51
|
Ghomlaghi M, Hart A, Hoang N, Shin S, Nguyen LK. Feedback, Crosstalk and Competition: Ingredients for Emergent Non-Linear Behaviour in the PI3K/mTOR Signalling Network. Int J Mol Sci 2021; 22:6944. [PMID: 34203293 PMCID: PMC8267830 DOI: 10.3390/ijms22136944] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The PI3K/mTOR signalling pathway plays a central role in the governing of cell growth, survival and metabolism. As such, it must integrate and decode information from both external and internal sources to guide efficient decision-making by the cell. To facilitate this, the pathway has evolved an intricate web of complex regulatory mechanisms and elaborate crosstalk with neighbouring signalling pathways, making it a highly non-linear system. Here, we describe the mechanistic biological details that underpin these regulatory mechanisms, covering a multitude of negative and positive feedback loops, feed-forward loops, competing protein interactions, and crosstalk with major signalling pathways. Further, we highlight the non-linear and dynamic network behaviours that arise from these regulations, uncovered through computational and experimental studies. Given the pivotal role of the PI3K/mTOR network in cellular homeostasis and its frequent dysregulation in pathologies including cancer and diabetes, a coherent and systems-level understanding of the complex regulation and consequential dynamic signalling behaviours within this network is imperative for advancing biology and development of new therapeutic approaches.
Collapse
Affiliation(s)
- Milad Ghomlaghi
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Anthony Hart
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Nhan Hoang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
| | - Sungyoung Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Lan K. Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
52
|
Pojer JM, Saiful Hilmi AJ, Kondo S, Harvey KF. Crumbs and the apical spectrin cytoskeleton regulate R8 cell fate in the Drosophila eye. PLoS Genet 2021; 17:e1009146. [PMID: 34097697 PMCID: PMC8211197 DOI: 10.1371/journal.pgen.1009146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 06/17/2021] [Accepted: 05/11/2021] [Indexed: 12/31/2022] Open
Abstract
The Hippo pathway is an important regulator of organ growth and cell fate. In the R8 photoreceptor cells of the Drosophila melanogaster eye, the Hippo pathway controls the fate choice between one of two subtypes that express either the blue light-sensitive Rhodopsin 5 (Hippo inactive R8 subtype) or the green light-sensitive Rhodopsin 6 (Hippo active R8 subtype). The degree to which the mechanism of Hippo signal transduction and the proteins that mediate it are conserved in organ growth and R8 cell fate choice is currently unclear. Here, we identify Crumbs and the apical spectrin cytoskeleton as regulators of R8 cell fate. By contrast, other proteins that influence Hippo-dependent organ growth, such as the basolateral spectrin cytoskeleton and Ajuba, are dispensable for the R8 cell fate choice. Surprisingly, Crumbs promotes the Rhodopsin 5 cell fate, which is driven by Yorkie, rather than the Rhodopsin 6 cell fate, which is driven by Warts and the Hippo pathway, which contrasts with its impact on Hippo activity in organ growth. Furthermore, neither the apical spectrin cytoskeleton nor Crumbs appear to regulate the Hippo pathway through mechanisms that have been observed in growing organs. Together, these results show that only a subset of Hippo pathway proteins regulate the R8 binary cell fate decision and that aspects of Hippo signalling differ between growing organs and post-mitotic R8 cells.
Collapse
Affiliation(s)
- Jonathan M. Pojer
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdul Jabbar Saiful Hilmi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Shu Kondo
- Laboratory of Invertebrate Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Kieran F. Harvey
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
53
|
Mohajan S, Jaiswal PK, Vatanmakarian M, Yousefi H, Sankaralingam S, Alahari SK, Koul S, Koul HK. Hippo pathway: Regulation, deregulation and potential therapeutic targets in cancer. Cancer Lett 2021; 507:112-123. [PMID: 33737002 PMCID: PMC10370464 DOI: 10.1016/j.canlet.2021.03.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/25/2023]
Abstract
Hippo pathway is a master regulator of development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size control. Hippo pathway relays signals from different extracellular and intracellular events to regulate cell behavior and functions. Hippo pathway is conserved from Protista to eukaryotes. Deregulation of the Hippo pathway is associated with numerous cancers. Alteration of the Hippo pathway results in cell invasion, migration, disease progression, and therapy resistance in cancers. However, the function of the various components of the mammalian Hippo pathway is yet to be elucidated in detail especially concerning tumor biology. In the present review, we focused on the Hippo pathway in different model organisms, its regulation and deregulation, and possible therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Suman Mohajan
- Department of Biochemistry and Molecular Biology, LSUHSC, Shreveport, USA
| | - Praveen Kumar Jaiswal
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA; Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA
| | - Mousa Vatanmakarian
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | | | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA; Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA
| | - Sweaty Koul
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA
| | - Hari K Koul
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA; Urology, LSUHSC, School of Medicine, New Orleans, USA; Stanley S. Scott Cancer Center, LSUHSC, New Orleans, USA.
| |
Collapse
|
54
|
Rhee JE, Choi JH, Park JH, Lee G, Pak B, Kwon SH, Jeon SH. CG11426 gene product negatively regulates glial population size in the Drosophila eye imaginal disc. Dev Neurobiol 2021; 81:805-816. [PMID: 34047015 DOI: 10.1002/dneu.22838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/03/2021] [Accepted: 05/16/2021] [Indexed: 11/09/2022]
Abstract
Glial cells play essential roles in the nervous system. Although glial populations are tightly regulated, the mechanisms regulating the population size remain poorly understood. Since Drosophila glial cells are similar to the human counterparts in their functions and shapes, rendering them an excellent model system to understand the human glia biology. Lipid phosphate phosphatases (LPPs) are important for regulating bioactive lipids. In Drosophila, there are three known LPP-encoding genes: wunen, wunen-2, and lazaro. The wunens are important for germ cell migration and survival and septate junction formation during tracheal development. Lazaro is involved in phototransduction. In the present study, we characterized a novel Drosophila LPP-encoding gene, CG11426. Suppression of CG11426 increased glial cell number in the eye imaginal disc during larval development, while ectopic CG11426 expression decreased it. Both types of mutation also caused defects in axon projection to the optic lobe in larval eye-brain complexes. Moreover, CG11426 promoted apoptosis via inhibiting ERK signaling in the eye imaginal disc. Taken together, these findings demonstrated that CG11426 gene product negatively regulates ERK signaling to promote apoptosis for proper maintenance of the glial population in the developing eye disc.
Collapse
Affiliation(s)
- Jong-Eun Rhee
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| | - Jin-Hyeon Choi
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| | - Jae H Park
- Department of Biochemistry & Cellular and Molecular Biology, and Neuronet Research Center, University of Tennessee, Knoxville, Tennessee, USA
| | - Gyunghee Lee
- Department of Biochemistry & Cellular and Molecular Biology, and Neuronet Research Center, University of Tennessee, Knoxville, Tennessee, USA
| | - Banya Pak
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hae Kwon
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Sang-Hak Jeon
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
55
|
Zheng Q, Chen X, Qiao C, Wang M, Chen W, Luan X, Yan Y, Shen C, Fang J, Hu X, Zheng B, Wu Y, Yu J. Somatic CG6015 mediates cyst stem cell maintenance and germline stem cell differentiation via EGFR signaling in Drosophila testes. Cell Death Discov 2021; 7:68. [PMID: 33824283 PMCID: PMC8024382 DOI: 10.1038/s41420-021-00452-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/21/2021] [Accepted: 03/18/2021] [Indexed: 11/09/2022] Open
Abstract
Stem cell niche is regulated by intrinsic and extrinsic factors. In the Drosophila testis, cyst stem cells (CySCs) support the differentiation of germline stem cells (GSCs). However, the underlying mechanisms remain unclear. In this study, we found that somatic CG6015 is required for CySC maintenance and GSC differentiation in a Drosophila model. Knockdown of CG6015 in CySCs caused aberrant activation of dpERK in undifferentiated germ cells in the Drosophila testis, and disruption of key downstream targets of EGFR signaling (Dsor1 and rl) in CySCs results in a phenotype resembling that of CG6015 knockdown. CG6015, Dsor1, and rl are essential for the survival of Drosophila cell line Schneider 2 (S2) cells. Our data showed that somatic CG6015 regulates CySC maintenance and GSC differentiation via EGFR signaling, and inhibits aberrant activation of germline dpERK signals. These findings indicate regulatory mechanisms of stem cell niche homeostasis in the Drosophila testis.
Collapse
Affiliation(s)
- Qianwen Zheng
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Xia Chen
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Chen Qiao
- Department of Clinical Pharmacy, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Min Wang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Wanyin Chen
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Xiaojin Luan
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Yidan Yan
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, 215002, Suzhou, Jiangsu, P.R. China
| | - Jie Fang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Xing Hu
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, 215002, Suzhou, Jiangsu, P.R. China.
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, 214062, Wuxi, Jiangsu, P.R. China.
| | - Jun Yu
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, 212001, Zhenjiang, Jiangsu, P.R. China.
| |
Collapse
|
56
|
Low dose of zearalenone elevated colon cancer cell growth through G protein-coupled estrogenic receptor. Sci Rep 2021; 11:7403. [PMID: 33795755 PMCID: PMC8016995 DOI: 10.1038/s41598-021-86788-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Colon cancer is one of the leading causes of cancer death worldwide. It is widely believed that environmental factors contribute to colon cancer development. Zearalenone (ZEA) is non-steroidal estrogenic mycotoxin that is widely found in the human diet and animal feeds. Most cancer studies of ZEA focused on estrogen sensitive cancers, while few focused on other types, such as colon cancer; despite the gastrointestinal tract being the first barrier exposed to food contaminants. This study investigated the stimulatory effects of ZEA on colon cancer cell lines and their underlying molecular mechanisms. ZEA promoted anchorage independent cell growth and cell cycle progression through promoting G1-to-S phase transition. Proliferative marker, cyclin D1 and Ki67 were found to be upregulated upon ZEA treatment. G protein-coupled estrogenic receptor 1 (GPER) protein expression was promoted upon ZEA treatment suggesting the involvement of GPER. The growth promoting effect mediated through GPER were suppressed by its antagonist G15. ZEA were found to promote the downstream parallel pathway, MAPK signaling pathway and Hippo pathway effector YAP1. Altogether, our observations suggest a novel mechanism by which ZEA could promote cancer growth and provide a new perspective on the carcinogenicity of ZEA.
