51
|
Wang M, Xu T, Feng W, Liu J, Wang Z. Advances in Understanding the LncRNA-Mediated Regulation of the Hippo Pathway in Cancer. Onco Targets Ther 2021; 14:2397-2415. [PMID: 33854336 PMCID: PMC8039192 DOI: 10.2147/ott.s283157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are a class of RNA molecules that are longer than 200 nucleotides and cannot encode proteins. Over the past decade, lncRNAs have been defined as regulatory elements of multiple biological processes, and their aberrant expression contributes to the development and progression of various malignancies. Recent studies have shown that lncRNAs are involved in key cancer-related signaling pathways, including the Hippo signaling pathway, which plays a prominent role in controlling organ size and tissue homeostasis by regulating cell proliferation, apoptosis, and differentiation. However, dysregulation of this pathway is associated with pathological conditions, especially cancer. Accumulating evidence has revealed that lncRNAs can modulate the Hippo signaling pathway in cancer. In this review, we elaborate on the role of the Hippo signaling pathway and the advances in the understanding of its lncRNA-mediated regulation in cancer. This review provides additional insight into carcinogenesis and will be of great clinical value for developing novel early detection and treatment strategies for this deadly disease.
Collapse
Affiliation(s)
- Mengwei Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Tianwei Xu
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wenyan Feng
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Junxia Liu
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Zhaoxia Wang
- Cancer Medical Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
52
|
YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming. Metabolites 2021; 11:metabo11030154. [PMID: 33800464 PMCID: PMC7999074 DOI: 10.3390/metabo11030154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Cell reprogramming can either refer to a direct conversion of a specialized cell into another or to a reversal of a somatic cell into an induced pluripotent stem cell (iPSC). It implies a peculiar modification of the epigenetic asset and gene regulatory networks needed for a new cell, to better fit the new phenotype of the incoming cell type. Cellular reprogramming also implies a metabolic rearrangement, similar to that observed upon tumorigenesis, with a transition from oxidative phosphorylation to aerobic glycolysis. The induction of a reprogramming process requires a nexus of signaling pathways, mixing a range of local and systemic information, and accumulating evidence points to the crucial role exerted by the Hippo pathway components Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ). In this review, we will first provide a synopsis of the Hippo pathway and its function during reprogramming and tissue regeneration, then we introduce the latest knowledge on the interplay between YAP/TAZ and metabolism and, finally, we discuss the possible role of YAP/TAZ in the orchestration of the metabolic switch upon cellular reprogramming.
Collapse
|
53
|
Guo T, Gupta A, Yu J, Granados JZ, Gandhi AY, Evers BM, Iyengar P, Infante RE. LIFR-α-dependent adipocyte signaling in obesity limits adipose expansion contributing to fatty liver disease. iScience 2021; 24:102227. [PMID: 33748712 PMCID: PMC7970148 DOI: 10.1016/j.isci.2021.102227] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/02/2021] [Accepted: 02/19/2021] [Indexed: 01/23/2023] Open
Abstract
The role of chronic adipose inflammation in diet-induced obesity (DIO) and its sequelae including fatty liver disease remains unclear. Leukemia inhibitory factor (LIF) induces JAK-dependent adipocyte lipolysis and altered adipo/cytokine expression, suppressing in vivo adipose expansion in normal and obese mouse models. To characterize LIF receptor (LIFR-α)-dependent cytokine signaling in DIO, we created an adipocyte-specific LIFR knockout mouse model (Adipoq-Cre;LIFRfl/fl). Differentiated adipocytes derived from this model blocked LIF-induced triacylglycerol lipolysis. Adipoq-Cre;LIFRfl/fl mice on a high-fat diet (HFD) displayed reduced adipose STAT3 activation, 50% expansion in adipose, 20% body weight increase, and a 75% reduction in total hepatic triacylglycerides compared with controls. To demonstrate that LIFR-α signals adipocytes through STAT3, we also created an Adipoq-Cre;STAT3fl/fl model that showed similar findings when fed a HFD as Adipoq-Cre;LIFRfl/fl mice. These findings establish the importance of obesity-associated LIFR-α/JAK/STAT3 inflammatory signaling in adipocytes, blocking further adipose expansion in DIO contributing to ectopic liver triacylglyceride accumulation. LIFR-α signaling induces adipocyte lipolysis, restricting adipose expansion in DIO LIFR-α signaling requires STAT3 for adipocyte lipolysis LIFR-α/JAK/STAT3 lipolysis signaling in adipocytes promotes hepatic steatosis
Collapse
Affiliation(s)
- Tong Guo
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5300 Harry Hines Boulevard, Dallas, TX, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arun Gupta
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jinhai Yu
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5300 Harry Hines Boulevard, Dallas, TX, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jorge Z Granados
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5300 Harry Hines Boulevard, Dallas, TX, USA
| | - Aakash Y Gandhi
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5300 Harry Hines Boulevard, Dallas, TX, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Puneeth Iyengar
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5300 Harry Hines Boulevard, Dallas, TX, USA.,Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rodney E Infante
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5300 Harry Hines Boulevard, Dallas, TX, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
54
|
LeBlanc L, Ramirez N, Kim J. Context-dependent roles of YAP/TAZ in stem cell fates and cancer. Cell Mol Life Sci 2021; 78:4201-4219. [PMID: 33582842 PMCID: PMC8164607 DOI: 10.1007/s00018-021-03781-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/30/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
Hippo effectors YAP and TAZ control cell fate and survival through various mechanisms, including transcriptional regulation of key genes. However, much of this research has been marked by conflicting results, as well as controversy over whether YAP and TAZ are redundant. A substantial portion of the discordance stems from their contradictory roles in stem cell self-renewal vs. differentiation and cancer cell survival vs. apoptosis. In this review, we present an overview of the multiple context-dependent functions of YAP and TAZ in regulating cell fate decisions in stem cells and organoids, as well as their mechanisms of controlling programmed cell death pathways in cancer.
Collapse
Affiliation(s)
- Lucy LeBlanc
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Nereida Ramirez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.,Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA. .,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
55
|
Passaro F, De Martino I, Zambelli F, Di Benedetto G, Barbato M, D'Erchia AM, Manzari C, Pesole G, Mutarelli M, Cacchiarelli D, Antonini D, Parisi S, Russo T. YAP contributes to DNA methylation remodeling upon mouse embryonic stem cell differentiation. J Biol Chem 2021; 296:100138. [PMID: 33268382 PMCID: PMC7948423 DOI: 10.1074/jbc.ra120.015896] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/26/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
The Yes-associated protein (YAP), one of the major effectors of the Hippo pathway together with its related protein WW-domain-containing transcription regulator 1 (WWTR1; also known as TAZ), mediates a range of cellular processes from proliferation and death to morphogenesis. YAP and WW-domain-containing transcription regulator 1 (WWTR1; also known as TAZ) regulate a large number of target genes, acting as coactivators of DNA-binding transcription factors or as negative regulators of transcription by interacting with the nucleosome remodeling and histone deacetylase complexes. YAP is expressed in self-renewing embryonic stem cells (ESCs), although it is still debated whether it plays any crucial roles in the control of either stemness or differentiation. Here we show that the transient downregulation of YAP in mouse ESCs perturbs cellular homeostasis, leading to the inability to differentiate properly. Bisulfite genomic sequencing revealed that this transient knockdown caused a genome-wide alteration of the DNA methylation remodeling that takes place during the early steps of differentiation, suggesting that the phenotype we observed might be due to the dysregulation of some of the mechanisms involved in regulation of ESC exit from pluripotency. By gene expression analysis, we identified two molecules that could have a role in the altered genome-wide methylation profile: the long noncoding RNA ephemeron, whose rapid upregulation is crucial for the transition of ESCs into epiblast, and the methyltransferase-like protein Dnmt3l, which, during the embryo development, cooperates with Dnmt3a and Dnmt3b to contribute to the de novo DNA methylation that governs early steps of ESC differentiation. These data suggest a new role for YAP in the governance of the epigenetic dynamics of exit from pluripotency.
Collapse
Affiliation(s)
- Fabiana Passaro
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Italy.
| | - Ilaria De Martino
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Italy
| | - Federico Zambelli
- Department of Biosciences, University of Milano, Italy; Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Italy
| | - Giorgia Di Benedetto
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Italy
| | - Matteo Barbato
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Italy
| | - Anna Maria D'Erchia
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Italy; Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Caterina Manzari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Italy
| | - Graziano Pesole
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Italy; Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Margherita Mutarelli
- Tigem and Department of Translational Medicine, University of Napoli Federico II, Italy
| | - Davide Cacchiarelli
- Tigem and Department of Translational Medicine, University of Napoli Federico II, Italy
| | - Dario Antonini
- Department of Biology, University of Napoli Federico II, Italy
| | - Silvia Parisi
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Italy
| | - Tommaso Russo
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli Federico II, Italy.
| |
Collapse
|
56
|
Xu Y, Liu L, Wang X, Huang Y, Cheng Y, Zhang J, Wang J, Tian Y, Xiong J, Yang Y, Ren B, Wu W, He P, Zhang Y, Zhao B, Wang J, Yu M, Wang J, Jian R, Liu Y, Ruan Y. Identification of novel Taz isoforms and functional comparison in pluripotency maintenance of mouse embryonic stem cells. Gene 2020; 773:145383. [PMID: 33383118 DOI: 10.1016/j.gene.2020.145383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/19/2020] [Accepted: 12/18/2020] [Indexed: 12/29/2022]
Abstract
Alternative splicing (AS) is a key process to expand the diversity of mRNA and protein from the genome and it is crucial for fate determination of embryonic stem cells (ESCs) by encoding isoforms with different functions to regulate the balance between pluripotency maintenance and differentiation. Since the role of the Hippo pathway in ESCs is controversial, there may be novel isoforms of Taz, a key effector of the Hippo pathway, previously unknown to us. Here, we identified three variants of Taz in mESCs. Apart from the canonical Taz1185, there were also two novel variants, Taz402 and Taz1086. We found their structure and subcellular localization to be different, while they could all interact with TEAD2 with similar binding affinities and activate transcription. Under the LIFlow condition, overexpression of them all induced apoptosis and differentiation of mESCs, among which the phenotype of Taz1086 was the most dramatic. Taken together, we discovered novel variants of Taz and compared their structure and functional differences in mESC pluripotency maintenance. These findings will help us to understand the Taz gene and clarify its role in mESC.
