Y Yang
- School of Life Science, East China Normal University, Shanghai, China
| | - S Huang
- Agricultural and Medical Biotechnology, University of Kentucky, Lexington, KY, USA
| | - K Li
- School of Life Science, East China Normal University, Shanghai, China
| | - L He
- School of Life Science, East China Normal University, Shanghai, China
| | - X Zhou
- Department of Entomology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
159
|
Hypermethylated LATS2 gene with decreased expression in female breast cancer: A case control study from North India. Gene 2018; 676:156-163. [PMID: 30010037 DOI: 10.1016/j.gene.2018.07.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/25/2018] [Accepted: 07/11/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND LATS2, a presumed tumor suppressor gene located on chromosome 13q11-12 is involved in cell growth related activity like regulation of cell cycle at G1/S. The reduced expression of LATS2 has been reported in many tumors; including tumors of Breast, which is to the best of our knowledge has not been studied in north Indian female breast cancer population. OBJECTIVE Here, we looked upon the expression pattern and methylation status of the LATS2 gene in north Indian female breast cancer cases to further strengthen its role as a tumor suppressor gene and more importantly as a cancer biomarker. METHODS mRNA expression level was determined by real time PCR in 140 Breast cancer patients, Protein expression was studied by Immunohistochemistry and Promoter methylation was studied by Methylation specific PCR. All findings were correlated with clinicopathological features. RESULTS LATS2 mRNA expression was remarkably downregulated in 67.85% (95/140) cases. The expression of Large Associated Tumor Suppressor 2 at protein level was also absent in 67.85% (95/140) cases. The absence of LATS2 protein strongly correlated with promoter hypermethylation where 91 out of a total of 107 hyper methylated cases showed absence of protein (91/107, 85%). The absence of LATS2 protein was strongly significant with HER2 neu status (0.01), TNM staging (0.009) and Molecular subtype (0.024). CONCLUSION The decreased expression in breast cancer seems to be associated with hypermethylation of LATS2 promoter regions. Further LATS2 as a tumor suppressor can be recognized as a promising Biomarker in Breast cancer pathogenesis. Though, further studies, targeting larger sets of breast cancer population are required to establish LATS2 as a promising biomarker.
Collapse
|
160
|
Zhang Q, Han X, Chen J, Xie X, Xu J, Zhao Y, Shen J, Hu L, Xu P, Song H, Zhang L, Zhao B, Wang YJ, Xia Z. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) mediate cell density-dependent proinflammatory responses. J Biol Chem 2018; 293:18071-18085. [PMID: 30315101 DOI: 10.1074/jbc.ra118.004251] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/14/2018] [Indexed: 12/21/2022] Open
Abstract
A proper inflammatory response is critical to the restoration of tissue homeostasis after injury or infection, but how such a response is modulated by the physical properties of the cellular and tissue microenvironments is not fully understood. Here, using H358, HeLa, and HEK293T cells, we report that cell density can modulate inflammatory responses through the Hippo signaling pathway. We found that NF-κΒ activation through the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα) is not affected by cell density. However, we also noted that specific NF-κΒ target genes, such as cyclooxygenase 2 (COX-2), are induced much less at low cell densities than at high cell densities. Mechanistically, we observed that the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are localized to the nucleus, bind to TEA domain transcription factors (TEADs), recruit histone deacetylase 7 (HDAC7) to the promoter region of COX-2, and repress its transcription at low cell density and that high cell density abrogates this YAP/TAZ-mediated transcriptional repression. Of note, IL-1β stimulation promoted cell migration and invasion mainly through COX-2 induction, but YAP inhibited this induction and thus cell migration and invasion. These results suggest that YAP/TAZ-TEAD interactions can repress COX-2 transcription and thereby mediate cell density-dependent modulation of proinflammatory responses. Our findings highlight that the cellular microenvironment significantly influences inflammatory responses via the Hippo pathway.
Collapse
Affiliation(s)
- Qiong Zhang
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China,; Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan, China,; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and
| | - Xu Han
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Jinfeng Chen
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan, China
| | - Xiaomei Xie
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China,; Youth League Committee of Zhejiang Gongshang University, Hangzhou, 310018 Zhejiang, China, and
| | - Jiafeng Xu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Yang Zhao
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Jie Shen
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 Zhejiang, China
| | - Lin Hu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215000 Jiangsu, China
| | - Pinglong Xu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Hai Song
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Long Zhang
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Bin Zhao
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China
| | - Ying-Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and
| | - Zongping Xia
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058 Zhejiang, China,; Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan, China,.
| |
Collapse
|
161
|
Melhuish TA, Kowalczyk I, Manukyan A, Zhang Y, Shah A, Abounader R, Wotton D. Myt1 and Myt1l transcription factors limit proliferation in GBM cells by repressing YAP1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:983-995. [PMID: 30312684 DOI: 10.1016/j.bbagrm.2018.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/05/2018] [Accepted: 10/06/2018] [Indexed: 12/19/2022]
Abstract
Myelin transcription factor 1 (Myt1) and Myt1l (Myt1-like) are zinc finger transcription factors that regulate neuronal differentiation. Reduced Myt1l expression has been implicated in glioblastoma (GBM), and the related St18 was originally identified as a potential tumor suppressor for breast cancer. We previously analyzed changes in gene expression in a human GBM cell line with re-expression of either Myt1 or Myt1l. This revealed largely overlapping gene expression changes, suggesting similar function in these cells. Here we show that re-expression of Myt1 or Myt1l reduces proliferation in two different GBM cell lines, activates gene expression programs associated with neuronal differentiation, and limits expression of proliferative and epithelial to mesenchymal transition gene-sets. Consistent with this, expression of both MYT1 and MYT1L is lower in more aggressive glioma sub-types. Examination of the gene expression changes in cells expressing Myt1 or Myt1l suggests that both repress expression of the YAP1 transcriptional coactivator, which functions primarily in the Hippo signaling pathway. Expression of YAP1 and its target genes is reduced in Myt-expressing cells, and there is an inverse correlation between YAP1 and MYT1/MYT1L expression in human brain cancer datasets. Proliferation of GBM cell lines is reduced by lowering YAP1 expression and increased with YAP1 over-expression, which overcomes the anti-proliferative effect of Myt1/Myt1l expression. Finally we show that reducing YAP1 expression in a GBM cell line slows the growth of orthotopic tumor xenografts. Together, our data suggest that Myt1 and Myt1l directly repress expression of YAP1, a protein which promotes proliferation and GBM growth.