Collapse
|
57
|
Venkatramanan S, Ibar C, Irvine KD. TRIP6 is required for tension at adherens junctions. J Cell Sci 2021; 134:jcs247866. [PMID: 33558314 PMCID: PMC7970510 DOI: 10.1242/jcs.247866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
Hippo signaling mediates influences of cytoskeletal tension on organ growth. TRIP6 and LIMD1 have each been identified as being required for tension-dependent inhibition of the Hippo pathway LATS kinases and their recruitment to adherens junctions, but the relationship between TRIP6 and LIMD1 was unknown. Using siRNA-mediated gene knockdown, we show that TRIP6 is required for LIMD1 localization to adherens junctions, whereas LIMD1 is not required for TRIP6 localization. TRIP6, but not LIMD1, is also required for the recruitment of vinculin and VASP to adherens junctions. Knockdown of TRIP6 or vinculin, but not of LIMD1, also influences the localization of myosin and F-actin. In TRIP6 knockdown cells, actin stress fibers are lost apically but increased basally, and there is a corresponding increase in the recruitment of vinculin and VASP to basal focal adhesions. Our observations identify a role for TRIP6 in organizing F-actin and maintaining tension at adherens junctions that could account for its influence on LIMD1 and LATS. They also suggest that focal adhesions and adherens junctions compete for key proteins needed to maintain attachments to contractile F-actin.
Collapse
Affiliation(s)
- Srividya Venkatramanan
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Consuelo Ibar
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| |
Collapse
|
58
|
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
|
59
|
Le Y, He Y, Bai M, Wang Y, Wu J, Yu L. Knockout of Ajuba Attenuates the Growth and Migration of Hepatocellular Carcinoma Cells. Cytogenet Genome Res 2021; 160:650-658. [PMID: 33640888 DOI: 10.1159/000512264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 10/12/2020] [Indexed: 11/19/2022] Open
Abstract
Ajuba has been found to be mutated or aberrantly regulated in several human cancers and plays important roles in cancer progression via different signaling pathways. However, little is known about the role of Ajuba in hepatocellular carcinoma (HCC). Here, we found an upregulation of Ajuba expression in HCC tissues compared with normal liver tissues, while a poor prognosis was observed in HCC patients with high Ajuba expression. Knockout of Ajuba in HCC cells inhibited cell growth in vitro and in vivo, suppressed cell migration, and enhanced the cell apoptosis under stress. Moreover, re-expression of Ajuba in Ajuba-deficient cells could restore the phenotype of Ajuba-deficient cells. In conclusion, these results indicate that Ajuba is upregulated in HCC and promotes cell growth and migration of HCC cells, suggesting that Ajuba could possibly be a new target for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Yichen Le
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yi He
- School of Life Sciences, Fudan University, Shanghai, China
| | - Meirong Bai
- School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Ying Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Jiaxue Wu
- School of Life Sciences, Fudan University, Shanghai, China,
| | - Long Yu
- School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
60
|
Schleicher K, Schramek D. AJUBA: A regulator of epidermal homeostasis and cancer. Exp Dermatol 2021; 30:546-559. [PMID: 33372298 DOI: 10.1111/exd.14272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/09/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022]
Abstract
The epidermis, outermost layer of the skin, is constantly renewing itself through proliferative and differentiation processes. These processes are vital to maintain proper epidermal integrity during skin development and homeostasis and for preventing skin diseases and cancers. The biological mechanisms that permit this balancing act are vast, where individual pathway regulators are known, but the exact regulatory control and cross-talk between simultaneously turning one biological pathway on and an opposing one off remain elusive. This review explores the diverse roles the scaffolding protein AJUBA plays during epidermal homeostasis and cancer. Initially identified for its role in promoting meiotic progression in oocytes through Grb2 and MAP kinase activity, AJUBA also maintains cytoskeletal tension permitting epidermal tissue development and responds to retinoic acid committing cells to initiate development of surface epidermal layer. AJUBA regulates proliferation of skin stem cells through Hippo and Wnt signalling and encourages mitotic commitment through Aurora-A, Aurora-B and CDK1. In addition, AJUBA also induces epidermal differentiation to maintain appropriate epidermal thickness and barrier function by activating Notch signalling and stabilizing catenins and actin during cellular remodelling. AJUBA also plays an imperative context-dependent tumor-promoting and tumor-suppressive role within epithelial cancers. AJUBA's abundant roles within the epidermis signify its importance as a molecular switchboard, vetting multiple signalling pathways to control epidermal biology.
Collapse
Affiliation(s)
- Krista Schleicher
- Molecular, Structural and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada.,Faculty of Medicine, Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Daniel Schramek
- Molecular, Structural and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada.,Faculty of Medicine, Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
61
|
Feltri ML, Weaver MR, Belin S, Poitelon Y. The Hippo pathway: Horizons for innovative treatments of peripheral nerve diseases. J Peripher Nerv Syst 2021; 26:4-16. [PMID: 33449435 DOI: 10.1111/jns.12431] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022]
Abstract
Initially identified in Drosophila, the Hippo signaling pathway regulates how cells respond to their environment by controlling proliferation, migration and differentiation. Many recent studies have focused on characterizing Hippo pathway function and regulation in mammalian cells. Here, we present a brief overview of the major components of the Hippo pathway, as well as their regulation and function. We comprehensively review the studies that have contributed to our understanding of the Hippo pathway in the function of the peripheral nervous system and in peripheral nerve diseases. Finally, we discuss innovative approaches that aim to modulate Hippo pathway components in diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
62
|
Li Y, Xu C, Wang H, Liu X, Jiang L, Liang S, Wu Z, Wang Z, Zhou J, Xiao W, Guo Z, Wang Y. Systems pharmacology reveals the multi-level synergetic mechanism of action of Ginkgo biloba L. leaves for cardiomyopathy treatment. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113279. [PMID: 32810617 DOI: 10.1016/j.jep.2020.113279] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/06/2020] [Accepted: 08/11/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiomyopathy is a common cause of heart failure and may lead to increased risk of sudden cardiac death, lacking simple, safe and effective treatment strategies due to unclear pathogenesis. Ginkgo biloba L. leaves (GBLs), a traditional Chinese medicine (TCM), has been widely used in clinical medicine for improving blood circulation, and was demonstrated to be effective on cardiomyopathy in preclinical studies. However, because of the widely known holistic therapeutic philosophy via multi-target and multi-pathway effect for most TCMs, to explore its underlying molecular mechanisms of action (MoA) remains a great challenge. AIM OF STUDY Decipher the underlying MoA of GBLs for cardiomyopathy treatment: Study design and methods: An integrated systems pharmacology framework was employed to screen potential active compounds, identify therapeutic targets, explore the action pathways and verify mechanisms of GBLs with in vitro experiments. RESULTS We firstly confirmed the therapeutic effect of GBLs on cardiomyopathy and subsequently screened 27 active compounds from GBLs according to their pharmacokinetic properties. Then Probability Ensemble Approach was applied to identify the compound combinations that exert synergetic effect from GBLs. Network analysis and functional enrichment analysis demonstrated that these compounds exhibit synergistic therapeutic effect by acting on multiple targets and thereby regulating multiple pathways mainly involved in pro-survival, anti-apoptotic and anti-inflammatory processes. Finally, using a doxorubicin-induced myocardial injury model, therapeutic effect of ginkgolide A, ginkgolide B, isorhamnetin, as well as their synergistic effect on PI3K-AKT and NF-κB signaling pathways were validated in vitro. Importantly, we demonstrated that Ginkgo diterpene lactone meglumine injection (GDJ), an approved injection derived from GBLs, could be a promising agent for cardiomyopathy treatment. CONCLUSION Collectively, the multi-level synergetic mechanism of GBLs on cardiomyopathy treatment was demonstrated with systems pharmacology approach, providing a paradigm for deciphering the complicated MoA of TCMs.
Collapse
Affiliation(s)
- Yueping Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, Xinjiang, 832002, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu, 222002, China.
| | - Chi Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China.
| | - Haiqing Wang
- College of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China.
| | - Xiaobing Liu
- Department of General Surgery, Chinese People's Liberation Army 69224 Army Hospital, Kuche, Xinjiang, 842000, China.
| | - Li Jiang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, Xinjiang, 832002, China.
| | - Shengnan Liang
- School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Ziyin Wu
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu, 222002, China.
| | - Zhenzhong Wang
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu, 222002, China
| | - Jun Zhou
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu, 222002, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu, 222002, China.
| | - Zihu Guo
- College of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China.
| | - Yonghua Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, Xinjiang, 832002, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu, 222002, China; College of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
63
|
Szulzewsky F, Holland EC, Vasioukhin V. YAP1 and its fusion proteins in cancer initiation, progression and therapeutic resistance. Dev Biol 2021; 475:205-221. [PMID: 33428889 DOI: 10.1016/j.ydbio.2020.12.018] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/14/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
YAP1 is a transcriptional co-activator whose activity is controlled by the Hippo signaling pathway. In addition to important functions in normal tissue homeostasis and regeneration, YAP1 has also prominent functions in cancer initiation, aggressiveness, metastasis, and therapy resistance. In this review we are discussing the molecular functions of YAP1 and its roles in cancer, with a focus on the different mechanisms of de-regulation of YAP1 activity in human cancers, including inactivation of upstream Hippo pathway tumor suppressors, regulation by intersecting pathways, miRNAs, and viral oncogenes. We are also discussing new findings on the function and biology of the recently identified family of YAP1 gene fusions, that constitute a new type of activating mutation of YAP1 and that are the likely oncogenic drivers in several subtypes of human cancers. Lastly, we also discuss different strategies of therapeutic inhibition of YAP1 functions.
Collapse
Affiliation(s)
- Frank Szulzewsky
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Eric C Holland
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA; Seattle Tumor Translational Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Valeri Vasioukhin
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| |
Collapse
|
64
|
Samji P, Rajendran MK, Warrier VP, Ganesh A, Devarajan K. Regulation of Hippo signaling pathway in cancer: A MicroRNA perspective. Cell Signal 2020; 78:109858. [PMID: 33253912 DOI: 10.1016/j.cellsig.2020.109858] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
Recent studies have suggested that Hippo signaling is not only involved in controlling organ size in Drosophila but can also regulate cell proliferation, tissue homeostasis, differentiation, apoptosis and regeneration. Any dysregulation of Hippo signaling, especially the hyper activation of its downstream effectors YAP/TAZ, can lead to uncontrolled cell proliferation and malignant transformation. In majority of cancers, expression of YAP/TAZ is extremely high and this increased expression of YAP/TAZ has been shown to be an independent predictor of prognosis and indicator of increased cell proliferation, metastasis and poor survival. In this review, we have summarized the most recent findings about the cross talk of Hippo signaling pathway with other signaling pathways and its regulation by different miRNAs in various cancer types. Recent evidence has suggested that Hippo pathway is also involved in mediating the resistance of different cancer cells to chemotherapeutic drugs and in a few cancer types, this is brought about by regulating miRNAs. Therefore, the delineation of the underlying mechanisms regulating the chemotherapeutic resistance might help in developing better treatment options. This review has attempted to provide an overview of different drugs/options which can be utilized to target oncogenic YAP/TAZ proteins for therapeutic interventions.