Collapse
Affiliation(s)
- Yixiao Xu
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing 400038, China
| | - Lianlian Liu
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Xueyue Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Yuyan Huang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Yuda Cheng
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Junlei Zhang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Jiali Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Yanping Tian
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Jiaxiang Xiong
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Bangqi Ren
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing 400038, China
| | - Wei Wu
- Thoracic Surgery Department, Southwest Hospital, the First Hospital Affiliated to Army Medical University, Chongqing 400038, China
| | - Ping He
- Cardiac Surgery Department, Southwest Hospital, the First Hospital Affiliated to Army Medical University, Chongqing 400038, China
| | - Yue Zhang
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing 400038, China
| | - Binyu Zhao
- Department of Physiology, Army Medical University, Chongqing 400038, China
| | - Jiaqi Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Meng Yu
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Jiangjun Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Rui Jian
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing 400038, China.
| | - Yan Ruan
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
57
|
Buckarma EH, Werneburg NW, Conboy CB, Kabashima A, O'Brien DR, Wang C, Ilyas SI, Smoot RL. The YAP-Interacting Phosphatase SHP2 Can Regulate Transcriptional Coactivity and Modulate Sensitivity to Chemotherapy in Cholangiocarcinoma. Mol Cancer Res 2020; 18:1574-1588. [PMID: 32646966 PMCID: PMC7541657 DOI: 10.1158/1541-7786.mcr-20-0165] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/15/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
The Hippo pathway effector Yes-associated protein (YAP) is localized to the nucleus and transcriptionally active in a number of tumor types, including a majority of human cholangiocarcinomas. YAP activity has been linked to chemotherapy resistance and has been shown to rescue KRAS and BRAF inhibition in RAS/RAF-driven cancers; however, the underlying mechanisms of YAP-mediated chemoresistance have yet to be elucidated. Herein, we report that the tyrosine phosphatase SHP2 directly regulates the activity of YAP by dephosphorylating pYAPY357 even in the setting of RAS/RAF mutations, and that diminished SHP2 phosphatase activity is associated with chemoresistance in cholangiocarcinomas. A screen for YAP-interacting tyrosine phosphatases identified SHP2, and characterization of cholangiocarcinomas cell lines demonstrated an inverse relationship between SHP2 levels and pYAPY357. Human sequencing data demonstrated lower SHP2 levels in cholangiocarcinomas tumors as compared with normal liver. Cell lines with low SHP2 expression and higher levels of pYAPY357 were resistant to gemcitabine and cisplatin. In cholangiocarcinomas cells with high levels of SHP2, pharmacologic inhibition or genetic deletion of SHP2 increased YAPY357 phosphorylation and expression of YAP target genes, including the antiapoptotic regulator MCL1, imparting resistance to gemcitabine and cisplatin. In vivo evaluation of chemotherapy sensitivity demonstrated significant resistance in xenografts with genetic deletion of SHP2, which could be overcome by utilizing an MCL1 inhibitor. IMPLICATIONS: These findings demonstrate a role for SHP2 in regulating YAP activity and chemosensitivity, and suggest that decreased phosphatase activity may be a mechanism of chemoresistance in cholangiocarcinoma via a MCL1-mediated mechanism.
Collapse
Affiliation(s)
| | - Nathan W Werneburg
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Ayano Kabashima
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Daniel R O'Brien
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Chen Wang
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Rory L Smoot
- Department of Surgery, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
58
|
Abstract
Derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming somatic cells to a pluripotent state has revolutionized stem cell research. Ensuing this, various groups have used genetic and non-genetic approaches to generate iPSCs from numerous cell types. However, achieving a pluripotent state in most of the reprogramming studies is marred by serious limitations such as low reprogramming efficiency and slow kinetics. These limitations are mainly due to the presence of potent barriers that exist during reprogramming when a mature cell is coaxed to achieve a pluripotent state. Several studies have revealed that intrinsic factors such as non-optimal stoichiometry of reprogramming factors, specific signaling pathways, cellular senescence, pluripotency-inhibiting transcription factors and microRNAs act as a roadblock. In addition, the epigenetic state of somatic cells and specific epigenetic modifications that occur during reprogramming also remarkably impede the generation of iPSCs. In this review, we present a comprehensive overview of the barriers that inhibit reprogramming and the understanding of which will pave the way to develop safe strategies for efficient reprogramming.
Collapse
|
59
|
Wu Z, Guan KL. Hippo Signaling in Embryogenesis and Development. Trends Biochem Sci 2020; 46:51-63. [PMID: 32928629 DOI: 10.1016/j.tibs.2020.08.008] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/27/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
Hippo pathway components are structurally and functionally conserved and are notable for their role in controlling organ size. More diverse functions of the Hippo pathway have been recognized, including development, tissue homeostasis, wound healing and regeneration, immunity, and tumorigenesis. During embryogenesis, different signaling pathways are repeatedly and cooperatively activated, leading to differential gene expression in specific developmental contexts. In this article, we present an overview on the regulation and function of the Hippo pathway in mammalian early development. We introduce the Hippo pathway components and major upstream signals that act through this pathway to influence embryogenesis. We also discuss the roles of Hippo pathway in tissue specification and organ development during organogenesis.
Collapse
Affiliation(s)
- Zhengming Wu
- 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
|
60
|
Liu J, Gao M, Xu S, Chen Y, Wu K, Liu H, Wang J, Yang X, Wang J, Liu W, Bao X, Chen J. YTHDF2/3 Are Required for Somatic Reprogramming through Different RNA Deadenylation Pathways. Cell Rep 2020; 32:108120. [DOI: 10.1016/j.celrep.2020.108120] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 07/12/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
|
61
|
Bian SB, Yang Y, Liang WQ, Zhang KC, Chen L, Zhang ZT. Leukemia inhibitory factor promotes gastric cancer cell proliferation, migration, and invasion via the LIFR-Hippo-YAP pathway. Ann N Y Acad Sci 2020; 1484:74-89. [PMID: 32827446 DOI: 10.1111/nyas.14466] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/02/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
The long-term outcome of gastric cancer (GC) patients remains unsatisfactory despite some recent improvements. Leukemia inhibitory factor (LIF) is a prognostic biomarker for some solid tumors, however its role in GC remains unknown. In this study, we demonstrated that LIF and LIF receptor (LIFR) are overexpressed in GC tissues and established that a correlation exists between them. LIF and LIFR expression are associated with tumor differentiation, lymphovascular invasion, tumor stage, lymph node metastasis, and pTNM stage, indicating that they may be useful prognostic factors. LIF promoted GC cell proliferation, colony formation, invasion, migration, and tumor growth; it also promoted cell cycle progression and inhibited apoptosis; and knocking out the LIFR gene reversed the effects of LIF. LIF inhibited the activity of the Hippo pathway, resulting in reduced phosphorylation of YAP, increased YAP nuclear translocation, and increased cell proliferation. Finally, silencing YAP mRNA expression suppressed cell proliferation. Overall, the results demonstrate that LIF promotes the malignant biological behavior of GC cells through LIFR-Hippo-YAP signaling. LIF may therefore be a useful biomarker for GC.
Collapse
Affiliation(s)
- Shi-Bo Bian
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China.,Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yun Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wen-Quan Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ke-Cheng Zhang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lin Chen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhong-Tao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
62
|
Ai LQY, Zhu JY, Chen X, Li X, Luo LL, Hu QM, Lin S, Ye J. Endothelial Yes-Associated Protein 1 Promotes Astrocyte Proliferation and Maturation via Cytoplasmic Leukemia Inhibitory Factor Secretion in Oxygen-Induced Retinopathy. Invest Ophthalmol Vis Sci 2020; 61:1. [PMID: 32271890 PMCID: PMC7401846 DOI: 10.1167/iovs.61.4.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Purpose The role of endothelial Yes-associated protein 1 (YAP) in the pathogenesis of retinal angiogenesis and the astrocyte network in the mouse oxygen-induced retinopathy (OIR) model is unknown. Methods For in vivo studies, OIR was induced in conditional endothelial YAP knockout mice and their wild-type littermates. Retinal vascularization and the astrocyte network were evaluated by whole-mount fluorescence and Western blotting. In vitro experiments were performed in astrocytes cultured with human microvascular endothelial cell-1–conditioned medium to analyze the mechanisms underlying the effect of endothelial YAP on astrocytes. Results Endothelial YAP deletion not only impaired retinal blood vessels, but also caused a sparse and disrupted astrocyte network in response to OIR. Levels of the immature astrocyte marker (platelet-derived growth factor A) in the retina were substantially increased owing to YAP deficiency, suggesting a possible failure in astrocyte maturation, whereas retinal expression of leukemia inhibitory factor (LIF) was decreased. In vitro studies suggested that loss or overexpression of YAP resulted in elevated or decreased LIF secretion by human microvascular endothelial cell-1, respectively. Increased LIF levels in the culture medium promoted astrocyte maturation and proliferation and rescued YAP inhibition-induced astrocyte loss. Finally, activating YAP could protect against the pathology of the astrocyte network and even suppress pathologic retinal vascularization in control OIR mice, but not in endothelial YAP-deficient OIR mice. Conclusions Endothelial YAP regulation of LIF secretion is required for normalized astrocyte network formation in OIR, thereby providing a novel target for protecting the astrocyte network and thus benefiting retinal blood vessels.
Collapse
|
63
|
Yamashita A, Yoshitomi H, Kihara S, Toguchida J, Tsumaki N. Culture substrate-associated YAP inactivation underlies chondrogenic differentiation of human induced pluripotent stem cells. Stem Cells Transl Med 2020; 10:115-127. [PMID: 32822104 PMCID: PMC7780802 DOI: 10.1002/sctm.20-0058] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/23/2020] [Accepted: 07/12/2020] [Indexed: 12/11/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are a promising cell source for the creation of cartilage to treat articular cartilage damage. The molecular mechanisms that translate culture conditions to the chondrogenic differentiation of hiPSCs remain to be analyzed. To analyze the effects of culture substrates, we chondrogenically differentiated hiPSCs on Matrigel or laminin 511‐E8 while holding the composition of the chondrogenic medium constant. Cartilage was formed from hiPSCs on Matrigel, but not on laminin 511‐E8. On Matrigel, the hiPSCs were round and yes‐associated protein (YAP) was inactive. In contrast, on laminin 511‐E8, the hiPSCs were flat and YAP was active. Treating the laminin 511‐E8 hiPSCs in a bioreactor caused cell aggregates, in which the cells were round and YAP was inactive. Subsequent culture of the aggregates in chondrogenic medium resulted in cartilage formation. Transient knockdown of YAP in hiPSCs around the start of chondrogenic differentiation successfully formed cartilage on laminin 511‐E8, suggesting that the activation of YAP is responsible for the failure of cartilage formation from hiPSCs on laminin 511‐E8. Consistently, the addition of YAP inhibitors to laminin 511‐E8 hiPSCs caused partial cartilage formation. This study contributes to identifying the molecules that mediate the effects of culture substrates on the chondrogenic differentiation of hiPSCs as well as to developing clinically applicable chondrogenic differentiation methods.