Collapse
Affiliation(s)
- Tiffany A Melhuish
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; Center for Cell Signaling, University of Virginia, Charlottesville, USA
| | - Izabela Kowalczyk
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; Center for Cell Signaling, University of Virginia, Charlottesville, USA
| | - Arkadi Manukyan
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; Center for Cell Signaling, University of Virginia, Charlottesville, USA
| | - Ying Zhang
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Anant Shah
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; Center for Cell Signaling, University of Virginia, Charlottesville, USA
| | - Roger Abounader
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - David Wotton
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; Center for Cell Signaling, University of Virginia, Charlottesville, USA.
| |
Collapse
|
162
|
Shi X, Liu Z, Liu Z, Feng X, Hua F, Hu X, Wang B, Lu K, Nie F. Long noncoding RNA PCAT6 functions as an oncogene by binding to EZH2 and suppressing LATS2 in non-small-cell lung cancer. EBioMedicine 2018; 37:177-187. [PMID: 30314898 PMCID: PMC6286630 DOI: 10.1016/j.ebiom.2018.10.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/28/2022] Open
Abstract
Background NSCLC (non-small-cell lung cancer) is the leading cause of cancer-related mortality worldwide. Both epigenetic and genetic changes contribute to the initiation, development and metastasis of NSCLC. Recently, accumulating data have begun to support the notion that long noncoding RNAs (lncRNAs) function as new crucial regulators of diverse biological processes, including proliferation, apoptosis and metastasis, and play crucial roles in tumorigenesis. Nevertheless, further study is warranted to comprehensively determine lncRNAs' functions and potential mechanism. Methods In this study, we performed a comprehensive analysis of the lncRNA expression profile of NSCLC using data from TCGA and Gene Expression Omnibus (GEO). PCAT6 expression level in a cohort of 60 pairs of NSCLC tissues using quantitative real-time PCR (qRT-PCR). Additionally, Loss-of-function assays and gain-of-function assays were used to assess the role of PCAT6 in promoting NSCLC progression. Tumor formation assay in a nude mouse model was performed to verity the role of PCAT6 in NSCLC in vivo. Meanwhile, RIP, ChIP, resue experiment and western blot assays were used to highlights the potential molecular mechanism of PCAT6 in NSCLC. Findings We identified that an oncogene, PCAT6, was upregulated in NSCLC, and this upregulation was verified in a cohort of 60 pairs of NSCLC tissues. Additionally, the expression level of PCAT6 was correlated with tumor size (P = .036), lymph node metastasis (P = .029) and TNM stage (P = .038). Loss-of-function and gain-of-function assays were used to assess the role of PCAT6 in promoting NSCLC progression. The results revealed that PCAT6 knockdown mitigated NSCLC cell growth by inducing G1-phase cell cycle arrest and apoptosis in vitro and in vivo. Whereas, PCAT6 overexpression could promoted tumor cell growth. Meanwhile, PCAT6 additionally promoted NSCLC cell migration and invasion. Furthermore, mechanistic investigation demonstrated that the oncogenic activity of PCAT6 is partially attributable to its repression of LATS2 via association with the epigenetic repressor EZH2 (Enhancer of zeste homolog 2). Overall, our study highlights the essential role of PCAT6 in NSCLC, suggesting that PCAT6 might be a potent therapeutic target for patients with NSCLC.
Collapse
Affiliation(s)
- Xuefei Shi
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Zhili Liu
- Department of Oncology, The Affiliated Jiangyin Hospital of Southeast University Medical College, Wuxi, People's Republic of China
| | - Zhicong Liu
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Xueren Feng
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Feng Hua
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Xixian Hu
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Bin Wang
- Department of Respiratory Medicine, Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China.
| | - Kaihua Lu
- Department of Oncology, First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| | - Fengqi Nie
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| |
Collapse
|
163
|
Wingless Signaling: A Genetic Journey from Morphogenesis to Metastasis. Genetics 2018; 208:1311-1336. [PMID: 29618590 DOI: 10.1534/genetics.117.300157] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
This FlyBook chapter summarizes the history and the current state of our understanding of the Wingless signaling pathway. Wingless, the fly homolog of the mammalian Wnt oncoproteins, plays a central role in pattern generation during development. Much of what we know about the pathway was learned from genetic and molecular experiments in Drosophila melanogaster, and the core pathway works the same way in vertebrates. Like most growth factor pathways, extracellular Wingless/Wnt binds to a cell surface complex to transduce signal across the plasma membrane, triggering a series of intracellular events that lead to transcriptional changes in the nucleus. Unlike most growth factor pathways, the intracellular events regulate the protein stability of a key effector molecule, in this case Armadillo/β-catenin. A number of mysteries remain about how the "destruction complex" destabilizes β-catenin and how this process is inactivated by the ligand-bound receptor complex, so this review of the field can only serve as a snapshot of the work in progress.
Collapse
|
164
|
Abstract
Since the founding of Drosophila genetics by Thomas Hunt Morgan and his colleagues over 100 years ago, the experimental induction of mosaicism has featured prominently in its recognition as an unsurpassed genetic model organism. The use of genetic mosaics has facilitated the discovery of a wide variety of developmental processes, identified specific cell lineages, allowed the study of recessive embryonic lethal mutations, and demonstrated the existence of cell competition. Here, we discuss how genetic mosaicism in Drosophila became an invaluable research tool that revolutionized developmental biology. We describe the prevailing methods used to produce mosaic animals, and highlight advantages and disadvantages of each genetic system. We cover methods ranging from simple "twin-spot" analysis to more sophisticated systems of multicolor labeling.
Collapse
|
165
|
Eales KL, Wilkinson EA, Cruickshank G, Tucker JHR, Tennant DA. Verteporfin selectively kills hypoxic glioma cells through iron-binding and increased production of reactive oxygen species. Sci Rep 2018; 8:14358. [PMID: 30254296 PMCID: PMC6156578 DOI: 10.1038/s41598-018-32727-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022] Open
Abstract
Gliomas are highly malignant brain tumours characterised by extensive areas of poor perfusion which subsequently leads to hypoxia and reduced survival. Therapies that address the hypoxic microenvironment are likely to significantly improve patient outcomes. Verteporfin, a benzoporphyrin-like drug, has been suggested to target the Yes-associated protein (YAP). Increased YAP expression and transcriptional activity has been proposed in other tumour types to promote malignant cell survival and thus YAP-inhibitor, verteporfin, may be predicted to impact glioma cell growth and viability. Due to the extensive hypoxic nature of gliomas, we investigated the effect of hypoxia on YAP expression and found that YAP transcription is increased under these conditions. Treatment of both primary and immortalised glioblastoma cell lines with verteporfin resulted in a significant decrease in viability but strikingly only under hypoxic conditions (1% O2). We discovered that cell death occurs through a YAP-independent mechanism, predominately involving binding of free iron and likely through redox cycling, contributes to production of reactive oxygen species. This results in disruption of normal cellular processes and death in cells already under oxidative stress - such as those in hypoxia. We suggest that through repurposing verteporfin, it represents a novel means of treating highly therapy-resistant, hypoxic cells in glioma.
Collapse
Affiliation(s)
- Katherine L Eales
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Edward A Wilkinson
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Garth Cruickshank
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, Birmingham, UK
| | - James H R Tucker
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
166
|
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
|
167
|
Liu X, Liu K, Li C, Cai J, Huang L, Chen H, Wang H, Zou J, Liu M, Wang K, Tan S, Zhang H. Heat-shock protein B1 upholds the cytoplasm reduced state to inhibit activation of the Hippo pathway in H9c2 cells. J Cell Physiol 2018; 234:5117-5133. [PMID: 30256412 DOI: 10.1002/jcp.27322] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/02/2018] [Indexed: 12/29/2022]
Abstract
Heat-shock protein B1 (HSPB1) is a multifunctional protein that protects against oxidative stress; however, its function in antioxidant pathways remains largely unknown. Here, we sought to determine the roles of HSPB1 in H9c2 cells subjected to oxidative stress. Using nonreducing sodium dodecyl sulfate polyacrylamide gel electrophoresis, we found that increased HSPB1 expression promoted the reduced states of glutathione reductase (GR), peroxiredoxin 1 (Prx1), and thioredoxin 1, whereas knockdown of HSPB1 attenuated these responses following oxidative stress. Increased HSPB1 expression promoted the activation of GR and thioredoxin reductase. Conversely, knockdown of HSPB1 attenuated these responses following oxidative stress. Importantly, overexpression of HSPB1 promoted the complex formation between HSPB1 and oxidized Prx1, leading to dephosphorylation of STE-mammalian STE20-like kinase 1 (MST1) in H9c2 cells exposed to H2 O 2 , whereas downregulation of HSPB1 induced the opposite results. Mechanistically, HSPB1 regulated the Hippo pathway by enhancing the dephosphorylation of MST1, resulting in reduced phosphorylation of LATS1 and Yes-associated protein (YAP). Moreover, HSPB1 regulated YAP-dependent gene expression. Thus, HSPB1 promoted the reduced state of endogenous antioxidant pathways following oxidative stress in H9c2 cells and improved the redox state of the cytoplasm via modulation of the Hippo signaling pathway.