Collapse
Affiliation(s)
- Priyanka Samji
- Cancer Biology Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India.
| | - Manoj K Rajendran
- Cancer Biology Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India
| | - Vidya P Warrier
- Cancer Biology Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India
| | - Akshayaa Ganesh
- Cancer Biology Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India
| | - Karunagaran Devarajan
- Cancer Biology Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India
| |
Collapse
|
65
|
Wang H, Wang J, Zhang S, Jia J, Liu X, Zhang J, Wang P, Song X, Che L, Liu K, Ribback S, Cigliano A, Evert M, Wu H, Calvisi DF, Zeng Y, Chen X. Distinct and Overlapping Roles of Hippo Effectors YAP and TAZ During Human and Mouse Hepatocarcinogenesis. Cell Mol Gastroenterol Hepatol 2020; 11:1095-1117. [PMID: 33232824 PMCID: PMC7903139 DOI: 10.1016/j.jcmgh.2020.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND & AIMS Yes-associated protein (YAP) and its paralog transcriptional co-activator with post synaptic density protein, drosophila disc large tumor suppressor and zonula occludens-1-binding motif (TAZ) are 2 co-activators downstream of Hippo tumor-suppressor cascade. Both have been implicated in the development of hepatocellular carcinoma (HCC). However, whether YAP and TAZ have distinct or overlapping functions during hepatocarcinogenesis remains unknown. METHODS Expression patterns of YAP and TAZ were analyzed in human HCC samples. The requirement of Yap and/or Taz in protein kinase B (Akt)/ neuroblastoma RAS viral oncogene homolog (NRas) -driven liver tumorigenesis was analyzed using conditional Yap, Taz, and Yap;Taz knockout mice. Transcriptional programs regulated by YAP and/or TAZ were identified via RNA sequencing. RESULTS We found that in human HCC samples, an almost ubiquitous activation of YAP or TAZ occurs, underlying their role in this tumor type. Intriguingly, 70% of HCC samples showed only nuclear YAP or TAZ immunoreactivity. In the Akt/NRas liver tumor model, where nuclear Yap and Taz can be detected readily, deletion of Yap or Taz alone only mildly delayed liver tumor development, whereas their concomitant ablation strongly inhibited tumor cell proliferation and significantly suppressed Akt/NRas-driven hepatocarcinogenesis. In HCC cell lines, silencing of either YAP or TAZ led to decreased expression of both overlapping and distinct sets of genes, with the most prominent gene signatures related to cell-cycle progression and DNA replication. CONCLUSIONS YAP and TAZ have overlapping and distinct roles in hepatocarcinogenesis. HCCs may display unique activation of YAP or TAZ, thus relying on either YAP or TAZ for their growth.
Collapse
Affiliation(s)
- Haichuan Wang
- Liver Transplantation Division, Department of Liver Surgery, Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Shanshan Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Jiaoyuan Jia
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Department of Oncology and Hematology, The Second Hospital, Jilin University, Changchun, China
| | - Xianqiong Liu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; School of Pharmacy, Hubei University of Chinese Medicine Wuhan, Hubei, China
| | - Jie Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Pan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Ke Liu
- Department of Pediatrics and Human Development, East Lansing, Michigan; Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, Michigan
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Antonio Cigliano
- Institute of Pathology, University Clinic of Regensburg, Regensburg, Germany
| | - Matthias Evert
- Institute of Pathology, University Clinic of Regensburg, Regensburg, Germany
| | - Hong Wu
- Liver Transplantation Division, Department of Liver Surgery, Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Diego F Calvisi
- Institute of Pathology, University Clinic of Regensburg, Regensburg, Germany.
| | - Yong Zeng
- Liver Transplantation Division, Department of Liver Surgery, Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California.
| |
Collapse
|
66
|
Vigneswaran K, Boyd NH, Oh SY, Lallani S, Boucher A, Neill SG, Olson JJ, Read RD. YAP/TAZ Transcriptional Coactivators Create Therapeutic Vulnerability to Verteporfin in EGFR-mutant Glioblastoma. Clin Cancer Res 2020; 27:1553-1569. [PMID: 33172899 DOI: 10.1158/1078-0432.ccr-20-0018] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 08/04/2020] [Accepted: 11/06/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastomas (GBMs), neoplasms derived from glia and neuroglial progenitor cells, are the most common and lethal malignant primary brain tumors diagnosed in adults, with a median survival of 14 months. GBM tumorigenicity is often driven by genetic aberrations in receptor tyrosine kinases, such as amplification and mutation of EGFR. EXPERIMENTAL DESIGN Using a Drosophila glioma model and human patient-derived GBM stem cells and xenograft models, we genetically and pharmacologically tested whether the YAP and TAZ transcription coactivators, effectors of the Hippo pathway that promote gene expression via TEA domain (TEAD) cofactors, are key drivers of GBM tumorigenicity downstream of oncogenic EGFR signaling. RESULTS YAP and TAZ are highly expressed in EGFR-amplified/mutant human GBMs, and their knockdown in EGFR-amplified/mutant GBM cells inhibited proliferation and elicited apoptosis. Our results indicate that YAP/TAZ-TEAD directly regulates transcription of SOX2, C-MYC, and EGFR itself to create a feedforward loop to drive survival and proliferation of human GBM cells. Moreover, the benzoporphyrin derivative verteporfin, a disruptor of YAP/TAZ-TEAD-mediated transcription, preferentially induced apoptosis of cultured patient-derived EGFR-amplified/mutant GBM cells, suppressed expression of YAP/TAZ transcriptional targets, including EGFR, and conferred significant survival benefit in an orthotopic xenograft GBM model. Our efforts led us to design and initiate a phase 0 clinical trial of Visudyne, an FDA-approved liposomal formulation of verteporfin, where we used intraoperative fluorescence to observe verteporfin uptake into tumor cells in GBM tumors in human patients. CONCLUSIONS Together, our data suggest that verteporfin is a promising therapeutic agent for EGFR-amplified and -mutant GBM.
Collapse
Affiliation(s)
| | - Nathaniel H Boyd
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Se-Yeong Oh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Shoeb Lallani
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Andrew Boucher
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| | - Stewart G Neill
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia.,Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Renee D Read
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia. .,Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
67
|
Chen Z, Li S, Mo J, Hawley E, Wang Y, He Y, Brosseau JP, Shipman T, Clapp DW, Carroll TJ, Le LQ. Schwannoma development is mediated by Hippo pathway dysregulation and modified by RAS/MAPK signaling. JCI Insight 2020; 5:141514. [PMID: 32960816 PMCID: PMC7605536 DOI: 10.1172/jci.insight.141514] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/11/2020] [Indexed: 01/19/2023] Open
Abstract
Schwannomas are tumors of the Schwann cells that cause chronic pain, numbness, and potentially life-threatening impairment of vital organs. Despite the identification of causative genes, including NF2 (Merlin), INI1/SMARCB1, and LZTR1, the exact molecular mechanism of schwannoma development is still poorly understood. Several studies have identified Merlin as a key regulator of the Hippo, MAPK, and PI3K signaling pathways; however, definitive evidence demonstrating the importance of these pathways in schwannoma pathogenesis is absent. Here, we provide direct genetic evidence that dysregulation of the Hippo pathway in the Schwann cell lineage causes development of multiple schwannomas in mice. We found that canonical Hippo signaling through the effectors YAP/TAZ is required for schwannomagenesis and that MAPK signaling modifies schwannoma formation. Furthermore, cotargeting YAP/TAZ transcriptional activity and MAPK signaling demonstrated a synergistic therapeutic effect on schwannomas. Our new model provides a tractable platform to dissect the molecular mechanisms underpinning schwannoma formation and the role of combinatorial targeted therapy in schwannoma treatment. Canonical Hippo signaling through the effectors YAP/TAZ is required for the development of peripheral nervous system tumors of Schwann cells, and MAPK signaling modifies schwannoma formation.
Collapse
Affiliation(s)
| | - Stephen Li
- Department of Dermatology and.,Medical Scientist Training Program, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Juan Mo
- Department of Dermatology and
| | - Eric Hawley
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Yongzheng He
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | - D Wade Clapp
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Thomas J Carroll
- Department of Molecular Biology.,Simmons Comprehensive Cancer Center, and
| | - Lu Q Le
- Department of Dermatology and.,Simmons Comprehensive Cancer Center, and.,Comprehensive Neurofibromatosis Clinic, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
68
|
Kronenberg NM, Tilston-Lunel A, Thompson FE, Chen D, Yu W, Dholakia K, Gather MC, Gunn-Moore FJ. Willin/FRMD6 Influences Mechanical Phenotype and Neuronal Differentiation in Mammalian Cells by Regulating ERK1/2 Activity. Front Cell Neurosci 2020; 14:552213. [PMID: 33088261 PMCID: PMC7498650 DOI: 10.3389/fncel.2020.552213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022] Open
Abstract
Willin/FRMD6 is part of a family of proteins with a 4.1 ezrin-radixin-moesin (FERM) domain. It has been identified as an upstream activator of the Hippo pathway and, when aberrant in its expression, is associated with human diseases and disorders. Even though Willin/FRMD6 was originally discovered in the rat sciatic nerve, most studies have focused on its functional roles in cells outside of the nervous system, where Willin/FRMD6 is involved in the formation of apical junctional cell-cell complexes and in regulating cell migration. Here, we investigate the biochemical and biophysical role of Willin/FRMD6 in neuronal cells, employing the commonly used SH-SY5Y neuronal model cell system and combining biochemical measurements with Elastic Resonator Interference Stress Micropscopy (ERISM). We present the first direct evidence that Willin/FRMD6 expression influences both the cell mechanical phenotype and neuronal differentiation. By investigating cells with increased and decreased Willin/FRMD6 expression levels, we show that Willin/FRMD6 not only affects proliferation and migration capacity of cells but also leads to changes in cell morphology and an enhanced formation of neurite-like membrane extensions. These changes were accompanied by alterations of biophysical parameters such as cell force, the organization of actin stress fibers and the formation of focal adhesions. At the biochemical level, changes in Willin/FRMD6 expression inversely affected the activity of the extracellular signal-regulated kinases (ERK) pathway and downstream transcriptional factor NeuroD1, which seems to prime SH-SY5Y cells for retinoic acid (RA)-induced neuronal differentiation.