Collapse
Affiliation(s)
- Akihiro Yamashita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hiroyuki Yoshitomi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University (CiRA), Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University (CiRA), Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Noriyuki Tsumaki
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
64
|
Sammarco A, Gomiero C, Sacchetto R, Beffagna G, Michieletto S, Orvieto E, Cavicchioli L, Gelain ME, Ferro S, Patruno M, Zappulli V. Wnt/β-Catenin and Hippo Pathway Deregulation in Mammary Tumors of Humans, Dogs, and Cats. Vet Pathol 2020; 57:774-790. [PMID: 32807036 DOI: 10.1177/0300985820948823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mammary cancer is a common neoplasm in women, dogs, and cats that still represents a therapeutic challenge. Wnt/β-catenin and Hippo pathways are involved in tumor progression, cell differentiation, and metastasis. The aim of this study was to evaluate mRNA and protein expression of molecules involved in these pathways in human (HBC), canine (CMT), and feline mammary tumors (FMT). Real-time quantitative polymerase chain reaction (qPCR) for β-catenin, CCND1, YAP, TAZ, CTGF, and ANKRD1, western blotting for YAP, TAZ, and β-catenin, and immunohistochemistry for estrogen receptor (ER), progesterone receptor (PR), ERBB2, β-catenin, and YAP/TAZ were performed on mammary tumor tissues. The protein expression of active β-catenin was higher in tumors than in healthy tissues in all 3 species. The mRNA expression of the downstream gene CCND1 was increased in HBC ER+ and CMTs compared to healthy tissues. Membranous and cytoplasmic protein expression of β-catenin were strongly negatively correlated in all 3 species. Tumors showed an increased protein expression of YAP/TAZ when compared to healthy tissues. Notably, YAP/TAZ expression was higher in triple negative breast cancers when compared to HBC ER+ and in FMTs when compared to CMTs. The mRNA expression of β-catenin, YAP, TAZ, CTGF, and ANKRD1 was not different between tumors and healthy mammary gland in the 3 species. This study demonstrates deregulation of Wnt/β-catenin and Hippo pathways in mammary tumors, which was more evident at the protein rather than the mRNA level. Wnt/β-catenin and Hippo pathways seem to be involved in mammary carcinogenesis and therefore represent interesting therapeutic targets that should be further investigated.
Collapse
Affiliation(s)
- Alessandro Sammarco
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| | - Chiara Gomiero
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy.,Department of Biomedical Sciences, 9308University of Padua, Italy.,Neuroscience Institute - Italian National Research Council (CNR), Padua, Italy
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| | - Giorgia Beffagna
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy.,Department of Cardio-Thoraco-Vascular Sciences and Public Health, 9308University of Padua, Italy
| | | | - Enrico Orvieto
- Department of Pathology, Azienda Ospedaliera di Padova, Padua, Italy.,Department of Pathology, 18674Santa Maria della Misericordia Hospital, Rovigo, Italy
| | - Laura Cavicchioli
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| | - Maria Elena Gelain
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| | - Silvia Ferro
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, 9308University of Padua, Italy
| |
Collapse
|
65
|
Heng BC, Zhang X, Aubel D, Bai Y, Li X, Wei Y, Fussenegger M, Deng X. Role of YAP/TAZ in Cell Lineage Fate Determination and Related Signaling Pathways. Front Cell Dev Biol 2020; 8:735. [PMID: 32850847 PMCID: PMC7406690 DOI: 10.3389/fcell.2020.00735] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
The penultimate effectors of the Hippo signaling pathways YAP and TAZ, are transcriptional co-activator proteins that play key roles in many diverse biological processes, ranging from cell proliferation, tumorigenesis, mechanosensing and cell lineage fate determination, to wound healing and regeneration. In this review, we discuss the regulatory mechanisms by which YAP/TAZ control stem/progenitor cell differentiation into the various major lineages that are of interest to tissue engineering and regenerative medicine applications. Of particular interest is the key role of YAP/TAZ in maintaining the delicate balance between quiescence, self-renewal, proliferation and differentiation of endogenous adult stem cells within various tissues/organs during early development, normal homeostasis and regeneration/healing. Finally, we will consider how increasing knowledge of YAP/TAZ signaling might influence the trajectory of future progress in regenerative medicine.
Collapse
Affiliation(s)
- Boon C. Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Faculty of Science and Technology, Sunway University, Subang Jaya, Malaysia
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
| | - Dominique Aubel
- IUTA Department Genie Biologique, Universite Claude Bernard Lyon 1, Villeurbanne, France
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH-Zürich, Basel, Switzerland
| | - Xuliang Deng
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
66
|
Sun X, Ren Z, Cun Y, Zhao C, Huang X, Zhou J, Hu R, Su X, Ji L, Li P, Mak K, Gao F, Yang Y, Xu H, Ding J, Cao N, Li S, Zhang W, Lan P, Sun H, Wang J, Yuan P. Hippo-YAP signaling controls lineage differentiation of mouse embryonic stem cells through modulating the formation of super-enhancers. Nucleic Acids Res 2020; 48:7182-7196. [PMID: 32510157 PMCID: PMC7367178 DOI: 10.1093/nar/gkaa482] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Hippo-YAP signaling pathway functions in early lineage differentiation of pluripotent stem cells, but the detailed mechanisms remain elusive. We found that knockout (KO) of Mst1 and Mst2, two key components of the Hippo signaling in mouse embryonic stem cells (ESCs), resulted in a disruption of differentiation into mesendoderm lineage. To further uncover the underlying regulatory mechanisms, we performed a series of ChIP-seq experiments with antibodies against YAP, ESC master transcription factors and some characterized histone modification markers as well as RNA-seq assays using wild type and Mst KO samples at ES and day 4 embryoid body stage respectively. We demonstrate that YAP is preferentially co-localized with super-enhancer (SE) markers such as Nanog, Sox2, Oct4 and H3K27ac in ESCs. The hyper-activation of nuclear YAP in Mst KO ESCs facilitates the binding of Nanog, Sox2 and Oct4 as well as H3K27ac modification at the loci where YAP binds. Moreover, Mst depletion results in novel SE formation and enhanced liquid-liquid phase-separated Med1 condensates on lineage associated genes, leading to the upregulation of these genes and the distortion of ESC differentiation. Our study reveals a novel mechanism on how Hippo-YAP signaling pathway dictates ESC lineage differentiation.
Collapse
Affiliation(s)
- Xiang Sun
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Zhijun Ren
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Yixian Cun
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Cai Zhao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Xianglin Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Jiajian Zhou
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Rong Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Xiaoxi Su
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
- China Hong Kong Children's Hospital, Hong Kong SAR
| | - Lu Ji
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - King Lun Kingston Mak
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
| | - Feng Gao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Yi Yang
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - He Xu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Junjun Ding
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- Department of Histology and embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Nan Cao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Shuo Li
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Wensheng Zhang
- Cam-Su Genomic Resource Center, Soochow University, Suzhou 215123, China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Jinkai Wang
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ping Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
67
|
Azad T, Rezaei R, Surendran A, Singaravelu R, Boulton S, Dave J, Bell JC, Ilkow CS. Hippo Signaling Pathway as a Central Mediator of Receptors Tyrosine Kinases (RTKs) in Tumorigenesis. Cancers (Basel) 2020; 12:cancers12082042. [PMID: 32722184 PMCID: PMC7463967 DOI: 10.3390/cancers12082042] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
The Hippo pathway plays a critical role in tissue and organ growth under normal physiological conditions, and its dysregulation in malignant growth has made it an attractive target for therapeutic intervention in the fight against cancer. To date, its complex signaling mechanisms have made it difficult to identify strong therapeutic candidates. Hippo signaling is largely carried out by two main activated signaling pathways involving receptor tyrosine kinases (RTKs)—the RTK/RAS/PI3K and the RTK-RAS-MAPK pathways. However, several RTKs have also been shown to regulate this pathway to engage downstream Hippo effectors and ultimately influence cell proliferation. In this text, we attempt to review the diverse RTK signaling pathways that influence Hippo signaling in the context of oncogenesis.
Collapse
Affiliation(s)
- Taha Azad
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Reza Rezaei
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Abera Surendran
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ragunath Singaravelu
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Stephen Boulton
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jaahnavi Dave
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - John C. Bell
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Carolina S. Ilkow
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.A.); (R.R.); (A.S.); (R.S.); (S.B.); (J.D.); (J.C.B.)
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Correspondence: ; Tel.: +1-613-737-8899 (ext. 75208)
| |
Collapse
|
68
|
Wang J, Zheng Z, Huang B, Wu H, Zhang X, Chen Y, Liu J, Shan Z, Fan S, Chen J, Zhao F. Osteal Tissue Macrophages Are Involved in Endplate Osteosclerosis through the OSM-STAT3/YAP1 Signaling Axis in Modic Changes. THE JOURNAL OF IMMUNOLOGY 2020; 205:968-980. [PMID: 32690652 DOI: 10.4049/jimmunol.1901001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 06/22/2020] [Indexed: 11/19/2022]
Abstract
Modic changes (MCs) are radiographic manifestations of lumbar degenerative diseases. Various types of MCs are often associated with endplate osteosclerosis. Osteal tissue macrophages (Osteomacs) were reported to be crucial for bone homeostasis and bone repair, but whether osteomacs participate in the endplate osteosclerosis in MCs remained unclear. In this study, we tried to explore the critical role of osteomacs in regulating osteogenesis in MCs. We collected MCs from patient samples and developed a Propionibacterium acnes-induced rat MCs model, using microcomputed tomography and immunohistochemistry to detect the endplate bone mass and distribution of osteomacs. In patients' MCs, osteomacs increased in endplate subchondral bone, especially in Modic type II. Endplate in Modic type III presented a stable osteosclerosis. In rat MCs model, osteomacs increased in the bone hyperplasia area but not in the inflammation area of the endplate region, whereas the distribution of osteomacs was consistent with the area of osteosclerosis. To further explore the functions of osteomacs in vitro, we isolated osteomacs using MACS technology and found osteomacs secreted oncostatin M (OSM) and strongly promoted osteoblast differentiation rather than osteoclast through the mechanism of OSM-mediated tyrosine phosphorylation and interaction of STAT3 and Yes-associated protein 1 (YAP1). STAT3 phosphorylation inhibition or YAP1 knockdown attenuated OSM-mediated osteoblast differentiation. Finally, we confirmed that blockade of OSM in vivo using anti-OSM-neutralizing Ab prevented endplate osteosclerosis in rat MCs model. Taken together, these findings confirmed that endplate osteosclerosis in MCs was accompanied by an increased number of osteomacs, which regulated osteogenesis via the OSM-STAT3/YAP1 signaling axis.