Collapse
Affiliation(s)
- Xiehong Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Ke Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Caiyan Li
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Jiaodi Cai
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Li Huang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Huan Chen
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Hao Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Jiang Zou
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Meidong Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Sipin Tan
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| | - Huali Zhang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| |
Collapse
|
168
|
Kim HB, Myung SJ. Clinical implications of the Hippo-YAP pathway in multiple cancer contexts. BMB Rep 2018; 51:119-125. [PMID: 29366445 PMCID: PMC5882218 DOI: 10.5483/bmbrep.2018.51.3.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 12/22/2022] Open
Abstract
The Hippo pathway plays prominent and widespread roles in various forms of human carcinogenesis. Specifically, the Yes-associated protein (YAP), a downstream effector of the Hippo pathway, can lead to excessive cell proliferation and the inhibition of apoptosis, resulting in tumorigenesis. It was reported that the YAP is strongly elevated in multiple types of human malignancies such as breast, lung, small intestine, colon, and liver cancers. Recent work indicates that, surprisingly, Hippo signaling components' (SAV1, MST1/2, Lats1/2) mutations are virtually absent in human cancer, rendering this signaling an unlikely candidate to explain the vigorous activation of the YAP in most, if not all human tumors and an activated YAP promotes the resistance to RAF-, MAPK/ERK Kinase (MEK)-, and Epidermal growth factor receptor (EGFR)-targeted inhibitor therapy. The analysis of YAP expressions can facilitate the identification of patients who respond better to an anti-cancer drug treatment comprising RAF-, MEK-, and EGFR-targeted inhibitors. The prominence of YAP for those aspects of cancer biology denotes that these factors are ideal targets for the development of anti-cancer medications. Therefore, our report strongly indicates that the YAP is of potential prognostic utility and druggability in various human cancers. [BMB Reports 2018; 51(3): 119-125].
Collapse
Affiliation(s)
- Han-Byul Kim
- LG Chem, Department of Life Sciences, R&D Park, Seoul 07796, Korea
| | - Seung-Jae Myung
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul 05505; Department of Gastroenterology and Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
169
|
Abstract
Organ growth is fundamental to animal development. One of major mechanisms for growth control is mediated by the conserved Hippo signaling pathway initially identified in Drosophila. The core of this pathway in Drosophila consists of a cascade of protein kinases Hippo and Warts that negatively regulate transcriptional coactivator Yorkie (Yki). Activation of Yki promotes cell survival and proliferation to induce organ growth. A key issue in Hippo signaling is to understand how core kinase cascade is activated. Activation of Hippo kinase cascade is regulated in the upstream by at least two transmembrane proteins Crumbs and Fat that act in parallel. These membrane proteins interact with additional factors such as FERM-domain proteins Expanded and Merlin to modulate subcellular localization and function of the Hippo kinase cascade. Hippo signaling is also influenced by cytoskeletal networks and cell tension in epithelia of developing organs. These upstream events in the regulation of Hippo signaling are only partially understood. This review focuses on our current understanding of some upstream processes involved in Hippo signaling in developing Drosophila organs.
Collapse
Affiliation(s)
- Kwang-Wook Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
170
|
Abstract
How the organ size is adjusted to the proper size during development and how organs know that they reach the original size during regeneration remain long-standing questions. Based on studies using multiple model organisms and approaches for over 20 years, a consensus has been established that the Hippo pathway plays crucial roles in controlling organ size and maintaining tissue homeostasis. Given the significance of these processes, the dysregulation of the Hippo pathway has also implicated various diseases, such as tissue degeneration and cancer. By regulating the downstream transcriptional coactivators YAP and TAZ, the Hippo pathway coordinates cell proliferation and apoptosis in response to a variety of signals including cell contact inhibition, polarity, mechanical sensation and soluble factors. Since the core components and their functions of the Hippo pathway are evolutionarily conserved, this pathway serves as a global regulator of organ size control. Therefore, further investigation of the regulatory mechanisms will provide physiological insights to better understand tissue homeostasis. In this review, the historical developments and current understandings of the regulatory mechanism of Hippo signaling pathway are discussed.
Collapse
Affiliation(s)
- Wantae Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Eek-Hoon Jho
- Departement of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
171
|
Liu S, Martin JF. The regulation and function of the Hippo pathway in heart regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e335. [PMID: 30169913 DOI: 10.1002/wdev.335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/30/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Heart failure caused by cardiomyocyte loss and fibrosis is a leading cause of death worldwide. Although current treatments for heart failure such as heart transplantation and left ventricular assist device implantation have obvious value, new approaches are needed. Endogenous adult cardiomyocyte renewal is measurable but inefficient and inadequate in response to extensive acute heart damage. Stimulating self-renewal of endogenous cardiomyocytes holds great promise for heart repair. Uncovering the genetic mechanisms underlying cardiomyocyte renewal is a critical step in developing new approaches to repairing the heart. Recent studies have revealed that the inhibition of the Hippo pathway is sufficient to promote the proliferation of endogenous cardiomyocytes, indicating that the manipulation of the Hippo pathway in the heart may be a promising treatment for heart failure in the future. We summarize recent findings that have shed light on the function of the Hippo pathway in heart regeneration. We also discuss the mechanisms by which Hippo pathway inhibition promotes heart regeneration and how the Hippo pathway responds to different types of injury or stress during the regenerative process. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
172
|
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are two homologous transcriptional coactivators that promote cell proliferation, stem cell maintenance, and tissue homeostasis. Under favorable conditions, YAP and TAZ are active to promote cell growth through a transcriptional program mediated by the TEAD family transcription factors. Given the indispensability of cellular energy and metabolites for survival and growth, YAP and TAZ are inhibited when energy level is low. Indeed, glucose, fatty acids, hormones, and other metabolic factors have been recently revealed to regulate YAP and TAZ. Conversely, YAP and TAZ are also involved in metabolism regulation, such as to promote glycolysis, lipogenesis, and glutaminolysis, suggesting YAP and TAZ as emerging nodes in coordinating nutrient availability with cell growth and tissue homeostasis. In this Review, we summarize recent findings and provide a current overview of YAP and TAZ in metabolism by focusing on the role of YAP and TAZ as integrators for metabolic cues and cell growth.