Collapse
Affiliation(s)
- Nils M Kronenberg
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom.,Centre for Nanobiophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Andrew Tilston-Lunel
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom.,Department of Biochemistry, School of Medicine, Boston University, Boston, MA, United States
| | - Frances E Thompson
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom
| | - Doris Chen
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom
| | - Wanjia Yu
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom
| | - Kishan Dholakia
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom.,Department of Physics, College of Science, Yonsei University, Seoul, South Korea
| | - Malte C Gather
- Centre of Biophotonics and SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, United Kingdom.,Centre for Nanobiophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Frank J Gunn-Moore
- Centre of Biophotonics, School of Biology, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
69
|
Genome-Wide Screen for Context-Dependent Tumor Suppressors Identified Using in Vivo Models for Neoplasia in Drosophila. G3-GENES GENOMES GENETICS 2020; 10:2999-3008. [PMID: 32737065 PMCID: PMC7467006 DOI: 10.1534/g3.120.401545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic approaches in Drosophila have successfully identified many genes involved in regulation of growth control as well as genetic interactions relevant to the initiation and progression of cancer in vivo. Here, we report on large-scale RNAi-based screens to identify potential tumor suppressor genes that interact with known cancer-drivers: the Epidermal Growth Factor Receptor and the Hippo pathway transcriptional cofactor Yorkie. These screens were designed to identify genes whose depletion drove tissue expressing EGFR or Yki from a state of benign overgrowth into neoplastic transformation in vivo. We also report on an independent screen aimed to identify genes whose depletion suppressed formation of neoplastic tumors in an existing EGFR-dependent neoplasia model. Many of the positives identified here are known to be functional in growth control pathways. We also find a number of novel connections to Yki and EGFR driven tissue growth, mostly unique to one of the two. Thus, resources provided here would be useful to all researchers who study negative regulators of growth during development and cancer in the context of activated EGFR and/or Yki and positive regulators of growth in the context of activated EGFR. Resources reported here are available freely for anyone to use.
Collapse
|
70
|
Zhu T, Ma Z, Wang H, Jia X, Wu Y, Fu L, Li Z, Zhang C, Yu G. YAP/TAZ affects the development of pulmonary fibrosis by regulating multiple signaling pathways. Mol Cell Biochem 2020; 475:137-149. [PMID: 32813142 DOI: 10.1007/s11010-020-03866-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
YAP and TAZ are important co-activators of various biological processes in human body. YAP/TAZ plays a vital role in the development of pulmonary fibrosis. Dysregulation of the YAP/TAZ signaling pathway is one of the most important causes of pulmonary fibrosis. Therefore, considering its crucial role, summary of the signal mechanism of YAP/TAZ is of certain guiding significance for the research of YAP/TAZ as a therapeutic target. The present review provided a detailed introduction to various YAP/TAZ-related signaling pathways and clarified the specific role of YAP/TAZ in these pathways. In the meantime, we summarized and evaluated possible applications of YAP/TAZ in the treatment of pulmonary fibrosis. Overall, our study is of guiding significance for future research on the functional mechanism of YAP/TAZ underlying lung diseases as well as for identification of novel therapeutic targets specific to pulmonary fibrosis.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Zhifeng Ma
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Haiyong Wang
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Xiaoxiao Jia
- Department of Pathology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China
| | - Yuanlin Wu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Linhai Fu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Zhupeng Li
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Chu Zhang
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China.
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China.
| |
Collapse
|
71
|
Lv X, He C, Huang C, Hua G, Chen X, Timm BK, Maclin VM, Haggerty AA, Aust SK, Golden DM, Dave BJ, Tseng YA, Chen L, Wang H, Chen P, Klinkebiel DL, Karpf AR, Dong J, Drapkin RI, Rueda BR, Davis JS, Wang C. Reprogramming of Ovarian Granulosa Cells by YAP1 Leads to Development of High-Grade Cancer with Mesenchymal Lineage and Serous Features. Sci Bull (Beijing) 2020; 65:1281-1296. [PMID: 34888112 PMCID: PMC8654108 DOI: 10.1016/j.scib.2020.03.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding the cell-of-origin of ovarian high grade serous cancer (HGSC) is the prerequisite for efficient prevention and early diagnosis of this most lethal gynecological cancer. Recently, a mesenchymal type of ovarian HGSC with the poorest prognosis among ovarian cancers was identified by both TCGA and AOCS studies. The cell-of-origin of this subtype of ovarian cancer is unknown. While pursuing studies to understand the role of the Hippo pathway in ovarian granulosa cell physiology and pathology, we unexpectedly found that the Yes-associated protein 1 (YAP1), the major effector of the Hippo signaling pathway, induced dedifferentiation and reprogramming of the ovarian granulosa cells, a unique type of ovarian follicular cells with mesenchymal lineage and high plasticity, leading to the development of high grade ovarian cancer with serous features. Our research results unveil a potential cell-of-origin for a subtype of HGSC with mesenchymal features.
Collapse
Affiliation(s)
- Xiangmin Lv
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chunbo He
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan 47000, China
| | - Cong Huang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Guohua Hua
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan 47000, China
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Barbara K. Timm
- Heartland Center for Reproductive Medicine, Omaha, NE 68198, USA
| | | | - Abigail A Haggerty
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shelly K Aust
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Denae M Golden
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bhavana J Dave
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yun-An Tseng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Li Chen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongbo Wang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Peichao Chen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - David L Klinkebiel
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adam R Karpf
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ronny I Drapkin
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - John S Davis
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
72
|
Gharanfoli S, Shahverdi A, Dalman A, Ghaznavi P, Alipour H, Eftekhari-Yazdi P. Effect of Maternal Age on Hippo Pathway Related Gene Expressions and Protein Localization Pattern in Human Embryos. CELL JOURNAL 2020; 22:74-80. [PMID: 32779436 PMCID: PMC7481894 DOI: 10.22074/cellj.2020.6860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/11/2019] [Indexed: 02/02/2023]
Abstract
Objective The Hippo pathway plays an important role in embryo development, and separation of trophectoderm
(TE) and inner cell mass (ICM) cell lines. Therefore, this study investigated effect of maternal age on activity of Hippo
pathway in human embryos.
Materials and Methods In this experimental study, the developed up embryos to the blastocyst stage and the
embryos whose growth stopped at the morula stage were collected from women aged 20-30 years old (young group,
94 embryos) and >37 years (old group, 89 embryos). Expression of OCT4, SOX2, CDX2, GATA3, YAP genes and the
relevant proteins, in the both groups were evaluated using respectively quantitative reverse transcription-polymerase
chain reaction (qRT-PCR) and immunofluorescence methods.
Results There was no significant difference in the expression level of OCT4, SOX2, CDX2, GATA3 and YAP genes in
blastocyst and morula stages, between the two groups. However, SOX2 and CDX2 gene expressions in morula stage
embryos of the old group was statistically lower than that of the young group (P=0.007 and P=0.008, respectively).
Additionally, in the embryos collected from women with >37 years of age, at the blastocyst stage, phospho-YAP (p-YAP)
protein was found to be accumulated in the TE, but it was almost disappeared from the ICM. Additionally, in the old
group, contrary to the expectation, YAP protein was expressed in the ICM, rather than TE.
Conclusion The results of this study showed that YAP and P-YAP among the Hippo signalling pathway may be altered
by increasing age.
Collapse
Affiliation(s)
- Sahar Gharanfoli
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.,Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Azam Dalman
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Pooneh Ghaznavi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hiva Alipour
- Biomedicine Group, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. Electronic Address:
| |
Collapse
|
73
|
Lupo B, Sassi F, Pinnelli M, Galimi F, Zanella ER, Vurchio V, Migliardi G, Gagliardi PA, Puliafito A, Manganaro D, Luraghi P, Kragh M, Pedersen MW, Horak ID, Boccaccio C, Medico E, Primo L, Nichol D, Spiteri I, Heide T, Vatsiou A, Graham TA, Élez E, Argiles G, Nuciforo P, Sottoriva A, Dienstmann R, Pasini D, Grassi E, Isella C, Bertotti A, Trusolino L. Colorectal cancer residual disease at maximal response to EGFR blockade displays a druggable Paneth cell-like phenotype. Sci Transl Med 2020; 12:eaax8313. [PMID: 32759276 PMCID: PMC7617202 DOI: 10.1126/scitranslmed.aax8313] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 12/19/2019] [Accepted: 05/22/2020] [Indexed: 12/11/2022]
Abstract
Blockade of epidermal growth factor receptor (EGFR) causes tumor regression in some patients with metastatic colorectal cancer (mCRC). However, residual disease reservoirs typically remain even after maximal response to therapy, leading to relapse. Using patient-derived xenografts (PDXs), we observed that mCRC cells surviving EGFR inhibition exhibited gene expression patterns similar to those of a quiescent subpopulation of normal intestinal secretory precursors with Paneth cell characteristics. Compared with untreated tumors, these pseudodifferentiated tumor remnants had reduced expression of genes encoding EGFR-activating ligands, enhanced activity of human epidermal growth factor receptor 2 (HER2) and HER3, and persistent signaling along the phosphatidylinositol 3-kinase (PI3K) pathway. Clinically, properties of residual disease cells from the PDX models were detected in lingering tumors of responsive patients and in tumors of individuals who had experienced early recurrence. Mechanistically, residual tumor reprogramming after EGFR neutralization was mediated by inactivation of Yes-associated protein (YAP), a master regulator of intestinal epithelium recovery from injury. In preclinical trials, Pan-HER antibodies minimized residual disease, blunted PI3K signaling, and induced long-term tumor control after treatment discontinuation. We found that tolerance to EGFR inhibition is characterized by inactivation of an intrinsic lineage program that drives both regenerative signaling during intestinal repair and EGFR-dependent tumorigenesis. Thus, our results shed light on CRC lineage plasticity as an adaptive escape mechanism from EGFR-targeted therapy and suggest opportunities to preemptively target residual disease.
Collapse
Affiliation(s)
- Barbara Lupo
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Francesco Sassi
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Marika Pinnelli
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Francesco Galimi
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | | | - Valentina Vurchio
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Giorgia Migliardi
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Paolo Armando Gagliardi
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Alberto Puliafito
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Daria Manganaro
- IEO, European Institute of Oncology IRCCS, 20139 Milano, Italy
| | - Paolo Luraghi
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | | | | | | | - Carla Boccaccio
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Enzo Medico
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Luca Primo
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Daniel Nichol
- The Institute of Cancer Research, London SW7 3RP, UK
| | | | - Timon Heide
- The Institute of Cancer Research, London SW7 3RP, UK
| | | | - Trevor A Graham
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Elena Élez
- Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Guillem Argiles
- Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Paolo Nuciforo
- Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | | | | | - Diego Pasini
- IEO, European Institute of Oncology IRCCS, 20139 Milano, Italy
- Department of Health Sciences, University of Milano, 20142 Milano, Italy
| | - Elena Grassi
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Claudio Isella
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Andrea Bertotti
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy.
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino, 10060 Candiolo, Torino, Italy.