Collapse
Affiliation(s)
- Jiasheng Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Zeyu Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Bao Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Hao Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Xuyang Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Yilei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Zhi Shan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; and Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| |
Collapse
|
69
|
Sorrentino G, Rezakhani S, Yildiz E, Nuciforo S, Heim MH, Lutolf MP, Schoonjans K. Mechano-modulatory synthetic niches for liver organoid derivation. Nat Commun 2020; 11:3416. [PMID: 32651372 PMCID: PMC7351772 DOI: 10.1038/s41467-020-17161-0] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 06/11/2020] [Indexed: 12/27/2022] Open
Abstract
The recent demonstration that primary cells from the liver can be expanded in vitro as organoids holds enormous promise for regenerative medicine and disease modelling. The use of three-dimensional (3D) cultures based on ill-defined and potentially immunogenic matrices, however, hampers the translation of liver organoid technology into real-life applications. We here use chemically defined hydrogels for the efficient derivation of both mouse and human hepatic organoids. Organoid growth is found to be highly stiffness-sensitive, a mechanism independent of acto-myosin contractility and requiring instead activation of the Src family of kinases (SFKs) and yes-associated protein 1 (YAP). Aberrant matrix stiffness, on the other hand, results in compromised proliferative capacity. Finally, we demonstrate the establishment of biopsy-derived human liver organoids without the use of animal components at any step of the process. Our approach thus opens up exciting perspectives for the establishment of protocols for liver organoid-based regenerative medicine. 3D liver organoids hold great promise for regenerative medicine but the use of ill-defined matrices limits their potential. Here, the authors generate human and mouse liver organoids using a chemically defined matrix, and reveal a link between matrix stiffness and organoid growth that does not require acto-myosin contraction.
Collapse
Affiliation(s)
- Giovanni Sorrentino
- Laboratory of Metabolic Signaling, Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Saba Rezakhani
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Ece Yildiz
- Laboratory of Metabolic Signaling, Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Sandro Nuciforo
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.,Clinic of Gastroenterology and Hepatology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
70
|
Yang T, Heng C, Zhou Y, Hu Y, Chen S, Wang H, Yang H, Jiang Z, Qian S, Wang Y, Wang J, Zhu X, Du L, Yin X, Lu Q. Targeting mammalian serine/threonine-protein kinase 4 through Yes-associated protein/TEA domain transcription factor-mediated epithelial-mesenchymal transition ameliorates diabetic nephropathy orchestrated renal fibrosis. Metabolism 2020; 108:154258. [PMID: 32376130 DOI: 10.1016/j.metabol.2020.154258] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/13/2020] [Accepted: 04/29/2020] [Indexed: 12/19/2022]
Abstract
RATIONALE Tubulointerstitial fibrosis, which is closely related to functional injury of the kidney, can be observed in advanced stages of diabetic nephropathy (DN). Mammalian serine/threonine-protein kinase 4 (MST1), a core component of the Hippo pathway that is involved in cellular proliferation and differentiation, plays a crucial role in the pathogenesis of multiple metabolic diseases, kidney diseases and cancer. METHODS In type 1 and type 2 diabetic animals, as well as in human proximal tubular epithelial cells (HK-2), activation of MST1 was analyzed by immunohistochemistry and western blotting. In db/db mice, MST1 protein was knocked down or overexpressed by shRNA, and renal function, fibrosis, and downstream signaling were then investigated. RNA silencing and overexpression were performed by using an MST1 or YAP knockdown/expression lentivirus to investigate the regulation of MST1-mediated YAP/TEAD signaling pathways in the fibrosis process in HK-2 cells. Luciferase and coimmunoprecipitation (co-IP) assays were used to identify whether YAP directly regulated TEAD activation by forming a YAP-TEAD heterodimer, which ultimately leads to tubulointerstitial fibrosis. RESULTS MST1 activation was significantly decreased in type 1 and type 2 diabetic nephropathy. Notably, the downregulation of MST1 activation was also observed in HK-2 cells in a glucose- and time-dependent manner. In vivo, downregulation of MST1 was sufficient to promote renal dysfunction and fibrosis in db/m mice, whereas overexpression of MST1 ameliorated diabetic nephropathy-induced renal fibrosis. Further mechanistic study demonstrated that activated YAP induced by MST1 inhibition directly upregulated TEAD activation by binding to TEAD and forming a YAP-TEAD heterodimer, resulting in the promotion of epithelial-mesenchymal transition (EMT) and fibrosis in renal tubular epithelial. CONCLUSIONS MST1 activation represents a potential therapeutic strategy to treat or prevent the progression of diabetic nephropathy-induced renal fibrosis.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Cai Heng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yi Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yinlu Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Shangxiu Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiyan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Hao Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Sitong Qian
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yinan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Jianyun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xia Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
71
|
Werneburg N, Gores GJ, Smoot RL. The Hippo Pathway and YAP Signaling: Emerging Concepts in Regulation, Signaling, and Experimental Targeting Strategies With Implications for Hepatobiliary Malignancies. Gene Expr 2020; 20:67-74. [PMID: 31253203 PMCID: PMC7284105 DOI: 10.3727/105221619x15617324583639] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Hippo pathway and its effector protein YAP (a transcriptional coactivator) have been identified as important in the biology of both hepatocellular carcinoma and cholangiocarcinoma. First identified as a tumor suppressor pathway in Drosophila, the understanding of the mammalian YAP signaling and its regulation continues to expand. In its "on" function, the canonical regulatory Hippo pathway, a well-described serine/threonine kinase module, regulates YAP function by restricting its subcellular localization to the cytoplasm. In contrast, when the Hippo pathway is "off," YAP translocates to the nucleus and drives cotranscriptional activity. Given the role of Hippo/YAP signaling in hepatic malignancies, investigators have sought to target these molecules; however, standard approaches have not been successful based on the pathways' negative regulatory role. More recently, additional regulatory mechanisms, such as tyrosine phosphorylation, of YAP have been described. These represent positive regulatory events that may be targetable. Additionally, several groups have identified potentiating feed-forward signaling for YAP in multiple contexts, suggesting other experimental therapeutic approaches to interrupt these signaling loops. Herein we explore the current data supporting alternative YAP regulatory pathways, review the described feed-forward signaling cascades that are YAP dependent, and explore targeting strategies that have been employed in preclinical models of hepatic malignancies.
Collapse
Affiliation(s)
- Nathan Werneburg
- *Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Gregory J. Gores
- *Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Rory L. Smoot
- †Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
72
|
Cao X, Wang C, Liu J, Zhao B. Regulation and functions of the Hippo pathway in stemness and differentiation. Acta Biochim Biophys Sin (Shanghai) 2020; 52:736-748. [DOI: 10.1093/abbs/gmaa048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract
The Hippo pathway plays important roles in organ development, tissue regeneration, and human diseases, such as cancer. In the canonical Hippo pathway, the MST1/2-LATS1/2 kinase cascade phosphorylates and inhibits transcription coactivators Yes-associated protein and transcription coactivator with PDZ-binding motif and thus regulates transcription of genes important for cell proliferation and apoptosis. However, recent studies have depicted a much more complicate picture of the Hippo pathway with many new components and regulatory stimuli involving both chemical and mechanical signals. Furthermore, accumulating evidence indicates that the Hippo pathway also plays important roles in the determination of cell fates, such as self-renewal and differentiation. Here, we review regulations of the Hippo pathway and its functions in stemness and differentiation emphasizing recent discoveries.
Collapse
Affiliation(s)
- Xiaolei Cao
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Chenliang Wang
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Jiyang Liu
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Bin Zhao
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
73
|
Chen X, Li Y, Luo J, Hou N. Molecular Mechanism of Hippo-YAP1/TAZ Pathway in Heart Development, Disease, and Regeneration. Front Physiol 2020; 11:389. [PMID: 32390875 PMCID: PMC7191303 DOI: 10.3389/fphys.2020.00389] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/01/2020] [Indexed: 01/20/2023] Open
Abstract
The Hippo-YAP1/TAZ pathway is a highly conserved central mechanism that controls organ size through the regulation of cell proliferation and other physical attributes of cells. The transcriptional factors Yes-associated protein 1 (YAP1) and PDZ-binding motif (TAZ) act as downstream effectors of the Hippo pathway, and their subcellular location and transcriptional activities are affected by multiple post-translational modifications (PTMs). Studies have conclusively demonstrated a pivotal role of the Hippo-YAP1/TAZ pathway in cardiac development, disease, and regeneration. Targeted therapeutics for the YAP1/TAZ could be an effective treatment option for cardiac regeneration and disease. This review article provides an overview of the Hippo-YAP1/TAZ pathway and the increasing impact of PTMs in fine-tuning YAP1/TAZ activation; in addition, we discuss the potential contributions of the Hippo-YAP1/TAZ pathway in cardiac development, disease, and regeneration.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yilang Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiandong Luo
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ning Hou
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, and The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
74
|
YAP Non-cell-autonomously Promotes Pluripotency Induction in Mouse Cells. Stem Cell Reports 2020; 14:730-743. [PMID: 32243844 PMCID: PMC7160372 DOI: 10.1016/j.stemcr.2020.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 01/11/2023] Open
Abstract
Yes-associated protein (YAP) is known to promote the stemness of multiple stem cell types, including pluripotent stem cells, while also antagonizing pluripotency during early embryogenesis. How YAP accomplishes these distinct functions remains unclear. Here, we report that, depending on the specific cells in which it is expressed, YAP could exhibit opposing effects on pluripotency induction from mouse somatic cells. Specifically, YAP inhibits pluripotency induction cell-autonomously but promotes it non-cell-autonomously. For its non-cell-autonomous role, YAP alters the expression of many secreted and matricellular proteins, including CYR61. YAP's non-cell-autonomous promoting effect could be recapitulated by recombinant CYR61 and abrogated by CYR61 depletion. Thus, we define a YAP-driven effect on enhancing pluripotency induction largely mediated by CYR61. Our work highlights the importance of considering the distinct contributions from heterologous cell types in deciphering cell fate control mechanisms and calls for careful re-examination of the co-existing bystander cells in complex cultures and tissues. YAP inhibits pluripotency induction when expressed cell-autonomously YAP promotes pluripotency induction when expressed non-cell-autonomously YAP expression alters the expression of genes that encode extracellular components CYR61 is secreted by YAP-expressing cells to promote nearby reprogramming
Collapse
|
75
|
Fathi Maroufi N, Hasegawa K, Vahedian V, Nazari Soltan Ahmad S, Zarebkohan A, Miresmaeili Mazrakhondi SA, Hosseini V, Rahbarghazi R. A glimpse into molecular mechanisms of embryonic stem cells pluripotency: Current status and future perspective. J Cell Physiol 2020; 235:6377-6392. [DOI: 10.1002/jcp.29616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Nazila Fathi Maroufi
- Stem Cell and Regenerative Medicine InstituteTabriz University of Medical Sciences Tabriz Iran
- Student Research CommitteeTabriz University of Medical Sciences Tabriz Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of MedicineTabriz University of Medical Sciences Tabriz Iran
| | - Kouichi Hasegawa
- Institute for Integrated Cell‐Material Sciences, Institute for Advanced StudyKyoto University Kyoto Japan
| | - Vahid Vahedian
- Department of Medical Laboratory Sciences, Faculty of MedicineIslamic Azad University Sari Iran
- Clinical Laboratory Medicine DepartmentRofeydeh Hospital University of Social Welfare and Rehabilitation Science Tehran Iran
| | - Saeed Nazari Soltan Ahmad
- Department of Biochemistry and Clinical Laboratories, Faculty of MedicineTabriz University of Medical Sciences Tabriz Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical Sciences Tabriz Iran
| | | | - Vahid Hosseini
- Department of Biochemistry and Clinical Laboratories, Faculty of MedicineTabriz University of Medical Sciences Tabriz Iran
- Tuberculosis and Lung Disease Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Reza Rahbarghazi
- Tuberculosis and Lung Disease Research CenterTabriz University of Medical Sciences Tabriz Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
76
|
Ai LQY, Yuan RD, Chen X, Liu YJ, Liu WY, Zhu JY, Zhang Z, Yan J, Chen CL, Lin S, Ye J. Retinal blood vessel-origin yes-associated protein (YAP) governs astrocytic maturation via leukaemia inhibitory factor (LIF). Cell Prolif 2020; 53:e12757. [PMID: 31916327 PMCID: PMC7046482 DOI: 10.1111/cpr.12757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/26/2019] [Accepted: 12/16/2019] [Indexed: 12/31/2022] Open
Abstract
Objectives To testify that endothelial cells (ECs) induce astrocyte maturation by leukaemia inhibitory factor (LIF) secretion. Materials and Methods In vivo experiments, mice bearing floxed alleles of YAP were crossed with mice expressing a Cre recombinase driven by the endothelial Tek promoter (Tek‐Cre) to finally obtain the following three genotypes: YAPf/f, Tek‐Cre; YAPf/w, Tek‐Cre; and YAPf/f. Retinal vascularization and astrocyte network were evaluated by whole‐mount fluorescence and Western blotting. In vitro, experiments were performed in an astrocyte and human microvascular endothelial cell (HMEC‐1) coculture model to analyse the mechanisms underlying the effect of endothelial YAP on astrocytes. Results In vivo, YAPf/f;Tek‐Cre mice showed delayed angiogenesis, sparse vessels and decreased glial fibrillary acidic protein (GFAP)+ astrocytes but aberrant growth of endothelial networks and immature astrocytes (platelet‐derived growth factor A, PDGFRA+ astrocytes) overgrowth. In vitro, Yap deletion attenuated the LIF release that delayed the maturation of retinal astrocyte which was consistent with the results of HMEC‐1—astrocyte coculture. The effect of YAP overexpression on LIF‐LIFR axis in HMEC‐1 interferes the GFAP expression of astrocyte. In contrast, LIF protein rescues the astrocytic GFAP expression when EC YAP was inhibited by siRNAs. Conclusions We show that EC yes‐associated protein (YAP) is not only a critical coactivator of Hippo signalling in retinal vessel development but also plays an essential role in retinal astrocyte maturation by regulating LIF production.