Collapse
Affiliation(s)
- Ja Hyun Koo
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
173
|
Xu J, Vanderzalm PJ, Ludwig M, Su T, Tokamov SA, Fehon RG. Yorkie Functions at the Cell Cortex to Promote Myosin Activation in a Non-transcriptional Manner. Dev Cell 2018; 46:271-284.e5. [PMID: 30032991 PMCID: PMC6086586 DOI: 10.1016/j.devcel.2018.06.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
The Hippo signaling pathway is an evolutionarily conserved mechanism that controls organ size in animals. Yorkie is well known as a transcriptional co-activator that functions downstream of the Hippo pathway to positively regulate transcription of genes that promote tissue growth. Recent studies have shown that increased myosin activity activates both Yorkie and its vertebrate orthologue YAP, resulting in increased nuclear localization and tissue growth. Here we show that Yorkie also can accumulate at the cell cortex in the apical junctional region. Moreover, Yorkie functions at the cortex to promote activation of myosin through a myosin regulatory light chain kinase, Stretchin-Mlck. This Yorkie function is not dependent on its transcriptional activity and is required for larval and adult tissues to achieve appropriate size. Based on these results, we suggest that Yorkie functions in a feedforward "amplifier" loop that promotes myosin activation, and thereby greater Yorkie activity, in response to tension.
Collapse
Affiliation(s)
- Jiajie Xu
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Pamela J Vanderzalm
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biology, John Carroll University, University Heights, OH 44118, USA
| | - Michael Ludwig
- Department of Ecology and Evolutionary Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Ting Su
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Sherzod A Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
174
|
A comprehensive evaluation of Hippo pathway silencing in sarcomas. Oncotarget 2018; 9:31620-31636. [PMID: 30167083 PMCID: PMC6114978 DOI: 10.18632/oncotarget.25824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/12/2018] [Indexed: 01/01/2023] Open
Abstract
TAZ and YAP are transcriptional coactivators negatively regulated by the Hippo pathway that have emerged as key oncoproteins in several cancers including sarcomas. We hypothesized that loss of expression of the Hippo kinases might be a mechanism of activating TAZ and YAP. By immunohistochemistry, TAZ/YAP activated clinical sarcoma samples demonstrated loss of MST1 (47%), MST2 (26%), LATS1 (19%), and LATS2 (27%). Western blot similarly demonstrated loss of MST1 (58%), MST2 (25%), and LATS2 (17%). Treatment with MG132 demonstrated an accumulation of MST2 in 25% of sarcoma cell lines, indicating that proteosomal degradation regulates MST2 expression. qRT-PCR in sarcoma cell lines demonstrated loss of expression of the Hippo kinases at the RNA level, most pronounced in MST1 (42%) and MST2 (25%). 5-azacytidine treatment in sarcoma cell lines modestly reversed expression of predominantly MST1 (8%) and MST2 (17%), indicating CpG island hypermethylation can silence expression of MST1 and MST2. Trichostatin A treatment reversed expression of MST1 (58%) and MST2 (67%), indicating histone deacetylation also plays a role in silencing expression of MST1 and MST2. Loss of expression of the Hippo kinases is frequent in sarcomas and is due to a variety of mechanisms including regulation at the post-translational level and epigenetic silencing.
Collapse
|
175
|
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
|
176
|
Moon S, Yeon Park S, Woo Park H. Regulation of the Hippo pathway in cancer biology. Cell Mol Life Sci 2018; 75:2303-2319. [PMID: 29602952 PMCID: PMC11105795 DOI: 10.1007/s00018-018-2804-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/17/2018] [Accepted: 03/22/2018] [Indexed: 01/23/2023]
Abstract
The Hippo tumor suppressor pathway, which is well conserved from Drosophila to humans, has emerged as the master regulator of organ size, as well as major cellular properties, such as cell proliferation, survival, stemness, and tissue homeostasis. The biological significance and deregulation of the Hippo pathway in tumorigenesis have received a surge of interest in the past decade. In the current review, we present the major discoveries that made substantial contributions to our understanding of the Hippo pathway and discuss how Hippo pathway components contribute to cellular signaling, physiology, and their potential implications in anticancer therapeutics.
Collapse
Affiliation(s)
- Sungho Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - So Yeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
177
|
de Vreede G, Morrison HA, Houser AM, Boileau RM, Andersen D, Colombani J, Bilder D. A Drosophila Tumor Suppressor Gene Prevents Tonic TNF Signaling through Receptor N-Glycosylation. Dev Cell 2018; 45:595-605.e4. [PMID: 29870719 PMCID: PMC5995582 DOI: 10.1016/j.devcel.2018.05.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/17/2018] [Accepted: 05/07/2018] [Indexed: 01/18/2023]
Abstract
Drosophila tumor suppressor genes have revealed molecular pathways that control tissue growth, but mechanisms that regulate mitogenic signaling are far from understood. Here we report that the Drosophila TSG tumorous imaginal discs (tid), whose phenotypes were previously attributed to mutations in a DnaJ-like chaperone, are in fact driven by the loss of the N-linked glycosylation pathway component ALG3. tid/alg3 imaginal discs display tissue growth and architecture defects that share characteristics of both neoplastic and hyperplastic mutants. Tumorous growth is driven by inhibited Hippo signaling, induced by excess Jun N-terminal kinase (JNK) activity. We show that ectopic JNK activation is caused by aberrant glycosylation of a single protein, the fly tumor necrosis factor (TNF) receptor homolog, which results in increased binding to the continually circulating TNF. Our results suggest that N-linked glycosylation sets the threshold of TNF receptor signaling by modifying ligand-receptor interactions and that cells may alter this modification to respond appropriately to physiological cues.
Collapse
Affiliation(s)
- Geert de Vreede
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Holly A Morrison
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Alexandra M Houser
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Ryan M Boileau
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Ditte Andersen
- University Nice Sophia Antipolis, CNRS, Inserm, iBV, Nice 06108, France
| | - Julien Colombani
- University Nice Sophia Antipolis, CNRS, Inserm, iBV, Nice 06108, France
| | - David Bilder
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
178
|
|
179
|
Serpin Facilitates Tumor-Suppressive Cell Competition by Blocking Toll-Mediated Yki Activation in Drosophila. Curr Biol 2018; 28:1756-1767.e6. [DOI: 10.1016/j.cub.2018.04.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/06/2018] [Accepted: 04/05/2018] [Indexed: 11/23/2022]
|
180
|
Artap S, Manderfield LJ, Smith CL, Poleshko A, Aghajanian H, See K, Li L, Jain R, Epstein JA. Endocardial Hippo signaling regulates myocardial growth and cardiogenesis. Dev Biol 2018; 440:22-30. [PMID: 29727635 DOI: 10.1016/j.ydbio.2018.04.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/19/2018] [Accepted: 04/19/2018] [Indexed: 01/17/2023]
Abstract
The Hippo signaling pathway has been implicated in control of cell and organ size, proliferation, and endothelial-mesenchymal transformation. This pathway impacts upon two partially redundant transcription cofactors, Yap and Taz, that interact with other factors, including members of the Tead family, to affect expression of downstream genes. Yap and Taz have been shown to regulate, in a cell-autonomous manner, myocardial proliferation, myocardial hypertrophy, regenerative potential, and overall size of the heart. Here, we show that Yap and Taz also play an instructive, non-cell-autonomous role in the endocardium of the developing heart to regulate myocardial growth through release of the paracrine factor, neuregulin. Without endocardial Yap and Taz, myocardial growth is impaired causing early post-natal lethality. Thus, the Hippo signaling pathway regulates cell size via both cell-autonomous and non-cell-autonomous mechanisms. Furthermore, these data suggest that Hippo may regulate organ size via a sensing and paracrine function in endothelial cells.