- Candiolo Cancer Institute-FPO IRCCS, 10060 Candiolo, Torino, Italy
| |
Collapse
|
74
|
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
|
75
|
Zhang C, Wei S, Sun WP, Teng K, Dai MM, Wang FW, Chen JW, Ling H, Ma XD, Feng ZH, Duan JL, Cai MY, Xie D. Super-enhancer-driven AJUBA is activated by TCF4 and involved in epithelial-mesenchymal transition in the progression of Hepatocellular Carcinoma. Am J Cancer Res 2020; 10:9066-9082. [PMID: 32802179 PMCID: PMC7415796 DOI: 10.7150/thno.45349] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background and Aims: Aberrant transcriptional programs are highly regulated processes that play important roles in the development and progression of hepatocellular carcinoma (HCC). Emerging evidence suggests that super-enhancers (SEs) often drive critical oncogene expression. However, SE-associated genes in HCC pathogenesis are still poorly understood. Methods: We performed integrative ChIP-seq and Hi-C analyses of HCC cells and identified ajuba LIM protein (AJUBA) as a SE-associated gene. We evaluated AJUBA expression in HCC using immunohistochemistry, immunoblotting, and qRT-PCR. ChIP and luciferase reporter assays were performed to demonstrate that transcription factor 4 (TCF4) bound to AJUBA-associated SEs. We then assessed the role of AJUBA in HCC using both in vitro and in vivo assays. Epithelial-mesenchymal transition (EMT) was examined using immunofluorescence and immunoblotting assays. Furthermore, we used immunoprecipitation and BiFC assays to explore the underlying mechanisms. Results: We identified AJUBA as a SE-associated oncogene in HCC regulated by TCF4. High AJUBA expression was related to an aggressive phenotype and unfavorable outcome in HCC patients. AJUBA knockdown significantly reduced cell migration and invasion capacities both in vitro and in vivo. Furthermore, AJUBA overexpression in HCC recruited tumor necrosis factor associated factor 6 (TRAF6), enhancing the phosphorylation of Akt and increasing Akt activity toward GSK-3β, thus promoting EMT. Conclusions: Our results provide functional and mechanistic links between the SE-associated gene AJUBA and tumor EMT in aggressive HCC.
Collapse
|
76
|
Chai TF, Manu KA, Casey PJ, Wang M. Isoprenylcysteine carboxylmethyltransferase is required for the impact of mutant KRAS on TAZ protein level and cancer cell self-renewal. Oncogene 2020; 39:5373-5389. [PMID: 32561852 PMCID: PMC7391290 DOI: 10.1038/s41388-020-1364-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
Cancer stem cells possess the capacity for self-renewal and resistance to chemotherapy. It is therefore crucial to understand the molecular regulators of stemness in the quest to develop effective cancer therapies. TAZ is a transcription activator that promotes stem cell functions in post-development mammalian cells; suppression of TAZ activity reduces or eliminates cancer stemness in select cancers. Isoprenylcysteine carboxylmethyltransferase (ICMT) is the unique enzyme of the last step of posttranslational prenylation processing pathway that modifies several oncogenic proteins, including RAS. We found that suppression of ICMT results in reduced self-renewal/stemness in KRAS-driven pancreatic and breast cancer cells. Silencing of ICMT led to significant reduction of TAZ protein levels and loss of self-renewal ability, which could be reversed by overexpressing mutant KRAS, demonstrating the functional impact of ICMT modification on the ability of KRAS to control TAZ stability and function. Contrary to expectation, YAP protein levels appear to be much less susceptible than TAZ to the regulation by ICMT and KRAS, and YAP is less consequential in regulating stemness characteristics in these cells. Further, we found that the ICMT-dependent KRAS regulation of TAZ was mediated through RAF, but not PI3K, signaling. Functionally, we demonstrate that a signaling cascade from ICMT modification of KRAS to TAZ protein stability supports cancer cell self-renewal abilities in both in vitro and in vivo settings. In addition, studies using the proof-of-concept small molecule inhibitors of ICMT confirmed its role in regulating TAZ and self-renewal, demonstrating the potential utility of targeting ICMT to control aggressive KRAS-driven cancers.
Collapse
Affiliation(s)
- Tin Fan Chai
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore
| | - Kanjoormana Aryan Manu
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Patrick J Casey
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore. .,Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore.
| |
Collapse
|
77
|
Dwivedi N, Tao S, Jamadar A, Sinha S, Howard C, Wallace DP, Fields TA, Leask A, Calvet JP, Rao R. Epithelial Vasopressin Type-2 Receptors Regulate Myofibroblasts by a YAP-CCN2-Dependent Mechanism in Polycystic Kidney Disease. J Am Soc Nephrol 2020; 31:1697-1710. [PMID: 32554753 DOI: 10.1681/asn.2020020190] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Fibrosis is a major cause of loss of renal function in autosomal dominant polycystic kidney disease (ADPKD). In this study, we examined whether vasopressin type-2 receptor (V2R) activity in cystic epithelial cells can stimulate interstitial myofibroblasts and fibrosis in ADPKD kidneys. METHODS We treated Pkd1 gene knockout (Pkd1KO) mice with dDAVP, a V2R agonist, for 3 days and evaluated the effect on myofibroblast deposition of extracellular matrix (ECM). We also analyzed the effects of conditioned media from primary cultures of human ADPKD cystic epithelial cells on myofibroblast activation. Because secretion of the profibrotic connective tissue growth factor (CCN2) increased significantly in dDAVP-treated Pkd1KO mouse kidneys, we examined its role in V2R-dependent fibrosis in ADPKD as well as that of yes-associated protein (YAP). RESULTS V2R stimulation using dDAVP increased the renal interstitial myofibroblast population and ECM deposition. Similarly, conditioned media from human ADPKD cystic epithelial cells increased myofibroblast activation in vitro, suggesting a paracrine mechanism. Renal collecting duct-specific gene deletion of CCN2 significantly reduced cyst growth and myofibroblasts in Pkd1KO mouse kidneys. We found that YAP regulates CCN2, and YAP inhibition or gene deletion reduces renal fibrosis in Pkd1KO mouse kidneys. Importantly, YAP inactivation blocks the dDAVP-induced increase in myofibroblasts in Pkd1KO kidneys. Further in vitro studies showed that V2R regulates YAP by an ERK1/2-dependent mechanism in human ADPKD cystic epithelial cells. CONCLUSIONS Our results demonstrate a novel mechanism by which cystic epithelial cells stimulate myofibroblasts in the pericystic microenvironment, leading to fibrosis in ADPKD. The V2R-YAP-CCN2 cell signaling pathway may present a potential therapeutic target for fibrosis in ADPKD.
Collapse
Affiliation(s)
- Nidhi Dwivedi
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Shixin Tao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sonali Sinha
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Christianna Howard
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Darren P Wallace
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Timothy A Fields
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew Leask
- School of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - James P Calvet
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas .,Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
78
|
DeAngelis MW, McGhie EW, Coolon JD, Johnson RI. Mask, a component of the Hippo pathway, is required for Drosophila eye morphogenesis. Dev Biol 2020; 464:53-70. [PMID: 32464117 DOI: 10.1016/j.ydbio.2020.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022]
Abstract
Hippo signaling is an important regulator of tissue size, but it also has a lesser-known role in tissue morphogenesis. Here we use the Drosophila pupal eye to explore the role of the Hippo effector Yki and its cofactor Mask in morphogenesis. We found that Mask is required for the correct distribution and accumulation of adherens junctions and appropriate organization of the cytoskeleton. Accordingly, disrupting mask expression led to severe mis-patterning and similar defects were observed when yki was reduced or in response to ectopic wts. Further, the patterning defects generated by reducing mask expression were modified by Hippo pathway activity. RNA-sequencing revealed a requirement for Mask for appropriate expression of numerous genes during eye morphogenesis. These included genes implicated in cell adhesion and cytoskeletal organization, a comprehensive set of genes that promote cell survival, and numerous signal transduction genes. To validate our transcriptome analyses, we then considered two loci that were modified by Mask activity: FER and Vinc, which have established roles in regulating adhesion. Modulating the expression of either locus modified mask mis-patterning and adhesion phenotypes. Further, expression of FER and Vinc was modified by Yki. It is well-established that the Hippo pathway is responsive to changes in cell adhesion and the cytoskeleton, but our data indicate that Hippo signaling also regulates these structures.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Emily W McGhie
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Joseph D Coolon
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Ruth I Johnson
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| |
Collapse
|
79
|
Gao J, He L, Zhou L, Jing Y, Wang F, Shi Y, Cai M, Sun J, Xu H, Jiang J, Zhang L, Wang H. Mechanical force regulation of YAP by F-actin and GPCR revealed by super-resolution imaging. NANOSCALE 2020; 12:2703-2714. [PMID: 31950964 DOI: 10.1039/c9nr09452k] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The Hippo signaling pathway plays critical roles in many biological processes including mechanotransduction. The key activator YAP of this pathway is considered as a central component of mechanotransduction signaling sensing the extracellular mechanical microenvironment changes, such as different cell density, the architecture of tissues and matrix stiffness. Although it has been largely studied that YAP is involved in these processes, the underlying mechanism of mechanical force-induced YAP regulation remains unclear. Here we exerted pressure on cell surfaces and investigated how YAP senses the extracellular mechanical force change using one of the super-resolution imaging techniques, dSTORM. We demonstrated that pressure promoted F-actin depolymerization, RhoA down-regulation, and LPAR1 (Gα12/13-coupled receptor) inactivation, which led to YAP cytoplasmic translocation and decreased clustering. Our work uncovers the role of GPCRs and F-actin in pressure-controlled YAP inactivation, and provides new insights into the mechanisms of mechanical regulation of the Hippo signaling pathway.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Lingli He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, P.R. China
| | - Lulu Zhou
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Yingying Jing
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China. and University of Science and Technology of China, Hefei, Anhui 230027, P.R. China
| | - Feng Wang
- Institute of Immunology, The First Bethune Hospital Academy of Translational Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Shi
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Jiayin Sun
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Junguang Jiang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | - Lei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, P.R. China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China. and Laboratory for Marine Biology and biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266237, P.R. China
| |
Collapse
|
80
|
Gou J, Stotsky JA, Othmer HG. Growth control in the Drosophila wing disk. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1478. [PMID: 31917525 DOI: 10.1002/wsbm.1478] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
The regulation of size and shape is a fundamental requirement of biological development and has been a subject of scientific study for centuries, but we still lack an understanding of how organisms know when to stop growing. Imaginal wing disks of the fruit fly Drosophila melanogaster, which are precursors of the adult wings, are an archetypal tissue for studying growth control. The growth of the disks is dependent on many inter- and intra-organ factors such as morphogens, mechanical forces, nutrient levels, and hormones that influence gene expression and cell growth. Extracellular signals are transduced into gene-control signals via complex signal transduction networks, and since cells typically receive many different signals, a mechanism for integrating the signals is needed. Our understanding of the effect of morphogens on tissue-level growth regulation via individual pathways has increased significantly in the last half century, but our understanding of how multiple biochemical and mechanical signals are integrated to determine whether or not a cell decides to divide is still rudimentary. Numerous fundamental questions are involved in understanding the decision-making process, and here we review the major biochemical and mechanical pathways involved in disk development with a view toward providing a basis for beginning to understand how multiple signals can be integrated at the cell level, and how this translates into growth control at the level of the imaginal disk. This article is categorized under: Analytical and Computational Methods > Computational Methods Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Jia Gou
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Jay A Stotsky
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
81
|
Zhang T, Liang Y, Zuo P, Jing S, Li T, Wang Y, Lv C, Li D, Zhang J, Wei Z. 20(S)-Protopanaxadiol blocks cell cycle progression by targeting epidermal growth factor receptor. Food Chem Toxicol 2020; 135:111017. [DOI: 10.1016/j.fct.2019.111017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022]
|
82
|
van Soldt BJ, Cardoso WV. Hippo-Yap/Taz signaling: Complex network interactions and impact in epithelial cell behavior. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e371. [PMID: 31828974 DOI: 10.1002/wdev.371] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/29/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022]
Abstract
The Hippo pathway has emerged as a crucial integrator of signals in biological events from development to adulthood and in diseases. Although extensively studied in Drosophila and in cell cultures, major gaps of knowledge still remain on how this pathway functions in mammalian systems. The pathway consists of a growing number of components, including core kinases and adaptor proteins, which control the subcellular localization of the transcriptional co-activators Yap and Taz through phosphorylation of serines at key sites. When localized to the nucleus, Yap/Taz interact with TEAD transcription factors to induce transcriptional programs of proliferation, stemness, and growth. In the cytoplasm, Yap/Taz interact with multiple pathways to regulate a variety of cellular functions or are targeted for degradation. The Hippo pathway receives cues from diverse intracellular and extracellular inputs, including growth factor and integrin signaling, polarity complexes, and cell-cell junctions. This review highlights the mechanisms of regulation of Yap/Taz nucleocytoplasmic shuttling and their implications for epithelial cell behavior using the lung as an intriguing example of this paradigm. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Signaling Pathways > Cell Fate Signaling Establishment of Spatial and Temporal Patterns > Cytoplasmic Localization.