Collapse
Affiliation(s)
- Li-Qian-Yu Ai
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Rong-Di Yuan
- Department of Ophthalmology, XinQiao Hospital, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yun-Jia Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Wen-Yi Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jing-Yi Zhu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Zhou Zhang
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jun Yan
- Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chun-Lin Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
77
|
Bae JS, Kim SM, Jeon Y, Sim J, Jang JY, Son J, Hong W, Park MK, Lee H. Loss of Mob1a/b impairs the differentiation of mouse embryonic stem cells into the three germ layer lineages. Exp Mol Med 2019; 51:1-12. [PMID: 31723125 PMCID: PMC6853965 DOI: 10.1038/s12276-019-0342-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 06/24/2019] [Accepted: 09/19/2019] [Indexed: 01/05/2023] Open
Abstract
The Hippo pathway plays a crucial role in cell proliferation and apoptosis and can regulate stem cell maintenance and embryonic development. MOB kinase activators 1A and 1B (Mob1a/b) are key components of the Hippo pathway, whose homozygous deletion in mice causes early embryonic lethality at the preimplantation stage. To investigate the role of Mob1a/b in stem cell maintenance and differentiation, an embryonic stem cell (ESC) clone in which Mob1a/b could be conditionally depleted was generated and characterized. Although Mob1a/b depletion did not affect the stemness or proliferation of mouse ESCs, this depletion caused defects in differentiation into the three germ layers. Yap knockdown rescued the in vitro and in vivo defects in differentiation caused by Mob1a/b depletion, suggesting that differentiation defects caused by Mob1a/b depletion were Yap-dependent. In teratoma experiments, Yap knockdown in Mob1a/b-depleted ESCs partially restored defects in differentiation, indicating that hyperactivation of Taz, another effector of the Hippo pathway, inhibited differentiation into the three germ layers. Taken together, these results suggest that Mob1a/b or Hippo signaling plays a critical role in the differentiation of mouse ESCs into the three germ layers, which is dependent on Yap. These close relationship of the Hippo pathway with the differentiation of stem cells supports its potential as a therapeutic target in regenerative medicine.
Collapse
Affiliation(s)
- June Sung Bae
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Sun Mi Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Yoon Jeon
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Juyeon Sim
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Ji Yun Jang
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Jaehyung Son
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Woosol Hong
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Mi Kyung Park
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Ho Lee
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea. .,Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea.
| |
Collapse
|
78
|
Kim DH, Choi HI, Park JS, Kim CS, Bae EH, Ma SK, Kim SW. Src-mediated crosstalk between FXR and YAP protects against renal fibrosis. FASEB J 2019; 33:11109-11122. [PMID: 31298930 DOI: 10.1096/fj.201900325r] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Renal fibrosis is the common pathway of chronic kidney disease progression. The nuclear receptor farnesoid X receptor [FXR, NR1H4 (nuclear receptor subfamily 1 group member 4)], a multifunctional transcription factor, plays a pivotal role in protecting against fibrosis. However, the mechanisms underlying these antifibrotic actions of FXR in kidney disease are largely unknown. Here, we show that agonist GW4064-mediated FXR activation inhibits the activity of the nonreceptor tyrosine kinase Src (proto-oncogene tyrosine-protein kinase), which is critical for regulation of yes-associated protein (YAP) phosphorylation and nuclear localization in renal fibrosis. Activation of FXR suppressed renal fibrosis and Tyr416-Src phosphorylation in TGF-β-treated human renal proximal tubule epithelial (HK2) cells. Moreover, GW4064 treatment in HK2 cells increased Ser127 phosphorylation, cytosolic accumulation of YAP, and interaction of the hippo core kinases (Ste20-like kinase 1, large tumor suppressor kinase 1, and salvador homolog 1). Inhibition of Src using PP2 (Src kinase inhibitor) prevented renal fibrosis and increased Ser127 phosphorylation and cytosolic accumulation of YAP. The expression of fibrosis markers, inflammatory genes, and YAP target genes was increased in the kidneys of FXR knockout mice compared with those of wild-type mice. In addition, GW4064 or WAY-362450 (turofexorate isopropyl) treatment protected against unilateral ureteral obstruction-induced renal fibrosis. Collectively, our data support the novel conclusion that Src-mediated crosstalk between FXR and YAP protects against renal fibrosis, making this pathway a possible therapeutic target for chronic kidney disease.-Kim, D.-H., Choi, H.-I., Park, J. S., Kim, C. S., Bae, E. H., Ma, S. K., Kim, S. W. Src-mediated crosstalk between FXR and YAP protects against renal fibrosis.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Hoon-In Choi
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Jung Sun Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
79
|
Han Z, Yu Y, Cai B, Xu Z, Bao Z, Zhang Y, Bamba D, Ma W, Gao X, Yuan Y, Zhang L, Yu M, Liu S, Yan G, Jin M, Huang Q, Wang X, Hua B, Yang F, Pan Z, Liang H, Liu Y. YAP/TEAD3 signal mediates cardiac lineage commitment of human-induced pluripotent stem cells. J Cell Physiol 2019; 235:2753-2760. [PMID: 31541452 DOI: 10.1002/jcp.29179] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes differentiated from human-induced pluripotent stem cells (hiPSCs) hold great potential for therapy of heart diseases. However, the underlying mechanisms of its cardiac differentiation have not been fully elucidated. Hippo-YAP signal pathway plays important roles in cell differentiation, tissue homeostasis, and organ size. Here, we identify the role of Hippo-YAP signal pathway in determining cardiac differentiation fate of hiPSCs. We found that cardiac differentiation of hiPSCs were significantly inhibited after treatment with verteporfin (a selective and potent YAP inhibitor). During hiPSCs differentiation from mesoderm cells (MESs) into cardiomyocytes, verteporfin treatment caused the cells retained in the earlier cardiovascular progenitor cells (CVPCs) stage. Interestingly, during hiPSCs differentiation from CVPC into cardiomyocytes, verteporfin treatment induced cells dedifferentiation into the earlier CVPC stage. Mechanistically, we found that YAP interacted with transcriptional enhanced associate domain transcription factor 3 (TEAD3) to regulate cardiac differentiation of hiPSCs during the CVPC stage. Consistently, RNAi-based silencing of TEAD3 mimicked the phenotype as the cells treated with verteporfin. Collectively, our study suggests that YAP-TEAD3 signaling is important for cardiomyocyte differentiation of hiPSCs. Our findings provide new insight into the function of Hippo-YAP signal in cardiovascular lineage commitment.
Collapse
Affiliation(s)
- Zhenbo Han
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Ying Yu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Benzhi Cai
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Zihang Xu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Zhengyi Bao
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Ying Zhang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Djibril Bamba
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Wenya Ma
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Xinlu Gao
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Ye Yuan
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Lai Zhang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Meixi Yu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Shenzhen Liu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Gege Yan
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Mengyu Jin
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Qi Huang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Xiuxiu Wang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Bingjie Hua
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Fan Yang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Zhenwei Pan
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Haihai Liang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Yu Liu
- Department of Clinical Laboratory at the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
80
|
A gain-of-functional screen identifies the Hippo pathway as a central mediator of receptor tyrosine kinases during tumorigenesis. Oncogene 2019; 39:334-355. [PMID: 31477837 DOI: 10.1038/s41388-019-0988-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 12/16/2022]
Abstract
The Hippo pathway has emerged as a key signaling pathway that regulates various biological functions. Dysregulation of the Hippo pathway has been implicated in a broad range of human cancer types. While a number of stimuli affecting the Hippo pathway have been reported, its upstream kinase and extracellular regulators remain largely unknown. Here we performed the first comprehensive gain-of-functional screen for receptor tyrosine kinases (RTKs) regulating the Hippo pathway using an RTK overexpression library and a Hippo signaling activity biosensor. Surprisingly, we found that the majority of RTKs could regulate the Hippo signaling activity. We further characterized several of these novel relationships [TAM family members (TYRO3, AXL, METRK), RET, and FGFR family members (FGFR1 and FGFR2)] and found that the Hippo effectors YAP/TAZ are central mediators of the tumorigenic phenotypes (e.g., increased cell proliferation, transformation, increased cell motility, and angiogenesis) induced by these RTKs and their extracellular ligands (Gas6, GDNF, and FGF) through either PI3K or MAPK signaling pathway. Significantly, we identify FGFR, RET, and MERTK as the first RTKs that can directly interact with and phosphorylate YAP/TAZ at multiple tyrosine residues independent of upstream Hippo signaling, thereby activating their functions in tumorigenesis. In conclusion, we have identified several novel kinases and extracellular stimuli regulating the Hippo pathway. Our findings also highlight the pivotal role of the Hippo pathway in mediating Gas6/GDNF/FGF-TAM/RET/FGFR-MAPK/PI3K signaling during tumorigenesis and provide a compelling rationale for targeting YAP/TAZ in RTK-driven cancers.