Collapse
Affiliation(s)
- Stanley Artap
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren J Manderfield
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cheryl L Smith
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrey Poleshko
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haig Aghajanian
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelvin See
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li Li
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, The Cardiovascular Institute and the Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
181
|
Abstract
The Hippo signal transduction pathway is an important regulator of organ growth and cell differentiation, and its deregulation contributes to the development of cancer. The activity of the Hippo pathway is strongly dependent on cell junctions, cellular architecture, and the mechanical properties of the microenvironment. In this review, we discuss recent advances in our understanding of how cell junctions transduce signals from the microenvironment and control the activity of the Hippo pathway. We also discuss how these mechanisms may control organ growth during development and regeneration, and how defects in them deregulate Hippo signaling in cancer cells.
Collapse
Affiliation(s)
- Ruchan Karaman
- VIB Center for Cancer Biology, University of Leuven, 3000 Leuven, Belgium.,Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| | - Georg Halder
- VIB Center for Cancer Biology, University of Leuven, 3000 Leuven, Belgium.,Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|
182
|
Cho YS, Zhu J, Li S, Wang B, Han Y, Jiang J. Regulation of Yki/Yap subcellular localization and Hpo signaling by a nuclear kinase PRP4K. Nat Commun 2018; 9:1657. [PMID: 29695716 PMCID: PMC5916879 DOI: 10.1038/s41467-018-04090-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 04/03/2018] [Indexed: 12/23/2022] Open
Abstract
Hippo (Hpo) signaling pathway controls tissue growth by regulating the subcellular localization of Yorkie (Yki)/Yap via a cytoplasmic kinase cassette containing an upstream kinase Hpo/MST1/2 and a downstream kinase Warts (Wts)/Lats1/2. Here we show that PRP4K, a kinase involved in mRNA splicing, phosphorylates Yki/Yap in the nucleus to prevent its nuclear accumulation and restrict Hpo pathway target gene expression. PRP4K inactivation accelerates whereas excessive PRP4K inhibits Yki-driven tissue overgrowth. PRP4K phosphorylates a subset of Wts/Lats1/2 sites on Yki/Yap to inhibit the binding of Yki/Yap to the Scalloped (Sd)/TEAD transcription factor and exclude Yki/Yap nuclear localization depending on nuclear export. Furthermore, PRP4K inhibits proliferation and invasiveness of cultured breast cancer cells and its high expression correlates with good prognosis in breast cancer patients. Our study unravels an unanticipated layer of Hpo pathway regulation and suggests that PRP4K-mediated Yki/Yap phosphorylation in the nucleus provides a fail-safe mechanism to restrict aberrant pathway activation. The Hippo signaling pathway controls tissue growth by regulating the subcellular localization of Yorkie /Yap. Here the authors show that PRP4K, a kinase involved in mRNA splicing, phosphorylates Yki/Yap in the nucleus, which prevents its nuclear accumulation and inhibits Hippo signaling.
Collapse
Affiliation(s)
- Yong Suk Cho
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Jian Zhu
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.,Henan Key Laboratory of immunology and targeted therapy, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, People's Republic of China
| | - Shuangxi Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Bing Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA. .,Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, TX, 75390, Dallas, USA.
| |
Collapse
|
183
|
Warren JSA, Xiao Y, Lamar JM. YAP/TAZ Activation as a Target for Treating Metastatic Cancer. Cancers (Basel) 2018; 10:cancers10040115. [PMID: 29642615 PMCID: PMC5923370 DOI: 10.3390/cancers10040115] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ) have both emerged as important drivers of cancer progression and metastasis. YAP and TAZ are often upregulated or nuclear localized in aggressive human cancers. There is abundant experimental evidence demonstrating that YAP or TAZ activation promotes cancer formation, tumor progression, and metastasis. In this review we summarize the evidence linking YAP/TAZ activation to metastasis, and discuss the roles of YAP and TAZ during each step of the metastatic cascade. Collectively, this evidence strongly suggests that inappropriate YAP or TAZ activity plays a causal role in cancer, and that targeting aberrant YAP/TAZ activation is a promising strategy for the treatment of metastatic disease. To this end, we also discuss several potential strategies for inhibiting YAP/TAZ activation in cancer and the challenges each strategy poses.
Collapse
Affiliation(s)
- Janine S A Warren
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
184
|
Taha Z, Janse van Rensburg HJ, Yang X. The Hippo Pathway: Immunity and Cancer. Cancers (Basel) 2018; 10:cancers10040094. [PMID: 29597279 PMCID: PMC5923349 DOI: 10.3390/cancers10040094] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Since its discovery, the Hippo pathway has emerged as a central signaling network in mammalian cells. Canonical signaling through the Hippo pathway core components (MST1/2, LATS1/2, YAP and TAZ) is important for development and tissue homeostasis while aberrant signaling through the Hippo pathway has been implicated in multiple pathologies, including cancer. Recent studies have uncovered new roles for the Hippo pathway in immunology. In this review, we summarize the mechanisms by which Hippo signaling in pathogen-infected or neoplastic cells affects the activities of immune cells that respond to these threats. We further discuss how Hippo signaling functions as part of an immune response. Finally, we review how immune cell-intrinsic Hippo signaling modulates the development/function of leukocytes and propose directions for future work.
Collapse
Affiliation(s)
- Zaid Taha
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.
| | | | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
185
|
Holden JK, Cunningham CN. Targeting the Hippo Pathway and Cancer through the TEAD Family of Transcription Factors. Cancers (Basel) 2018; 10:cancers10030081. [PMID: 29558384 PMCID: PMC5876656 DOI: 10.3390/cancers10030081] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/11/2022] Open
Abstract
The Hippo pathway is a critical transcriptional signaling pathway that regulates cell growth, proliferation and organ development. The transcriptional enhanced associate domain (TEAD) protein family consists of four paralogous transcription factors that function to modulate gene expression in response to the Hippo signaling pathway. Transcriptional activation of these proteins occurs upon binding to the co-activator YAP/TAZ whose entry into the nucleus is regulated by Lats1/2 kinase. In recent years, it has become apparent that the dysregulation and/or overexpression of Hippo pathway effectors is implicated in a wide range of cancers, including prostate, gastric and liver cancer. A large body of work has been dedicated to understanding the therapeutic potential of modulating the phosphorylation and localization of YAP/TAZ. However, YAP/TAZ are considered to be natively unfolded and may be intractable as drug targets. Therefore, TEAD proteins present themselves as an excellent therapeutic target for intervention of the Hippo pathway. This review summarizes the functional role of TEAD proteins in cancer and assesses the therapeutic potential of antagonizing TEAD function in vivo.
Collapse
Affiliation(s)
- Jeffrey K Holden
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, CA 94080, USA.
| | - Christian N Cunningham
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
186
|
Abstract
The Hippo pathway is a central regulator of tissue development and homeostasis, and has been reported to have a role during vascular development. Here we develop a bioluminescence-based biosensor that monitors the activity of the Hippo core component LATS kinase. Using this biosensor and a library of small molecule kinase inhibitors, we perform a screen for kinases modulating LATS activity and identify VEGFR as an upstream regulator of the Hippo pathway. We find that VEGFR activation by VEGF triggers PI3K/MAPK signaling, which subsequently inhibits LATS and activates the Hippo effectors YAP and TAZ. We further show that the Hippo pathway is a critical mediator of VEGF-induced angiogenesis and tumor vasculogenic mimicry. Thus, our work offers a biosensor tool for the study of the Hippo pathway and suggests a role for Hippo signaling in regulating blood vessel formation in physiological and pathological settings. The Hippo pathway is a major orchestrator of organ development and homeostasis. Here Azad and colleagues develop a biosensor to monitor the activity of the Hippo pathway component LATS and identify VEGF signalling as an upstream regulator of LATS, supporting a role for Hippo signalling during angiogenesis.