Collapse
Affiliation(s)
- Benjamin J van Soldt
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care Medicine, Columbia University Irving Medical Center, New York, New York.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
| | - Wellington V Cardoso
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care Medicine, Columbia University Irving Medical Center, New York, New York.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
83
|
Zinatizadeh MR, Miri SR, Zarandi PK, Chalbatani GM, Rapôso C, Mirzaei HR, Akbari ME, Mahmoodzadeh H. The Hippo Tumor Suppressor Pathway (YAP/TAZ/TEAD/MST/LATS) and EGFR-RAS-RAF-MEK in cancer metastasis. Genes Dis 2019; 8:48-60. [PMID: 33569513 PMCID: PMC7859453 DOI: 10.1016/j.gendis.2019.11.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/24/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023] Open
Abstract
Hippo Tumor Suppressor Pathway is the main pathway for cell growth that regulates tissue enlargement and organ size by limiting cell growth. This pathway is activated in response to cell cycle arrest signals (cell polarity, transduction, and DNA damage) and limited by growth factors or mitogens associated with EGF and LPA. The major pathway consists of the central kinase of Ste20 MAPK (Saccharomyces cerevisiae), Hpo (Drosophila melanogaster) or MST kinases (mammalian) that activates the mammalian AGC kinase dmWts or LATS effector (MST and LATS). YAP in the nucleus work as a cofactor for a wide range of transcription factors involved in proliferation (TEA domain family, TEAD1-4), stem cells (Oct4 mononuclear factor and SMAD-related TGFβ effector), differentiation (RUNX1), and Cell cycle/apoptosis control (p53, p63, and p73 family members). This is due to the diverse roles of YAP and may limit tumor progression and establishment. TEAD also coordinates various signal transduction pathways such as Hippo, WNT, TGFβ and EGFR, and effects on lack of regulation of TEAD cancerous genes, such as KRAS, BRAF, LKB1, NF2 and MYC, which play essential roles in tumor progression, metastasis, cancer metabolism, immunity, and drug resistance. However, RAS signaling is a pivotal factor in the inactivation of Hippo, which controls EGFR-RAS-RAF-MEK-ERK-mediated interaction of Hippo signaling. Thus, the loss of the Hippo pathway may have significant consequences on the targets of RAS-RAF mutations in cancer.
Collapse
Affiliation(s)
- Mohammad Reza Zinatizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
- Corresponding author. Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Rouhollah Miri
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Peyman Kheirandish Zarandi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Ghanbar Mahmoodi Chalbatani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Catarina Rapôso
- Faculty of Pharmaceutical Sciences State University of Campinas – UNICAMP Campinas, SP, Brazil
| | - Hamid Reza Mirzaei
- Cancer Research Center, Shohadae Tajrish Hospital, Department of Radiation Oncology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Habibollah Mahmoodzadeh
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
- Corresponding author. Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
84
|
Saab S, Chang OSS, Nagaoka K, Hung MC, Yamaguchi H. The potential role of YAP in Axl-mediated resistance to EGFR tyrosine kinase inhibitors. Am J Cancer Res 2019; 9:2719-2729. [PMID: 31911857 PMCID: PMC6943358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 06/10/2023] Open
Abstract
Yes-associated protein (YAP) is a transcription co-regulator downstream of the Hippo pathway, and plays a critical role in cancer. Although YAP regulation in the canonical Hippo pathway is well established, the Hippo-independent regulation of YAP is not well explored. Here, we showed the possible new mechanism of YAP regulation by the receptor tyrosine kinase Axl. Co-immunoprecipitation and Western blot analysis demonstrated the interaction between YAP and Axl, which was enhanced by Axl ligand Growth Arrest Specific 6 (GAS6) stimulation. Furthermore, we found that YAP is phosphorylated at tyrosine residues by GAS6 stimulation in vivo and Axl directly phosphorylates YAP in vitro. Axl overexpression or GAS6 stimulation increased YAP-mediated transcriptional activity, and YAP-mediated colony forming activity in soft agar was enhanced by co-expression of Axl. In EGFR tyrosine kinase inhibitor (TKI)-sensitive lung cancer cells, YAP protein was downregulated in response to TKI treatment, while overexpression of YAP attenuated TKI sensitivity, suggesting that YAP is a key determinant of TKI response. Moreover Axl overexpression reversed TKI-induced YAP downregulation and induced TKI-resistance, which was reversed by YAP knockdown, further supporting the notion that YAP functions downstream of Axl. Together, these findings suggest a novel role of YAP in Axl-mediated TKI resistance.
Collapse
Affiliation(s)
- Sarah Saab
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar FoundationPO Box 34110, Doha, Qatar
| | - Olin Shih-Shin Chang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterUnit 108, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Present address: Bristol-Myers SquibbRedwood City, CA 94063, USA
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and TechnologyFuchu, Tokyo 183-8509, Japan
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterUnit 108, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, Office of The President, China Medical UniversityTaichung 404, Taiwan
- Department of Biotechnology, Asia UniversityTaichung 413, Taiwan
| | - Hirohito Yamaguchi
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar FoundationPO Box 34110, Doha, Qatar
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer CenterUnit 108, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
85
|
Xu B, Li Q, Chen N, Zhu C, Meng Q, Ayyanathan K, Qian W, Jia H, Wang J, Ni P, Hou Z. The LIM protein Ajuba recruits DBC1 and CBP/p300 to acetylate ERα and enhances ERα target gene expression in breast cancer cells. Nucleic Acids Res 2019; 47:2322-2335. [PMID: 30597111 PMCID: PMC6412004 DOI: 10.1093/nar/gky1306] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 11/13/2022] Open
Abstract
Estrogen/ERα signaling is critical for breast cancer progression and therapeutic treatments. Thus, identifying new regulators of this pathway will help to develop new therapeutics to overcome chemotherapy resistance of the breast cancer cells. Here, we report Ajuba directly interacts with ERα to potentiate ERα target gene expression, and biologically Ajuba promotes breast cancer cell growth and contributes to tamoxifen resistance of these cells. Ajuba constitutively binds the DBD and AF2 regions of ERα, and these interactions can be markedly enhanced by estrogen treatment. Mechanistically, Ajuba recruits DBC1 and CBP/p300 and forms a ternary complex to co-activate ERα transcriptional activity and concomitantly enhances ERα acetylation. Moreover, components of this complex can be found at endogenous promoters containing functional ERα responsive elements. Taken together, these data demonstrate that Ajuba functions as a novel co-activator of ERα and that Ajuba/DBC1/CBP/p300 ternary complex may be a new target for developing therapeutics to treat breast cancer.
Collapse
Affiliation(s)
- Beihui Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Clinical Laboratory, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Li
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ning Chen
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chunxiao Zhu
- Department of Allergy, Linyi Hospital of Traditional Chinese Medicine, Shandong Province, China
| | - Qingrong Meng
- Department of Gynecology, Lanling People's Hospital, Shandong Province, China
| | | | - Wenli Qian
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hao Jia
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiamin Wang
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peihua Ni
- Faculty of Medical Laboratory Science, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoyuan Hou
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Gynecology, Lanling People's Hospital, Shandong Province, China.,Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cellular Biology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
86
|
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
|
87
|
Hsu PC, Jablons DM, Yang CT, You L. Epidermal Growth Factor Receptor (EGFR) Pathway, Yes-Associated Protein (YAP) and the Regulation of Programmed Death-Ligand 1 (PD-L1) in Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2019; 20:ijms20153821. [PMID: 31387256 PMCID: PMC6695603 DOI: 10.3390/ijms20153821] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/02/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) pathway is a well-studied oncogenic pathway in human non-small cell lung cancer (NSCLC). A subset of advanced NSCLC patients (15–55%) have EGFR-driven mutations and benefit from treatment with EGFR-tyrosine kinase inhibitors (TKIs). Immune checkpoint inhibitors (ICIs) targeting the PD-1/PDL-1 axis are a new anti-cancer therapy for metastatic NSCLC. The anti-PD-1/PDL-1 ICIs showed promising efficacy (~30% response rate) and improved the survival of patients with metastatic NSCLC, but the role of anti-PD-1/PDL-1 ICIs for EGFR mutant NSCLC is not clear. YAP (yes-associated protein) is the main mediator of the Hippo pathway and has been identified as promoting cancer progression, drug resistance, and metastasis in NSCLC. Here, we review recent studies that examined the correlation between the EGFR, YAP pathways, and PD-L1 and demonstrate the mechanism by which EGFR and YAP regulate PD-L1 expression in human NSCLC. About 50% of EGFR mutant NSCLC patients acquire resistance to EGFR-TKIs without known targetable secondary mutations. Targeting YAP therapy is suggested as a potential treatment for NSCLC with acquired resistance to EGFR-TKIs. Future work should focus on the efficacy of YAP inhibitors in combination with immune checkpoint PD-L1/PD-1 blockade in EGFR mutant NSCLC without targetable resistant mutations.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - David M Jablons
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Liang You
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA.