Collapse
|
81
|
Hamanaka N, Nakanishi Y, Mizuno T, Horiguchi-Takei K, Akiyama N, Tanimura H, Hasegawa M, Satoh Y, Tachibana Y, Fujii T, Sakata K, Ogasawara K, Ebiike H, Koyano H, Sato H, Ishii N, Mio T. YES1 Is a Targetable Oncogene in Cancers Harboring YES1 Gene Amplification. Cancer Res 2019; 79:5734-5745. [PMID: 31391186 DOI: 10.1158/0008-5472.can-18-3376] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/18/2019] [Accepted: 07/23/2019] [Indexed: 11/16/2022]
Abstract
Targeting genetic alterations of oncogenes by molecular-targeted agents (MTA) is an effective approach for treating cancer. However, there are still no clinical MTA options for many cancers, including esophageal cancer. We used a short hairpin RNA library to screen for a new oncogene in the esophageal cancer cell line KYSE70 and identified YES proto-oncogene 1 (YES1) as having a significant impact on tumor growth. An analysis of clinical samples showed that YES1 gene amplification existed not only in esophageal cancer but also in lung, head and neck, bladder, and other cancers, indicating that YES1 would be an attractive target for a cancer drug. Because there is no effective YES1 inhibitor so far, we generated a YES1 kinase inhibitor, CH6953755. YES1 kinase inhibition by CH6953755 led to antitumor activity against YES1-amplified cancers in vitro and in vivo. Yes-associated protein 1 (YAP1) played a role downstream of YES1 and contributed to the growth of YES1-amplified cancers. YES1 regulated YAP1 transcription activity by controlling its nuclear translocation and serine phosphorylation. These findings indicate that the regulation of YAP1 by YES1 plays an important role in YES1-amplified cancers and that CH6953755 has therapeutic potential in such cancers. SIGNIFICANCE: These findings identify the SRC family kinase YES1 as a targetable oncogene in esophageal cancer and describe a new inhibitor for YES1 that has potential for clinical utility.See related commentary by Rai, p. 5702.
Collapse
Affiliation(s)
- Natsuki Hamanaka
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Yoshito Nakanishi
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan.
| | - Takakazu Mizuno
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | | | - Nukinori Akiyama
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Hiromi Tanimura
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Masami Hasegawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Yasuko Satoh
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Yukako Tachibana
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Toshihiko Fujii
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Kiyoaki Sakata
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Kiyomoto Ogasawara
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Hirosato Ebiike
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Hiroshi Koyano
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Haruhiko Sato
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Nobuya Ishii
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| | - Toshiyuki Mio
- Research Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Kanagawa, Japan
| |
Collapse
|
82
|
Blockade of leukemia inhibitory factor as a therapeutic approach to KRAS driven pancreatic cancer. Nat Commun 2019; 10:3055. [PMID: 31296870 PMCID: PMC6624260 DOI: 10.1038/s41467-019-11044-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 06/17/2019] [Indexed: 11/09/2022] Open
Abstract
KRAS mutations are present in over 90% of pancreatic ductal adenocarcinomas (PDAC), and drive their poor outcomes and failure to respond to targeted therapies. Here we show that Leukemia Inhibitory Factor (LIF) expression is induced specifically by oncogenic KRAS in PDAC and that LIF depletion by genetic means or by neutralizing antibodies prevents engraftment in pancreatic xenograft models. Moreover, LIF-neutralizing antibodies synergize with gemcitabine to eradicate established pancreatic tumors in a syngeneic, KrasG12D-driven, PDAC mouse model. The related cytokine IL-6 cannot substitute for LIF, suggesting that LIF mediates KRAS-driven malignancies through a non-STAT-signaling pathway. Unlike IL-6, LIF inhibits the activity of the Hippo-signaling pathway in PDACs. Depletion of YAP inhibits the function of LIF in human PDAC cells. Our data suggest a crucial role of LIF in KRAS-driven pancreatic cancer and that blockade of LIF by neutralizing antibodies represents an attractive approach to improving therapeutic outcomes. KRAS mutations are frequent in pancreatic ductal adenocarcinoma, leading to bad prognosis and resistance to targeted therapies. Here, the authors show that LIF expression is specifically induced by KRAS and constitutes a potential target to treat these KRAS-mutated cancers.
Collapse
|
83
|
Vázquez-Marín J, Gutiérrez-Triana JA, Almuedo-Castillo M, Buono L, Gómez-Skarmeta JL, Mateo JL, Wittbrodt J, Martínez-Morales JR. yap1b, a divergent Yap/Taz family member, cooperates with yap1 in survival and morphogenesis via common transcriptional targets. Development 2019; 146:dev.173286. [PMID: 31142542 DOI: 10.1242/dev.173286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/17/2019] [Indexed: 11/20/2022]
Abstract
Yap1/Taz are well-known Hippo effectors triggering complex transcriptional programs controlling growth, survival and cancer progression. Here, we describe yap1b, a new Yap1/Taz family member with a unique transcriptional activation domain that cannot be phosphorylated by Src/Yes kinases. We show that yap1b evolved specifically in euteleosts (i.e. including medaka but not zebrafish) by duplication and adaptation of yap1. Using DamID-seq, we generated maps of chromatin occupancy for Yap1, Taz (Wwtr1) and Yap1b in gastrulating zebrafish and medaka embryos. Our comparative analyses uncover the genetic programs controlled by Yap family proteins during early embryogenesis, and show largely overlapping targets for Yap1 and Yap1b. CRISPR/Cas9-induced mutation of yap1b in medaka does not result in an overt phenotype during embryogenesis or adulthood. However, yap1b mutation strongly enhances the embryonic malformations observed in yap1 mutants. Thus yap1 -/-; yap1b -/- double mutants display more severe body flattening, eye misshaping and increased apoptosis than yap1 -/- single mutants, thus revealing overlapping gene functions. Our results indicate that, despite its divergent transactivation domain, Yap1b cooperates with Yap1 to regulate cell survival and tissue morphogenesis during early development.
Collapse
Affiliation(s)
| | - José Arturo Gutiérrez-Triana
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany.,Escuela de Microbiología, Facultad de la Salud, Universidad Industrial de Santander, Bucaramanga, 680002, Colombia
| | | | - Lorena Buono
- Centro Andaluz de Biología del Desarrollo (CSIC/UPO/JA), 41013 Seville, Spain
| | | | - Juan Luis Mateo
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany.,Departamento de Informática, Universidad de Oviedo, Oviedo 33005, Spain
| | - Joachim Wittbrodt
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| | | |
Collapse
|
84
|
Hashimoto M, Sasaki H. Epiblast Formation by TEAD-YAP-Dependent Expression of Pluripotency Factors and Competitive Elimination of Unspecified Cells. Dev Cell 2019; 50:139-154.e5. [PMID: 31204175 DOI: 10.1016/j.devcel.2019.05.024] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 03/29/2019] [Accepted: 05/10/2019] [Indexed: 01/15/2023]
Abstract
The epiblast is a pluripotent cell population first formed in preimplantation embryos, and its quality is important for proper development. Here, we examined the mechanisms of epiblast formation and found that the Hippo pathway transcription factor TEAD and its coactivator YAP regulate expression of pluripotency factors. After specification of the inner cell mass, YAP accumulates in the nuclei and activates TEAD. TEAD activity is required for strong expression of pluripotency factors and is variable in the forming epiblast. Cells showing low TEAD activity are eliminated from the epiblast through cell competition. Pluripotency factor expression and MYC control cell competition downstream of TEAD activity. Cell competition eliminates unspecified cells and is required for proper organization of the epiblast. These results suggest that induction of pluripotency factors by TEAD activity and elimination of unspecified cells via cell competition ensure the production of an epiblast with naive pluripotency.
Collapse
Affiliation(s)
- Masakazu Hashimoto
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
85
|
Murakami M, Kamimura D, Hirano T. Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines. Immunity 2019; 50:812-831. [DOI: 10.1016/j.immuni.2019.03.027] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
|
86
|
Yeung YT, Guerrero-Castilla A, Cano M, Muñoz MF, Ayala A, Argüelles S. Dysregulation of the Hippo pathway signaling in aging and cancer. Pharmacol Res 2019; 143:151-165. [PMID: 30910741 DOI: 10.1016/j.phrs.2019.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/04/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
Human beings are facing emerging degenerative and cancer diseases, in large part, as a consequence of increased life expectancy. In the near future, researchers will have to put even more effort into fighting these new challenges, one of which will be prevention of cancer while continuing to improve the aging process through this increased life expectancy. In the last few decades, relevance of the Hippo pathway on cancer has become an important study since it is a major regulator of organ size control and proliferation. However, its deregulation can induce tumors throughout the body by regulating cell proliferation, disrupting cell polarity, releasing YAP and TAZ from the Scribble complexes and facilitating survival gene expression via activation of TEAD transcription factors. This pathway is also involved in some of the most important mechanisms that control the aging processes, such as the AMP-activated protein kinase and sirtuin pathways, along with autophagy and oxidative stress response/antioxidant defense. This could be the link between two tightly connected processes that could open a broader range of targeted molecular therapies to fight aging and cancer. Therefore, available knowledge of the processes involved in the Hippo pathway during aging and cancer must necessarily be well understood.