Collapse
|
187
|
Cairns L, Tran T, Fowl BH, Patterson A, Kim YJ, Bothner B, Kavran JM. Salvador has an extended SARAH domain that mediates binding to Hippo kinase. J Biol Chem 2018. [PMID: 29519817 DOI: 10.1074/jbc.ra117.000923] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Hippo pathway controls cell proliferation and differentiation through the precisely tuned activity of a core kinase cassette. The activity of Hippo kinase is modulated by interactions between its C-terminal coiled-coil, termed the SARAH domain, and the SARAH domains of either dRassF or Salvador. Here, we wanted to understand the molecular basis of SARAH domain-mediated interactions and their influence on Hippo kinase activity. We focused on Salvador, a positive effector of Hippo activity and the least well-characterized SARAH domain-containing protein. We determined the crystal structure of a complex between Salvador and Hippo SARAH domains from Drosophila This structure provided insight into the organization of the Salvador SARAH domain including a folded N-terminal extension that expands the binding interface with Hippo SARAH domain. We also found that this extension improves the solubility of the Salvador SARAH domain, enhances binding to Hippo, and is unique to Salvador. We therefore suggest expanding the definition of the Salvador SARAH domain to include this extended region. The heterodimeric assembly observed in the crystal was confirmed by cross-linked MS and provided a structural basis for the mutually exclusive interactions of Hippo with either dRassF or Salvador. Of note, Salvador influenced the kinase activity of Mst2, the mammalian Hippo homolog. In co-transfected HEK293T cells, human Salvador increased the levels of Mst2 autophosphorylation and Mst2-mediated phosphorylation of select substrates, whereas Salvador SARAH domain inhibited Mst2 autophosphorylation in vitro These results suggest Salvador enhances the effects of Hippo kinase activity at multiple points in the Hippo pathway.
Collapse
Affiliation(s)
- Leah Cairns
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Thao Tran
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Brendan H Fowl
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Angela Patterson
- the Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, and
| | - Yoo Jin Kim
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Brian Bothner
- the Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, and
| | - Jennifer M Kavran
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215, .,the Departments of Biophysics and Biophysical Chemistry, School of Medicine, and.,Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 20215
| |
Collapse
|
188
|
BmYki is transcribed into four functional splicing isoforms in the silk glands of the silkworm Bombyx mori. Gene 2018; 646:39-46. [DOI: 10.1016/j.gene.2017.12.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
|
189
|
Khanal P, Jia Z, Yang X. Cysteine residues are essential for dimerization of Hippo pathway components YAP2L and TAZ. Sci Rep 2018; 8:3485. [PMID: 29472569 PMCID: PMC5823869 DOI: 10.1038/s41598-018-21828-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/12/2018] [Indexed: 12/24/2022] Open
Abstract
Hippo signalling pathway is an emerging signalling pathway that plays important roles in organ size control, tumorigenesis, metastasis, stress response, apoptosis, stem cell differentiation and renewal during development and tissue homeostasis. Recent studies reported that human serine/threonine protein kinase, Mst1, a core component of the Hippo pathway can be activated through formation of homodimer. However, it is still unclear whether or not other components of the Hippo pathway are also regulated through dimerization. Here we provide the first evidence that Hippo components and oncoprotein YAP2L and TAZ can form homodimer in vitro and in vivo by forming disulphide bond through cysteine residue(s). We have also shown that the homodimers of YAP2L/TAZ are more stable and showed more oncogenic behaviour than their corresponding monomers as revealed by colony formation and cell transformation assay. Since cysteine post-translational regulation plays important roles in redox signalling, tumorigenesis and drug resistance, further studies on the functional effect of this dimerization through post-translational modulation of cysteine residues in YAP2L/TAZ will provide a significant contribution to our understanding of the roles of YAP2L/TAZ in cancer development and therapy.
Collapse
Affiliation(s)
- Prem Khanal
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Zongchao Jia
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada.
| |
Collapse
|
190
|
Elbediwy A, Thompson BJ. Evolution of mechanotransduction via YAP/TAZ in animal epithelia. Curr Opin Cell Biol 2018; 51:117-123. [PMID: 29477107 DOI: 10.1016/j.ceb.2018.02.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Abstract
Mechanical stretch forces can control the growth of epithelial tissues such as mammalian skin, whose surface area is precisely coordinated with body size. In skin keratinocytes cultured in vitro, mechanical forces acting via Integrin adhesions and the actin cytoskeleton have been shown to induce nuclear translocation of YAP/TAZ co-activators to induce cell proliferation. Furthermore, conditional knockouts of both YAP (also called YAP1) and TAZ (also called WWTR1) in mouse skin resemble the phenotype of skin-specific loss of Integrin beta1 (ITGB1), indicating that this signalling mechanism is important in vivo. Curiously, Integrins are dispensable in Drosophila to activate the sole YAP/TAZ homolog Yorkie (Yki), which has lost the C-terminal PDZ-binding motif needed to promote nuclear localization of YAP/TAZ in mammalian cells. Differences in the structure of the epidermis between deuterostomes (e.g.: stratified squamous skin of mammals) and protostomes (e.g.: monolayered columnar epidermis of Drosophila) may explain this evolutionary divergence. Monolayered columnar epithelia feature a well-differentiated apical membrane domain, where proteins such as Crumbs, Expanded, Merlin and Kibra activate the Hippo pathway to repress Drosophila Yki. Stratified squamous epithelia lack an apical domain and thus depend primarily on basal Integrin adhesions to activate YAP/TAZ in basal layer stem cells via multiple postulated signalling mechanisms. Finally, YAP and TAZ retain the ability to sense the apical domain in the columnar epithelial cells lining internal organs such as the lung bronchus, where YAP/TAZ localize to the nucleus in proliferating basal layer stem cells but translocate to the cytoplasm in differentiated columnar cells.
Collapse
Affiliation(s)
- Ahmed Elbediwy
- Epithelial Biology Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, United Kingdom
| | - Barry J Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, United Kingdom.
| |
Collapse
|
191
|
Yu J, Pan D. Validating upstream regulators of Yorkie activity in Hippo signaling through scalloped-based genetic epistasis. Development 2018; 145:145/4/dev157545. [PMID: 29467233 DOI: 10.1242/dev.157545] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 01/19/2018] [Indexed: 12/19/2022]
Abstract
Genetic studies in Drosophila have been instrumental in characterizing the Hippo pathway, which converges on the co-activator Yorkie to regulate target gene transcription. A routinely used strategy to interrogate upstream regulators of Yorkie involves the examination of selected Hippo target genes upon loss or gain of function of a suspected pathway regulator. A caveat with this strategy is that aberrant expression of a given Hippo target per se does not distinguish whether it is caused by changes in Yorkie or Yorkie-independent inputs converging on the same target gene. Building on previous findings that the DNA-binding transcription factor Scalloped mediates both Yorkie overexpression and loss-of-function phenotypes yet is itself dispensable for normal eye development, we describe a simple strategy to distinguish these possibilities by analyzing double-mutant clones of scalloped and a suspected Yorkie regulator. We provide proof of principle that this strategy can be used effectively to validate canonical Yorkie regulators and to exclude proteins that impact target expression independent of Yorkie. The described methodology and reagents should facilitate efforts to assess the expanding repertoire of proteins implicated in regulation of Yorkie activity.