| |
Collapse
|
88
|
Wang P, Zhang H, Yang J, Li Z, Wang Y, Leng X, Ganapathy S, Isakson P, Chen C, Zhu T. Mu‐KRAS attenuates Hippo signaling pathway through PKCι to sustain the growth of pancreatic cancer. J Cell Physiol 2019; 235:408-420. [PMID: 31230347 DOI: 10.1002/jcp.28981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Peipei Wang
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| | - Hongmei Zhang
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| | - Jinhe Yang
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| | - Zongxian Li
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| | - Yiren Wang
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| | - Xiaohong Leng
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| | - Suthakar Ganapathy
- The Center of Drug Discovery Northeastern University Boston Massachusetts
| | - Pauline Isakson
- Clinical Immunology & Transfusion Medicine Sahlgrenska University Hospital Gothenburg Sweden
| | - Changyan Chen
- The Center of Drug Discovery Northeastern University Boston Massachusetts
| | - Tongbo Zhu
- Department of Immunology West China School of Basic Medical Sciences & Forensic Medicine Sichuan University Chengdu Sichuan China
| |
Collapse
|
89
|
Mao J, Tian Y, Wang C, Jiang K, Li R, Yao Y, Zhang R, Sun D, Liang R, Gao Z, Wang Q, Wang L. CBX2 Regulates Proliferation and Apoptosis via the Phosphorylation of YAP in Hepatocellular Carcinoma. J Cancer 2019; 10:2706-2719. [PMID: 31258779 PMCID: PMC6584921 DOI: 10.7150/jca.31845] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/09/2019] [Indexed: 12/29/2022] Open
Abstract
Chromobox 2 (CBX2), a chromobox family protein, is a crucial component of the polycomb group complex: polycomb repressive complex 1 (PRC1). Research on CBX2 as an oncogene has been published in recent years. However, the connection between CBX2 and hepatocellular carcinoma (HCC) has not been studied. In this article, based on the results of immunohistochemical (IHC) staining of HCC and adjacent liver tissue microarrays, we found that high CBX2 expression is associated with poor prognosis in HCC patients. The results of a CCK8 assay, a clonogenic survival assay and a nude mouse tumorigenicity assay showed that knockdown of CBX2 inhibited the proliferation of HCC cells. According to the results of Annexin V-FITC/propidium iodide (PI) staining-based fluorescence activated cell sorting (FACS) analysis, knockdown of CBX2 increased HCC cell apoptosis. Furthermore, the RNA-seq results revealed that knockdown of CBX2 inhibited the expression of WTIP, which is an inhibitor of the Hippo pathway. We used western blotting to validate the mechanism and discovered that knockdown of CBX2 increased the phosphorylation of YAP, which explains why knockdown of CBX2 inhibits proliferation and increases apoptosis in HCC cells. In conclusion, CBX2 could be a potential target for HCC anticancer treatment.
Collapse
Affiliation(s)
- Jiakai Mao
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu Tian
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.,Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chengye Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Keqiu Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yifan Yao
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rixin Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Deguang Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Liang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhenming Gao
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qi Wang
- Department of Pulmonary Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
90
|
Single-Cell Analysis of the Liver Epithelium Reveals Dynamic Heterogeneity and an Essential Role for YAP in Homeostasis and Regeneration. Cell Stem Cell 2019; 25:23-38.e8. [PMID: 31080134 DOI: 10.1016/j.stem.2019.04.004] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 02/04/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022]
Abstract
The liver can substantially regenerate after injury, with both main epithelial cell types, hepatocytes and biliary epithelial cells (BECs), playing important roles in parenchymal regeneration. Beyond metabolic functions, BECs exhibit substantial plasticity and in some contexts can drive hepatic repopulation. Here, we performed single-cell RNA sequencing to examine BEC and hepatocyte heterogeneity during homeostasis and after injury. Instead of evidence for a transcriptionally defined progenitor-like BEC cell, we found significant homeostatic BEC heterogeneity that reflects fluctuating activation of a YAP-dependent program. This transcriptional signature defines a dynamic cellular state during homeostasis and is highly responsive to injury. YAP signaling is induced by physiological bile acids (BAs), required for BEC survival in response to BA exposure, and is necessary for hepatocyte reprogramming into biliary progenitors upon injury. Together, these findings uncover molecular heterogeneity within the ductal epithelium and reveal YAP as a protective rheostat and regenerative regulator in the mammalian liver.
Collapse
|
91
|
Zhaojie L, Yuchen L, Miao C, Yacun C, Shayi W, Anbang H, Xinhui L, Meng Z, Peipei W, Hongbing M, Feng W, Zhiming C, Xinyuan G. Gelsolin-like actin-capping protein has prognostic value and promotes tumorigenesis and epithelial-mesenchymal transition via the Hippo signaling pathway in human bladder cancer. Ther Adv Med Oncol 2019; 11:1758835919841235. [PMID: 31068979 PMCID: PMC6492362 DOI: 10.1177/1758835919841235] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Transitional cell carcinoma (TCC) of the bladder, the major histologic subtype of bladder cancer, is increasing in incidence and mortality, which requires the identification of effective biomarkers. Actin-regulating proteins have recently been proposed as important antitumor druggable targets. As a gelsolin-family actin-modulating protein, CAPG (gelsolin-like actin-capping protein) generated great interest due to its crucial effects in various biological and physiological processes; however, the role and mechanism of CAPG in TCCs remain unknown. Materials and methods: Bioinformatic analysis and immunohistochemistry of clinical specimens were performed to detect the expression level of CAPG. Both in vitro and in vivo assays were used to determine the oncogenic effect of CAPG in TCCs. Male 4–5-week-old BALB/c nude mice were used for in vivo tumorigenesis assays, while SCID mice were used for in vivo metastatic assays. Affymetrix microarray was used to identify the underlying molecular mechanism. Western blot and immunofluorescence were used to validate the expression and localization of proteins. Results: CAPG was frequently upregulated in TCCs and associated with clinical aggressiveness and worse prognosis. Functional assays demonstrated that CAPG could contribute to the tumorigenesis, metastasis and epithelial-mesenchymal transition (EMT) of TCCs both in vitro and in vivo. A novel mechanism that CAPG promoted TCC development via inactivating the Hippo pathway, leading to a nucleus translocation of Yes-associated protein was suggested. Conclusions: The current study identified CAPG as a novel and critical oncogene in TCCs, supporting the pursuit of CAPG as a potential target for TCC intervention.
Collapse
Affiliation(s)
- Lyu Zhaojie
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Liu Yuchen
- Key Laboratory of Medical Reprogramming Technology, Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Chen Miao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chen Yacun
- Department of Pathology, The Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wu Shayi
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - He Anbang
- Department of Urology, Peking University First Hospital, The Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Liao Xinhui
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, ChinaDepartment of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhang Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wu Peipei
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mei Hongbing
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Wang Feng
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Cai Zhiming
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 518035 Shenzhen, China
| | - Guan Xinyuan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Room L10-56, 10/F, Laboratory Block 21 Sassoon Road, Hong Kong State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, China
| |
Collapse
|
92
|
Boopathy GTK, Hong W. Role of Hippo Pathway-YAP/TAZ Signaling in Angiogenesis. Front Cell Dev Biol 2019; 7:49. [PMID: 31024911 PMCID: PMC6468149 DOI: 10.3389/fcell.2019.00049] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a highly coordinated process of formation of new blood vessels from pre-existing blood vessels. The process of development of the proper vascular network is a complex process that is crucial for the vertebrate development. Several studies have defined essential roles of Hippo pathway-YAP/TAZ in organ size control, tissue regeneration, and self-renewal. Thus Hippo pathway is one of the central components in tissue homeostasis. There are mounting evidences on the eminence of Hippo pathway-YAP/TAZ in angiogenesis in multiple model organisms. Hippo pathway-YAP/TAZ is now demonstrated to regulate endothelial cell proliferation, migration and survival; subsequently regulating vascular sprouting, vascular barrier formation, and vascular remodeling. Major intracellular signaling programs that regulate angiogenesis concomitantly activate YAP/TAZ to regulate key events in angiogenesis. In this review, we provide a brief overview of the recent findings in the Hippo pathway and YAP/TAZ signaling in angiogenesis.
Collapse
Affiliation(s)
- Gandhi T K Boopathy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
93
|
Gupta S, Varshney B, Chatterjee S, Ray K. Somatic ERK activation during transit amplification is essential for maintaining the synchrony of germline divisions in Drosophila testis. Open Biol 2019; 8:rsob.180033. [PMID: 30045884 PMCID: PMC6070716 DOI: 10.1098/rsob.180033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/28/2018] [Indexed: 12/23/2022] Open
Abstract
Transit amplification (TA) of progenitor cells maintains tissue homeostasis by balancing proliferation and differentiation. In Drosophila testis, the germline proliferation is tightly regulated by factors present in both the germline and the neighbouring somatic cyst cells (SCCs). Although the exact mechanism is unclear, the epidermal growth factor receptor (EGFR) activation in SCCs has been reported to control spermatogonial divisions within a cyst, through downstream activations of Rac1-dependent pathways. Here, we report that somatic activation of the mitogen-activated protein kinase (Rolled/ERK) downstream of EGFR is required to synchronize the mitotic divisions and regulate the transition to meiosis. The process operates independently of the Bag-of-marble activity in the germline. Also, the integrity of the somatic cyst enclosure is inessential for this purpose. Together, these results suggest that synchronization of germ-cell divisions through somatic activation of distinct ERK-downstream targets independently regulates TA and subsequent differentiation of neighbouring germline cells.
Collapse
Affiliation(s)
- Samir Gupta
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Bhavana Varshney
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Shambhabi Chatterjee
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| |
Collapse
|
94
|
Callus BA, Finch-Edmondson ML, Fletcher S, Wilton SD. YAPping about and not forgetting TAZ. FEBS Lett 2019; 593:253-276. [PMID: 30570758 DOI: 10.1002/1873-3468.13318] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/05/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022]
Abstract
The Hippo pathway has emerged as a major eukaryotic signalling pathway and is increasingly the subject of intense interest, as are the key effectors of canonical Hippo signalling, YES-associated protein (YAP) and TAZ. The Hippo pathway has key roles in diverse biological processes, including network signalling regulation, development, organ growth, tissue repair and regeneration, cancer, stem cell regulation and mechanotransduction. YAP and TAZ are multidomain proteins and function as transcriptional coactivators of key genes to evoke their biological effects. YAP and TAZ interact with numerous partners and their activities are controlled by a complex set of processes. This review provides an overview of Hippo signalling and its role in growth. In particular, the functional domains of YAP and TAZ and the complex mechanisms that regulate their protein stability and activity are discussed. Notably, the similarities and key differences are highlighted between the two paralogues including which partner proteins interact with which functional domains to regulate their activity.