Collapse
Affiliation(s)
- Yiu To Yeung
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | | | - Mercedes Cano
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mario F Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Antonio Ayala
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
87
|
Gérard D, Schmidt F, Ginolhac A, Schmitz M, Halder R, Ebert P, Schulz MH, Sauter T, Sinkkonen L. Temporal enhancer profiling of parallel lineages identifies AHR and GLIS1 as regulators of mesenchymal multipotency. Nucleic Acids Res 2019; 47:1141-1163. [PMID: 30544251 PMCID: PMC6380961 DOI: 10.1093/nar/gky1240] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 11/23/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023] Open
Abstract
Temporal data on gene expression and context-specific open chromatin states can improve identification of key transcription factors (TFs) and the gene regulatory networks (GRNs) controlling cellular differentiation. However, their integration remains challenging. Here, we delineate a general approach for data-driven and unbiased identification of key TFs and dynamic GRNs, called EPIC-DREM. We generated time-series transcriptomic and epigenomic profiles during differentiation of mouse multipotent bone marrow stromal cell line (ST2) toward adipocytes and osteoblasts. Using our novel approach we constructed time-resolved GRNs for both lineages and identifed the shared TFs involved in both differentiation processes. To take an alternative approach to prioritize the identified shared regulators, we mapped dynamic super-enhancers in both lineages and associated them to target genes with correlated expression profiles. The combination of the two approaches identified aryl hydrocarbon receptor (AHR) and Glis family zinc finger 1 (GLIS1) as mesenchymal key TFs controlled by dynamic cell type-specific super-enhancers that become repressed in both lineages. AHR and GLIS1 control differentiation-induced genes and their overexpression can inhibit the lineage commitment of the multipotent bone marrow-derived ST2 cells.
Collapse
Affiliation(s)
- Deborah Gérard
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Florian Schmidt
- Excellence Cluster for Multimodal Computing and Interaction, Saarland Informatics Campus, 66123 Saarbrücken, Germany
- Computational Biology & Applied Algorithmics, Max Planck Institute for Informatics, Saarland Informatics Campus, 66123 Saarbrücken, Germany
| | - Aurélien Ginolhac
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Martine Schmitz
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
| | - Peter Ebert
- Computational Biology & Applied Algorithmics, Max Planck Institute for Informatics, Saarland Informatics Campus, 66123 Saarbrücken, Germany
| | - Marcel H Schulz
- Excellence Cluster for Multimodal Computing and Interaction, Saarland Informatics Campus, 66123 Saarbrücken, Germany
- Computational Biology & Applied Algorithmics, Max Planck Institute for Informatics, Saarland Informatics Campus, 66123 Saarbrücken, Germany
| | - Thomas Sauter
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Lasse Sinkkonen
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
88
|
Madl CM, LeSavage BL, Dewi RE, Lampe KJ, Heilshorn SC. Matrix Remodeling Enhances the Differentiation Capacity of Neural Progenitor Cells in 3D Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801716. [PMID: 30828535 PMCID: PMC6382308 DOI: 10.1002/advs.201801716] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/09/2018] [Indexed: 05/14/2023]
Abstract
Neural progenitor cells (NPCs) are a promising cell source to repair damaged nervous tissue. However, expansion of therapeutically relevant numbers of NPCs and their efficient differentiation into desired mature cell types remains a challenge. Material-based strategies, including culture within 3D hydrogels, have the potential to overcome these current limitations. An ideal material would enable both NPC expansion and subsequent differentiation within a single platform. It has recently been demonstrated that cell-mediated remodeling of 3D hydrogels is necessary to maintain the stem cell phenotype of NPCs during expansion, but the role of matrix remodeling on NPC differentiation and maturation remains unknown. By culturing NPCs within engineered protein hydrogels susceptible to degradation by NPC-secreted proteases, it is identified that a critical amount of remodeling is necessary to enable NPC differentiation, even in highly degradable gels. Chemical induction of differentiation after sufficient remodeling time results in differentiation into astrocytes and neurotransmitter-responsive neurons. Matrix remodeling modulates expression of the transcriptional co-activator Yes-associated protein, which drives expression of NPC stemness factors and maintains NPC differentiation capacity, in a cadherin-dependent manner. Thus, cell-remodelable hydrogels are an attractive platform to enable expansion of NPCs followed by differentiation of the cells into mature phenotypes for therapeutic use.
Collapse
Affiliation(s)
| | | | - Ruby E. Dewi
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Kyle J. Lampe
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
- Department of Chemical EngineeringUniversity of VirginiaCharlottesvilleVA22904USA
| | - Sarah C. Heilshorn
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| |
Collapse
|
89
|
Chen YA, Lu CY, Cheng TY, Pan SH, Chen HF, Chang NS. WW Domain-Containing Proteins YAP and TAZ in the Hippo Pathway as Key Regulators in Stemness Maintenance, Tissue Homeostasis, and Tumorigenesis. Front Oncol 2019; 9:60. [PMID: 30805310 PMCID: PMC6378284 DOI: 10.3389/fonc.2019.00060] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/21/2019] [Indexed: 12/29/2022] Open
Abstract
The Hippo pathway is a conserved signaling pathway originally defined in Drosophila melanogaster two decades ago. Deregulation of the Hippo pathway leads to significant overgrowth in phenotypes and ultimately initiation of tumorigenesis in various tissues. The major WW domain proteins in the Hippo pathway are YAP and TAZ, which regulate embryonic development, organ growth, tissue regeneration, stem cell pluripotency, and tumorigenesis. Recent reports reveal the novel roles of YAP/TAZ in establishing the precise balance of stem cell niches, promoting the production of induced pluripotent stem cells (iPSCs), and provoking signals for regeneration and cancer initiation. Activation of YAP/TAZ, for example, results in the expansion of progenitor cells, which promotes regeneration after tissue damage. YAP is highly expressed in self-renewing pluripotent stem cells. Overexpression of YAP halts stem cell differentiation and yet maintains the inherent stem cell properties. A success in reprograming iPSCs by the transfection of cells with Oct3/4, Sox2, and Yap expression constructs has recently been shown. In this review, we update the current knowledge and the latest progress in the WW domain proteins of the Hippo pathway in relevance to stem cell biology, and provide a thorough understanding in the tissue homeostasis and identification of potential targets to block tumor development. We also provide the regulatory role of tumor suppressor WWOX in the upstream of TGF-β, Hyal-2, and Wnt signaling that cross talks with the Hippo pathway.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yu Lu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tian-You Cheng
- Department of Optics and Photonics, National Central University, Chungli, Taiwan
| | - Szu-Hua Pan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University, Taipei, Taiwan
| | - Nan-Shan Chang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, United States.,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
90
|
Abstract
Transcription coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ, also known as WWTR1) are homologs of the Drosophila Yorkie (Yki) protein and are major downstream effectors of the evolutionarily conserved Hippo pathway. YAP/TAZ play critical roles in regulation of cell proliferation, apoptosis, and stemness, thus mediate functions of the Hippo pathway in organ size control and tumorigenesis. The Hippo pathway inhibits YAP/TAZ through phosphorylation, which leads to YAP/TAZ cytoplasmic retention and degradation. Dephosphorylated and nuclear-localized YAP/TAZ bind to transcription factors, especially the TEAD family proteins, thus transactivate the expression of specific genes. Therefore, measuring the expression level of YAP/TAZ target genes is a critical approach to assess Hippo pathway activity. Through gene expression profiling in different tissues and cells using techniques such as microarray and RNA-seq, many target genes of YAP/TAZ have been identified. Some of these genes were confirmed to be direct YAP/TAZ targets by chromatin immunoprecipitation (ChIP)-PCR or ChIP-seq. These works made it possible to quickly determine YAP/TAZ activity by measuring the mRNA levels of several YAP/TAZ target genes, such as CTGF, CYR61, and miR-130a by quantitative real-time PCR (qPCR). In this chapter, we demonstrate the use of qPCR to measure YAP/TAZ activity in MCF10A cells.
Collapse
|
91
|
Lokau J, Garbers C. Activating mutations of the gp130/JAK/STAT pathway in human diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 116:283-309. [PMID: 31036294 DOI: 10.1016/bs.apcsb.2018.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cytokines of the interleukin-6 (IL-6) family are involved in numerous physiological and pathophysiological processes. Dysregulated and increased activities of its members can be found in practically all human inflammatory diseases including cancer. All cytokines activate several intracellular signaling cascades, including the Jak/STAT, MAPK, PI3K, and Src/YAP signaling pathways. Additionally, several mutations in proteins involved in these signaling cascades have been identified in human patients, which render these proteins constitutively active and result in a hyperactivation of the signaling pathway. Interestingly, some of these mutations are associated with or even causative for distinct human diseases, making them interesting targets for therapy. This chapter describes the basic biology of the gp130/Jak/STAT pathway, summarizes what is known about the molecular mechanisms of the activating mutations, and gives an outlook how this knowledge can be exploited for targeted therapy in human diseases.
Collapse
Affiliation(s)
- Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
92
|
Afanassieff M, Aksoy I, Beaujean N, Bourillot PY, Savatier P. [Fifty shades of pluripotency]. Med Sci (Paris) 2018; 34:944-953. [PMID: 30526839 DOI: 10.1051/medsci/2018240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Since the derivation of the first pluripotent embryonic stem cell lines in mice in the early 1980s, a plethora of lines has been obtained from various mammalian species including rodents, lagomorphs and primates. These lines are distinguished by their molecular and functional characteristics and correspond to the different pluripotency states observed in the developing embryo between the "blastocyst" and "gastrula" stages. These cell lines are positioned along a gradient, or continuum of pluripotency, the ends of which are epitomized by the naïve and primed states, respectively. Conventional human pluripotent stem cells self-renew in the primed state of pluripotency (ie, at the bottom of the gradient), a position that is undoubtedly the cause of their natural instability. Recent studies aim to generate naive human pluripotent stem cells (at the top of the gradient). The importance of this research in the perspective of medical applications will be discussed.
Collapse
Affiliation(s)
- Marielle Afanassieff
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Irène Aksoy
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Nathalie Beaujean
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Pierre-Yves Bourillot
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Pierre Savatier
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| |
Collapse
|
93
|
Chu WK, Hung LM, Hou CW, Chen JK. Heterogeneous ribonucleoprotein F regulates YAP expression via a G-tract in 3'UTR. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:12-24. [PMID: 30312683 DOI: 10.1016/j.bbagrm.2018.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/05/2018] [Accepted: 10/06/2018] [Indexed: 10/28/2022]
Abstract
The Yes-associated protein (YAP) is a transcription coactivator that plays crucial roles in organ size control and tumorigenesis, and was demonstrated to be inhibited by the Hippo signaling pathway. To date, the molecular mechanisms regulating the expression of YAP in human cells remain unknown. In the present study, we found that hnRNP F and hnRNP U negatively regulate YAP expression. We also showed that downregulation of YAP expression by hnRNP F and hnRNP U was not at the transcriptional level. Knockdown of hnRNP F or hnRNP U increased YAP mRNA stability, suggesting the downregulation of YAP expression was by a post-transcriptional mechanism. A putative hnRNP F binding site was identified in the YAP 3'UTR at 685 to 698, and deletion of this putative hnRNP F element abolished the down-regulation effect of YAP mRNA stability by hnRNP F. Binding of the hnRNP F to the YAP 3'UTR was demonstrated by Cross-linked RNA Immunoprecipitation. mRNA stability is a possible secondary effect of alternative splicing or other nuclear process. Understanding the regulation of YAP expression would provide insights into the mechanisms underlying the maintenance of tissue size homeostasis and tumorigenesis.