Collapse
Affiliation(s)
- Jianzhong Yu
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.,Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| |
Collapse
|
192
|
POSH regulates Hippo signaling through ubiquitin-mediated expanded degradation. Proc Natl Acad Sci U S A 2018; 115:2150-2155. [PMID: 29440430 DOI: 10.1073/pnas.1715165115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The Hippo signaling pathway is a master regulator of organ growth, tissue homeostasis, and tumorigenesis. The activity of the Hippo pathway is controlled by various upstream components, including Expanded (Ex), but the precise molecular mechanism of how Ex is regulated remains poorly understood. Here we identify Plenty of SH3s (POSH), an E3 ubiquitin ligase, as a key component of Hippo signaling in DrosophilaPOSH overexpression synergizes with loss of Kibra to induce overgrowth and up-regulation of Hippo pathway target genes. Furthermore, knockdown of POSH impedes dextran sulfate sodium-induced Yorkie-dependent intestinal stem cell renewal, suggesting a physiological role of POSH in modulating Hippo signaling. Mechanistically, POSH binds to the C-terminal of Ex and is essential for the Crumbs-induced ubiquitination and degradation of Ex. Our findings establish POSH as a crucial regulator that integrates the signal from the cell surface to negatively regulate Ex-mediated Hippo activation in Drosophila.
Collapse
|
193
|
Melucci E, Casini B, Ronchetti L, Pizzuti L, Sperati F, Pallocca M, De Nicola F, Goeman F, Gallo E, Amoreo CA, Sergi D, Terrenato I, Vici P, Di Lauro L, Diodoro MG, Pescarmona E, Barba M, Mazzotta M, Mottolese M, Fanciulli M, Ciliberto G, De Maria R, Buglioni S, Maugeri-Saccà M. Expression of the Hippo transducer TAZ in association with WNT pathway mutations impacts survival outcomes in advanced gastric cancer patients treated with first-line chemotherapy. J Transl Med 2018; 16:22. [PMID: 29402328 PMCID: PMC5800016 DOI: 10.1186/s12967-018-1385-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/11/2018] [Indexed: 01/02/2023] Open
Abstract
Background An extensive crosstalk co-regulates the Hippo and Wnt pathway. Preclinical studies revealed that the Hippo transducers YAP/TAZ mediate a number of oncogenic functions in gastric cancer (GC). Moreover, comprehensive characterization of GC demonstrated that the Wnt pathway is targeted by oncogenic mutations. On this ground, we hypothesized that YAP/TAZ- and Wnt-related biomarkers may predict clinical outcomes in GC patients treated with chemotherapy. Methods In the present study, we included 86 patients with advanced GC treated with first-line chemotherapy in prospective phase II trials or in routine clinical practice. Tissue samples were immunostained to evaluate the expression of YAP/TAZ. Mutational status of key Wnt pathway genes (CTNNB1, APC and FBXW7) was assessed by targeted DNA next-generation sequencing (NGS). Survival curves were estimated and compared by the Kaplan–Meier product-limit method and the log-rank test, respectively. Variables potentially affecting progression-free survival (PFS) were verified in univariate Cox proportional hazard models. The final multivariate Cox models were obtained with variables testing significant at the univariate analysis, and by adjusting for all plausible predictors of the outcome of interest (PFS). Results We observed a significant association between TAZ expression and Wnt mutations (Chi-squared p = 0.008). Combined TAZ expression and Wnt mutations (TAZpos/WNTmut) was more frequently observed in patients with the shortest progression-free survival (negative outliers) (Fisher p = 0.021). Uni-and multivariate Cox regression analyses revealed that patients whose tumors harbored the TAZpos/WNTmut signature had an increased risk of disease progression (univariate Cox: HR 2.27, 95% CI 1.27–4.05, p = 0.006; multivariate Cox: HR 2.73, 95% CI 1.41–5.29, p = 0.003). Finally, the TAZpos/WNTmut signature negatively impacted overall survival. Conclusions Collectively, our findings indicate that the oncogenic YAP/TAZ–Wnt crosstalk may be active in GC, conferring chemoresistant traits that translate into adverse survival outcomes. Electronic supplementary material The online version of this article (10.1186/s12967-018-1385-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisa Melucci
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Beatrice Casini
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Livia Ronchetti
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Francesca Sperati
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Frauke Goeman
- Oncogenomic and Epigenetic Unit, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Enzo Gallo
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Carla Azzurra Amoreo
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Irene Terrenato
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.,Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marco Mazzotta
- Medical Oncology Unit, Policlinico Sant'Andrea, Via Di Grotta Rossa, 1035/1039, 00189, Rome, Italy
| | - Marcella Mottolese
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 10, 00168, Rome, Italy.
| | - Simonetta Buglioni
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy. .,Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
194
|
Carvalho Naves MP, de Morais CR, Silva ACA, Dantas NO, Spanó MA, de Rezende AAA. Assessment of mutagenic, recombinogenic and carcinogenic potential of titanium dioxide nanocristals in somatic cells of Drosophila melanogaster. Food Chem Toxicol 2018; 112:273-281. [DOI: 10.1016/j.fct.2017.12.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 01/02/2023]
|
195
|
Cho SY, Gwak JW, Shin YC, Moon D, Ahn J, Sol HW, Kim S, Kim G, Shin HM, Lee KH, Kim JY, Kim JS. Expression of Hippo pathway genes and their clinical significance in colon adenocarcinoma. Oncol Lett 2018. [PMID: 29541248 PMCID: PMC5835912 DOI: 10.3892/ol.2018.7911] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Yes-associated protein 1 (YAP1) is a transcriptional regulator of the Hippo pathway, which regulates the development and progression of a number of types of cancer, including that of the colon. In the present study, the expression levels of Hippo pathway genes and their clinical significance were investigated in 458 patients with colon adenocarcinoma (COAD), the most frequently diagnosed neoplastic disease globally, using data obtained from The Cancer Genome Atlas database. Notably, mRNA expression of YAP1 was higher in COAD than in other types of gastrointestinal tract cancer. Expression of YAP1 mRNA was higher in COAD than in normal colon samples and was significantly higher in Tumor-Node-Metastasis (TNM) stages III-IV than in stages I-II. YAP1 protein levels, a protein primarily localized in the nucleus, was greater in TNM stages III-IV than in stages I-II. The level of pYAP1, which is inactive and localized in the cytoplasm, was significantly higher in TNM stages III-IV than in stages I-II. However, the YAP1/pYAP1 ratio, which is representative of activity, was higher in TNM stages III-IV than in stages I-II. High mRNA expression of YAP1, TAZ and TEAD4 was associated with a poor prognosis in patients with COAD. Bioinformatics analysis revealed that YAP1 was associated with DNA duplication, cell proliferation and development. Wnt signaling and transforming growth factor-β signaling were significantly higher in the high-YAP1 group, according to data from Gene Set Enrichment Analysis. Taken together, the results indicate that the subcellular distribution of YAP1 and high mRNA expression of YAP1, TAZ and TEAD4 may be associated with poorer overall survival rates in patients with COAD.