Collapse
Affiliation(s)
| | - Megan L Finch-Edmondson
- Discipline of Child and Adolescent Health, Children's Hospital at Westmead Clinical School, University of Sydney Medical School, Australia.,Cerebral Palsy Alliance Research Institute, University of Sydney, Australia
| | - Sue Fletcher
- Centre for Comparative Genomics, Murdoch University, Australia.,Perron Institute for Neurological and Translational Research, Nedlands, Australia
| | - Steve D Wilton
- Centre for Comparative Genomics, Murdoch University, Australia.,Perron Institute for Neurological and Translational Research, Nedlands, Australia
| |
Collapse
|
95
|
Abstract
The Hippo Pathway comprises a vast network of components that integrate diverse signals including mechanical cues and cell surface or cell-surface-associated molecules to define cellular outputs of growth, proliferation, cell fate, and cell survival on both the cellular and tissue level. Because of the importance of the regulators, core components, and targets of this pathway in human health and disease, individual components were often identified by efforts in mammalian models or for a role in a specific process such as stress response or cell death. However, multiple components were originally discovered in the Drosophila system, and the breakthrough of conceiving that these components worked together in a signaling pathway came from a series of Drosophila genetic screens and fundamental genetic and phenotypic characterization efforts. In this chapter, we will review the original discoveries leading to the conceptual framework of these components as a tumor suppressor network. We will review chronologically the early efforts that established our initial understanding of the core machinery that then launched the growing and vibrant field to be discussed throughout later chapters of this book.
Collapse
Affiliation(s)
- Rewatee Gokhale
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cathie M Pfleger
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
96
|
Xekouki P, Lodge EJ, Matschke J, Santambrogio A, Apps JR, Sharif A, Jacques TS, Aylwin S, Prevot V, Li R, Flitsch J, Bornstein SR, Theodoropoulou M, Andoniadou CL. Non-secreting pituitary tumours characterised by enhanced expression of YAP/TAZ. Endocr Relat Cancer 2019; 26:215-225. [PMID: 30139767 PMCID: PMC6215911 DOI: 10.1530/erc-18-0330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022]
Abstract
Tumours of the anterior pituitary can manifest from all endocrine cell types but the mechanisms for determining their specification are not known. The Hippo kinase cascade is a crucial signalling pathway regulating growth and cell fate in numerous organs. There is mounting evidence implicating this in tumour formation, where it is emerging as an anti-cancer target. We previously demonstrated activity of the Hippo kinase cascade in the mouse pituitary and nuclear association of its effectors YAP/TAZ with SOX2-expressing pituitary stem cells. Here, we sought to investigate whether these components are expressed in the human pituitary and if they are deregulated in human pituitary tumours. Analysis of pathway components by immunofluorescence reveals pathway activity during normal human pituitary development and in the adult gland. Poorly differentiated pituitary tumours (null-cell adenomas, adamantinomatous craniopharyngiomas (ACPs) and papillary craniopharyngiomas (PCPs)), displayed enhanced expression of pathway effectors YAP/TAZ. In contrast, differentiated adenomas displayed lower or absent levels. Knockdown of the kinase-encoding Lats1 in GH3 rat mammosomatotropinoma cells suppressed Prl and Gh promoter activity following an increase in YAP/TAZ levels. In conclusion, we have demonstrated activity of the Hippo kinase cascade in the human pituitary and association of high YAP/TAZ with repression of the differentiated state both in vitro and in vivo. Characterisation of this pathway in pituitary tumours is of potential prognostic value, opening up putative avenues for treatments.
Collapse
Affiliation(s)
- Paraskevi Xekouki
- Centre for Craniofacial and Regenerative BiologyKing’s College London, London, UK
- Department of EndocrinologyKing’s College Hospital NHS Foundation Trust, London, UK
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative BiologyKing’s College London, London, UK
- Department of Endocrinology and DiabetesKing’s College London, London, UK
| | - Jakob Matschke
- Institute of NeuropathologyUniversity Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Alice Santambrogio
- Centre for Craniofacial and Regenerative BiologyKing’s College London, London, UK
- Department of Internal Medicine IIICarl Gustav Carus Medical School, Technical University of Dresden, Dresden, Germany
| | - John R Apps
- Birth Defects Research CentreDevelopmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Histopathology DepartmentGreat Ormond Street Hospital NHS Trust, London, UK
| | - Ariane Sharif
- Laboratory of Development and Plasticity of the Neuroendocrine BrainInserm U1172, Jean-Pierre Aubert Research Centre, Lille, France
| | - Thomas S Jacques
- Birth Defects Research CentreDevelopmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Histopathology DepartmentGreat Ormond Street Hospital NHS Trust, London, UK
| | - Simon Aylwin
- Department of EndocrinologyKing’s College Hospital NHS Foundation Trust, London, UK
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine BrainInserm U1172, Jean-Pierre Aubert Research Centre, Lille, France
| | - Ran Li
- Department of NeurosurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jörg Flitsch
- Department of NeurosurgeryHamburg University Medical Center, Hamburg, Germany
| | - Stefan R Bornstein
- Department of Endocrinology and DiabetesKing’s College London, London, UK
- Department of Internal Medicine IIICarl Gustav Carus Medical School, Technical University of Dresden, Dresden, Germany
| | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IVLudwig-Maximilians-Universität München, Munich, Germany
- Correspondence should be addressed to C L Andoniadou or M Theodoropoulou: or
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative BiologyKing’s College London, London, UK
- Department of Internal Medicine IIICarl Gustav Carus Medical School, Technical University of Dresden, Dresden, Germany
- Correspondence should be addressed to C L Andoniadou or M Theodoropoulou: or
| |
Collapse
|
97
|
Bum-Erdene K, Zhou D, Gonzalez-Gutierrez G, Ghozayel MK, Si Y, Xu D, Shannon HE, Bailey BJ, Corson TW, Pollok KE, Wells CD, Meroueh SO. Small-Molecule Covalent Modification of Conserved Cysteine Leads to Allosteric Inhibition of the TEAD⋅Yap Protein-Protein Interaction. Cell Chem Biol 2018; 26:378-389.e13. [PMID: 30581134 DOI: 10.1016/j.chembiol.2018.11.010] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/27/2018] [Accepted: 11/15/2018] [Indexed: 01/12/2023]
Abstract
The Hippo pathway coordinates extracellular signals onto the control of tissue homeostasis and organ size. Hippo signaling primarily regulates the ability of Yap1 to bind and co-activate TEA domain (TEAD) transcription factors. Yap1 tightly binds to TEAD4 via a large flat interface, making the development of small-molecule orthosteric inhibitors highly challenging. Here, we report small-molecule TEAD⋅Yap inhibitors that rapidly and selectively form a covalent bond with a conserved cysteine located within the unique deep hydrophobic palmitate-binding pocket of TEADs. Inhibition of TEAD4 binding to Yap1 by these compounds was irreversible and occurred on a longer time scale. In mammalian cells, the compounds formed a covalent complex with TEAD4, inhibited its binding to Yap1, blocked its transcriptional activity, and suppressed expression of connective tissue growth factor. The compounds inhibited cell viability of patient-derived glioblastoma spheroids, making them suitable as chemical probes to explore Hippo signaling in cancer.
Collapse
Affiliation(s)
- Khuchtumur Bum-Erdene
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Donghui Zhou
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Mona K Ghozayel
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yubing Si
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David Xu
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indianapolis, IN 46202, USA
| | - Harlan E Shannon
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Barbara J Bailey
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Timothy W Corson
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen E Pollok
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Clark D Wells
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
98
|
Zhang Q, Lou L, Cai X, Hao Z, Nie S, Liu Y, Su L, Wu W, Shen H, Li Y. Clinical significance of AJUBA, YAP1, and MMP14 expression in esophageal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:6018-6024. [PMID: 31949690 PMCID: PMC6963081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/25/2018] [Indexed: 06/10/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is generally known to be a highly fatal cancer, and thus novel molecular targets are needed to improve its diagnosis and treatment. AJUBA has been shown to regulate cell cycle, adhesion, proliferation, apoptosis, and migration in many malignant tumors. However, the clinical significance of AJUBA in ESCC tumor metastasis remains unclear. In this study, we explored the role of AJUBA, Yes-associated protein 1 (YAP1), and matrix metalloproteinase 14 (MMP14) in the clinical presentation and survival of ESCC. Immunohistochemical staining showed higher expression of these proteins in cancer tissues than in paired adjacent tissues, and this upregulation was differently related to lymph node metastasis and TNM stage. AJUBA expression was positively correlated with that of YAP1. High expression of MMP14 was associated with reduced survival. In general, our findings reveal that AJUBA, YAP1, and MMP14 might function as oncoproteins and contribute to novel targeted therapy in ESCC.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Lei Lou
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Xiaoli Cai
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Zengfang Hao
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Saisai Nie
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Ying Liu
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Lingrui Su
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Wenxin Wu
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical UniversityShijiazhuang, PR China
| | - Yuehong Li
- Department of Pathology, The Second Hospital of Hebei Medical UniversityShijiazhuang, PR China
| |
Collapse
|
99
|
Abstract
Hippo signaling is an evolutionarily conserved network that has a central role in regulating cell proliferation and cell fate to control organ growth and regeneration. It promotes activation of the LATS kinases, which control gene expression by inhibiting the activity of the transcriptional coactivator proteins YAP and TAZ in mammals and Yorkie in Drosophila. Diverse upstream inputs, including both biochemical cues and biomechanical cues, regulate Hippo signaling and enable it to have a key role as a sensor of cells' physical environment and an integrator of growth control signals. Several components of this pathway localize to cell-cell junctions and contribute to regulation of Hippo signaling by cell polarity, cell contacts, and the cytoskeleton. Downregulation of Hippo signaling promotes uncontrolled cell proliferation, impairs differentiation, and is associated with cancer. We review the current understanding of Hippo signaling and highlight progress in the elucidation of its regulatory mechanisms and biological functions.
Collapse
Affiliation(s)
- Jyoti R Misra
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA;
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA;
| |
Collapse
|
100
|
Hans VR, Wendt TI, Patel AM, Patel MM, Perez L, Talbot DE, Jemc JC. Raw regulates glial population of the eye imaginal disc. Genesis 2018; 56:e23254. [DOI: 10.1002/dvg.23254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/25/2018] [Accepted: 09/29/2018] [Indexed: 01/25/2023]
Affiliation(s)
| | - Taylor I. Wendt
- Department of BiologyLoyola University Chicago Chicago Illinois
| | | | - Mit M. Patel
- Department of BiologyLoyola University Chicago Chicago Illinois
| | - Luselena Perez
- Department of BiologyLoyola University Chicago Chicago Illinois
| | | | | |
Collapse
|