Collapse
Affiliation(s)
- Wing-Keung Chu
- Department of Physiology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Healthy and Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Li-Man Hung
- Healthy and Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan; Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chun-Wei Hou
- Healthy and Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Jan-Kan Chen
- Department of Physiology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Healthy and Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
94
|
PYK2 negatively regulates the Hippo pathway in TNBC by stabilizing TAZ protein. Cell Death Dis 2018; 9:985. [PMID: 30250159 PMCID: PMC6155151 DOI: 10.1038/s41419-018-1005-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/05/2018] [Accepted: 08/27/2018] [Indexed: 01/01/2023]
Abstract
The tumor suppressor Hippo pathway negatively regulates the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) to inhibit cell growth and control organ size, whereas activation of YAP and TAZ is implicated in tumorigenesis and cancer metastasis. Here, we report that the nonreceptor tyrosine kinase PYK2 positively regulates TAZ and YAP transcriptional activity in triple-negative breast cancer (TNBC). We found that inhibition of PYK2 expression or its kinase activity substantially affects the steady-state level of TAZ and markedly facilitates its proteasomal degradation. This effect was specific to PYK2 inhibition and was not obtained by inhibition of FAK. Destabilization of TAZ was associated with profound effect of PYK2 inhibition on cell growth at low-density concomitant with reduced expression of TAZ-target genes and induction of cell apoptosis. We further show that PYK2 enhances the tyrosine phosphorylation of both TAZ and LATS1/2 and concomitantly TAZ stability, and that PYK2 protein level correlates with the level of TAZ protein in primary breast tumors. Together these observations suggest that PYK2 is an important regulator of the Hippo pathway, and its tyrosine kinase activity has a striking effect on TAZ stabilization and activation in TNBC.
Collapse
|
95
|
Mechanoregulation and pathology of YAP/TAZ via Hippo and non-Hippo mechanisms. Clin Transl Med 2018; 7:23. [PMID: 30101371 PMCID: PMC6087706 DOI: 10.1186/s40169-018-0202-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023] Open
Abstract
Yes-associated protein (YAP) and its paralog WW domain containing transcription regulator 1 (TAZ) are important regulators of multiple cellular functions such as proliferation, differentiation, and survival. On the tissue level, YAP/TAZ are essential for embryonic development, organ size control and regeneration, while their deregulation leads to carcinogenesis or other diseases. As an underlying principle for YAP/TAZ-mediated regulation of biological functions, a growing body of research reveals that YAP/TAZ play a central role in delivering information of mechanical environments surrounding cells to the nucleus transcriptional machinery. In this review, we discuss mechanical cue-dependent regulatory mechanisms for YAP/TAZ functions, as well as their clinical significance in cancer progression and treatment.
Collapse
|
96
|
Chen X, Li J, Huang Y, Liu P, Fan Y. Insoluble Microenvironment Facilitating the Generation and Maintenance of Pluripotency. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:267-278. [PMID: 29327674 DOI: 10.1089/ten.teb.2017.0415] [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] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold enormous potential as a tool to generate cells for tissue engineering and regenerative medicine. Since the initial report of iPSCs in 2006, many different methods have been developed to enhance the safety and efficiency of this technology. Recent studies indicate that the extracellular signals can promote the production of iPSCs, and even replace the Yamanaka factors. Noticeably, abundant evidences suggest that the insoluble microenvironment, including the culture substrate and neighboring cells, directly regulates the expression of core pluripotency genes and the epigenetic modification of the chromatins, hence, impacts the reprogramming dynamics. These studies provide new strategies for developing safer and more efficient method for iPSC generation. In this review, we examine the publications addressing the insoluble extracellular microenvironment that boosts iPSC generation and self-renewal. We also discuss cell adhesion-mediated molecular mechanisms, through which the insoluble extracellular cues interplay with reprogramming.
Collapse
Affiliation(s)
- Xiaofang Chen
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Jiaqi Li
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
| | - Yan Huang
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Peng Liu
- 3 Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University , Beijing, China
| | - Yubo Fan
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
- 4 National Research Center for Rehabilitation Technical Aids , Beijing, China
| |
Collapse
|
97
|
Ege N, Dowbaj AM, Jiang M, Howell M, Hooper S, Foster C, Jenkins RP, Sahai E. Quantitative Analysis Reveals that Actin and Src-Family Kinases Regulate Nuclear YAP1 and Its Export. Cell Syst 2018; 6:692-708.e13. [PMID: 29909276 PMCID: PMC6035388 DOI: 10.1016/j.cels.2018.05.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 02/08/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
The transcriptional regulator YAP1 is critical for the pathological activation of fibroblasts. In normal fibroblasts, YAP1 is located in the cytoplasm, while in activated cancer-associated fibroblasts, it is nuclear and promotes the expression of genes required for pro-tumorigenic functions. Here, we investigate the dynamics of YAP1 shuttling in normal and activated fibroblasts, using EYFP-YAP1, quantitative photobleaching methods, and mathematical modeling. Imaging of migrating fibroblasts reveals the tight temporal coupling of cell shape change and altered YAP1 localization. Both 14-3-3 and TEAD binding modulate YAP1 shuttling, but neither affects nuclear import. Instead, we find that YAP1 nuclear accumulation in activated fibroblasts results from Src and actomyosin-dependent suppression of phosphorylated YAP1 export. Finally, we show that nuclear-constrained YAP1, upon XPO1 depletion, remains sensitive to blockade of actomyosin function. Together, these data place nuclear export at the center of YAP1 regulation and indicate that the cytoskeleton can regulate YAP1 within the nucleus.
Collapse
Affiliation(s)
- Nil Ege
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Cell and Developmental Biology Department, University College London, London WC1E 6BT, UK
| | - Anna M Dowbaj
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Ming Jiang
- High Throughput Screening, The Francis Crick Institute, London NW1 1AT, UK
| | - Michael Howell
- High Throughput Screening, The Francis Crick Institute, London NW1 1AT, UK
| | - Steven Hooper
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Charles Foster
- Transcription Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Robert P Jenkins
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
98
|
Dehghanian F, Hojati Z, Esmaeili F, Masoudi-Nejad A. Network-based expression analyses and experimental validations revealed high co-expression between Yap1 and stem cell markers compared to differentiated cells. Genomics 2018; 111:831-839. [PMID: 29775782 DOI: 10.1016/j.ygeno.2018.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/22/2018] [Accepted: 05/08/2018] [Indexed: 01/06/2023]
Abstract
The Hippo signaling pathway is identified as a potential regulatory pathway which plays critical roles in differentiation and stem cell self-renewal. Yap1 is a primary transcriptional effector of this pathway. The importance of Yap1 in embryonic stem cells (ESCs) and differentiation procedure remains a challenging question, since two different observations have been reported. To answer this question we used co-expression network and differential co-expression analyses followed by experimental validations. Our results indicate that Yap1 is highly co-expressed with stem cell markers in ESCs but not in differentiated cells (DCs). The significant Yap1 down-regulation and also translocation of Yap1 into the cytoplasm during P19 differentiation was also detected. Moreover, our results suggest the E2f7, Lin28a and Dppa4 genes as possible regulatory nuclear factors of Hippo pathway in stem cells. The present findings are actively consistent with studies that suggested Yap1 as an essential factor for stem cell self-renewal.
Collapse
Affiliation(s)
- Fariba Dehghanian
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Zohreh Hojati
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | - Fariba Esmaeili
- Division of Animal Sciences, Department of Biology, Faculty of Sciences, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
99
|
Papatsenko D, Waghray A, Lemischka IR. Feedback control of pluripotency in embryonic stem cells: Signaling, transcription and epigenetics. Stem Cell Res 2018; 29:180-188. [DOI: 10.1016/j.scr.2018.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 02/06/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022] Open
|
100
|
Gopinath M, Di Liddo R, Marotta F, Murugesan R, Banerjee A, Sriramulu S, Jothimani G, Subramaniam VD, Narasimhan S, Priya K S, Sun XF, Pathak S. Role of Hippo Pathway Effector Tafazzin Protein in Maintaining Stemness of Umbilical Cord-Derived Mesenchymal Stem Cells (UC-MSC). Int J Hematol Oncol Stem Cell Res 2018; 12:153-165. [PMID: 30233778 PMCID: PMC6141435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/24/2017] [Indexed: 11/22/2022] Open
Abstract
Tafazzin (TAZ) protein has been upregulated in various types of human cancers, although the basis for elevation is uncertain, it has been made definite that the effect of mutation in the hippo pathway, particularly when it is switched off, considerably activates tafazzin transcriptionally and thus this results in tissue or tumor overgrowth. Recent perceptions into the activity of tafazzin, have ascribed to it, a role as stem cell factor in mouse mesenchymal and as well as in neural stem cells. Being a downstream molecule in Hippo signalling, phosphorylation or dephosphorylation of tafazzin gene regulates its transcriptional activity and the stemness of mesenchymal stem cells. Commonly, extracellular matrix controls the stem cell fate commitment and perhaps tafazzin controls stemness through altering the extra cellular matrix. Extracellular matrix is generally made up of prime proteoglycans and the fate stabilization of the resulting lineages is surveilled by engineering these glycans. Tafazzin degradation and addition of proteoglycans affect physical attributes of the extracellular matrix that drives cell differentiation into various lineages. Thus, tafazzin along with major glycans present in the extracellular matrix is involved in imparting stemness. However, there are incoherent molecular events, wherein both tafazzin and the extracellular matrix components, together either activate or inhibit differentiation of stem cells. This review discusses about the role of tafazzin oncoprotein as a stemness factor.
Collapse
Affiliation(s)
- Madhumala Gopinath
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Rosa Di Liddo
- Department of Pharmacology and Pharmaceutical Sciences, University of Padova, Padova, Italy
| | - Francesco Marotta
- ReGenera R&D International for Aging Intervention, Milano-Beijing, Italy-China, VCC Preventive Medical Promotion Foundation, Beijing, China
| | - Ramachandran Murugesan
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Antara Banerjee
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Sushmitha Sriramulu
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Ganesan Jothimani
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Vimala Devi Subramaniam
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Srinivasan Narasimhan
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Swarna Priya K
- Department of Gynecology and Pediatrics, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| | - Xiao-Feng Sun
- Department of Oncology and Department of Clinical and Experimental Medicine, University of Linköping, Linköping, Sweden
| | - Surajit Pathak
- Department of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai-603103, India
| |
Collapse
|