Collapse
Affiliation(s)
- Sang Yeon Cho
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jang Wook Gwak
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Yoo Chul Shin
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Daeju Moon
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jihyuok Ahn
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyon Woo Sol
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sungha Kim
- Clinical Research Department, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Gwanghun Kim
- Department of Anatomy, Seoul National University, College of Medicine, Seoul 03080, Republic of Korea
| | - Hyun Mu Shin
- Department of Anatomy, Seoul National University, College of Medicine, Seoul 03080, Republic of Korea
| | - Kyung Ha Lee
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Ji Yeon Kim
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin Soo Kim
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
196
|
Expression of Hippo signaling pathway and Aurora kinase genes in chronic myeloid leukemia. Med Oncol 2018; 35:26. [PMID: 29387948 DOI: 10.1007/s12032-018-1079-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm resulting from clonal expansion of hematopoietic stem cells positive for the Philadelphia chromosome. The CML pathogenesis is associated with expression of the BCR-ABL1 oncogene, which encodes the Bcr-Abl protein with tyrosine kinase activity, promoting the leukemic cell exacerbated myeloproliferation and resistance to apoptosis. CML patients are usually treated with tyrosine kinase inhibitors (TKI), but some of them acquire resistance or are refractory to TKI. Thus, it is still relevant to elucidate the CML pathogenesis and seek new therapeutic targets, such as the Hippo signaling pathway and cell cycle regulatory genes from the Aurora kinase family. The present study quantified the expression level of genes encoding components of the Hippo signaling pathway (LATS1, LATS2, YAP, and TAZ), AURKA and AURKB in CML patients at different stages of the disease, who were resistant or sensitive to imatinib mesylate therapy, and in healthy individuals. The expression levels of the target genes were correlated with the CML Sokal's prognostic score. The most striking results were the LATS2 and AURKA overexpression in CML patients, the overexpression of TAZ and AURKB in CML patients at advanced phases and TAZ in CML IM-resistant. The development of drugs and/or identification of tumor markers for the Hippo signaling pathway and the Aurora kinase family, either alone or in combination, can optimize CML treatment by enhancing the susceptibility of leukemic cells to apoptosis and leading to a better disease prognosis.
Collapse
|
197
|
Verma A, Jing-Song F, Finch-Edmondson ML, Velazquez-Campoy A, Balasegaran S, Sudol M, Sivaraman J. Biophysical studies and NMR structure of YAP2 WW domain - LATS1 PPxY motif complexes reveal the basis of their interaction. Oncotarget 2018; 9:8068-8080. [PMID: 29487715 PMCID: PMC5814282 DOI: 10.18632/oncotarget.23909] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/05/2017] [Indexed: 11/30/2022] Open
Abstract
YES-associated protein (YAP) is a major effector protein of the Hippo tumor suppressor pathway, and is phosphorylated by the serine/threonine kinase LATS. Their binding is mediated by the interaction between WW domains of YAP and PPxY motifs of LATS. Their isoforms, YAP2 and LATS1 contain two WW domains and two PPxY motifs respectively. Here, we report the study of the interaction of these domains both in vitro and in human cell lines, to better understand the mechanism of their binding. We show that there is a reciprocal binding preference of YAP2-WW1 with LATS1-PPxY2, and YAP2-WW2 with LATS1-PPxY1. We solved the NMR structures of these complexes and identified several conserved residues that play a critical role in binding. We further created a YAP2 mutant by swapping the WW domains, and found that YAP2 phosphorylation at S127 by LATS1 is not affected by the spatial configuration of its WW domains. This is likely because the region between the PPxY motifs of LATS1 is unstructured, even upon binding with its partner. Based on our observations, we propose possible models for the interaction between YAP2 and LATS1.
Collapse
Affiliation(s)
- Apoorva Verma
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Fan Jing-Song
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Adrian Velazquez-Campoy
- Institute of Biocomputation and Physics of Complex Systems (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, Universidad de Zaragoza, Spain, Department of Biochemistry and Molecular and Cellular Biology, Universidad de Zaragoza, Fundacion ARAID, Gobierno de Aragon, Spain, Aragon Health Research Institute (IIS Aragon), Universidad de Zaragoza, Zaragoza, Spain
| | - Shanker Balasegaran
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Marius Sudol
- Mechanobiology Institute, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, MD9, Singapore.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research, Singapore
| | - Jayaraman Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore
| |
Collapse
|
198
|
|
199
|
Stepan J, Anderzhanova E, Gassen NC. Hippo Signaling: Emerging Pathway in Stress-Related Psychiatric Disorders? Front Psychiatry 2018; 9:715. [PMID: 30627107 PMCID: PMC6309125 DOI: 10.3389/fpsyt.2018.00715] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/06/2018] [Indexed: 12/20/2022] Open
Abstract
Discovery of the Hippo pathway and its core components has made a significant impact on our progress in the understanding of organ development, tissue homeostasis, and regeneration. Upon diverse extracellular and intracellular stimuli, Hippo signaling regulates stemness, cell proliferation and apoptosis by a well-conserved signaling cascade, and disruption of these systems has been implicated in cancer as well as metabolic and neurodegenerative diseases. The central role of Hippo signaling in cell biology also results in prominent links to stress-regulated pathways. Genetic variations, epigenetically provoked upregulation of Hippo pathway members and dysregulation of cellular processes implicated in learning and memory, are linked to an increased risk of stress-related psychiatric disorders (SRPDs). In this review, we summarize recent findings, supporting the role of Hippo signaling in SRPDs by canonical and non-canonical Hippo pathway interactions.
Collapse
Affiliation(s)
- Jens Stepan
- Department Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elmira Anderzhanova
- Clinic and Polyclinic of Psychiatry and Psychotherapy, Bonn University Clinic, Bonn, Germany
| | - Nils C Gassen
- Clinic and Polyclinic of Psychiatry and Psychotherapy, Bonn University Clinic, Bonn, Germany
| |
Collapse
|
200
|
Fullenkamp CA, Hall SL, Jaber OI, Pakalniskis BL, Savage EC, Savage JM, Ofori-Amanfo GK, Lambertz AM, Ivins SD, Stipp CS, Miller BJ, Milhem MM, Tanas MR. TAZ and YAP are frequently activated oncoproteins in sarcomas. Oncotarget 2017; 7:30094-108. [PMID: 27129148 PMCID: PMC5058666 DOI: 10.18632/oncotarget.8979] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/09/2016] [Indexed: 12/02/2022] Open
Abstract
TAZ (WWTR1) and YAP are transcriptional coactivators and oncoproteins inhibited by the Hippo pathway. Herein we evaluate 159 sarcomas representing the most prevalent sarcoma types by immunohistochemistry for expression and activation (nuclear localization) of TAZ and YAP. We show that 50% of sarcomas demonstrate activation of YAP while 66% of sarcomas demonstrate activated TAZ. Differential activation of TAZ and YAP are identified in various sarcoma types. At an RNA level, expression of WWTR1 or YAP1 predicts overall survival in undifferentiated pleomorphic sarcoma and dedifferentiated liposarcoma. Immunohistochemistry demonstrates that TAZ and YAP expression and activation are positively correlated with grade in the well-differentiated liposarcoma to dedifferentiated liposarcoma tumor progression sequence as well as conventional chondrosarcomas. TAZ and YAP are constitutively activated oncoproteins in sarcoma cell lines. Knock-down of TAZ and YAP demonstrate differential activity for the two proteins. Verteporfin decreases colony formation in soft agar as well as CTGF expression in sarcoma cell lines harboring activated TAZ and YAP.
Collapse
Affiliation(s)
| | - Sarah L Hall
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Omar I Jaber
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | | | - Erica C Savage
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | | | | | | | | | | | - Benjamin J Miller
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, USA
| | - Mohammed M Milhem
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Munir R Tanas
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|