101
|
Zheng Y, Pan D. The Hippo Signaling Pathway in Development and Disease. Dev Cell 2019; 50:264-282. [PMID: 31386861 PMCID: PMC6748048 DOI: 10.1016/j.devcel.2019.06.003] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/23/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway regulates diverse physiological processes, and its dysfunction has been implicated in an increasing number of human diseases, including cancer. Here, we provide an updated review of the Hippo pathway; discuss its roles in development, homeostasis, regeneration, and diseases; and highlight outstanding questions for future investigation and opportunities for Hippo-targeted therapies.
Collapse
Affiliation(s)
- Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.
| |
Collapse
|
102
|
Kück U, Radchenko D, Teichert I. STRIPAK, a highly conserved signaling complex, controls multiple eukaryotic cellular and developmental processes and is linked with human diseases. Biol Chem 2019; 400:1005-1022. [PMID: 31042639 DOI: 10.1515/hsz-2019-0173] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/28/2019] [Indexed: 01/17/2023]
Abstract
The striatin-interacting phosphatases and kinases (STRIPAK) complex is evolutionary highly conserved and has been structurally and functionally described in diverse lower and higher eukaryotes. In recent years, this complex has been biochemically characterized better and further analyses in different model systems have shown that it is also involved in numerous cellular and developmental processes in eukaryotic organisms. Further recent results have shown that the STRIPAK complex functions as a macromolecular assembly communicating through physical interaction with other conserved signaling protein complexes to constitute larger dynamic protein networks. Here, we will provide a comprehensive and up-to-date overview of the architecture, function and regulation of the STRIPAK complex and discuss key issues and future perspectives, linked with human diseases, which may form the basis of further research endeavors in this area. In particular, the investigation of bi-directional interactions between STRIPAK and other signaling pathways should elucidate upstream regulators and downstream targets as fundamental parts of a complex cellular network.
Collapse
Affiliation(s)
- Ulrich Kück
- Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 Bochum, Germany
| | - Daria Radchenko
- Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 Bochum, Germany
| | - Ines Teichert
- Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 Bochum, Germany
| |
Collapse
|
103
|
Rap1 Negatively Regulates the Hippo Pathway to Polarize Directional Protrusions in Collective Cell Migration. Cell Rep 2019; 22:2160-2175. [PMID: 29466741 DOI: 10.1016/j.celrep.2018.01.080] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 12/12/2017] [Accepted: 01/25/2018] [Indexed: 02/08/2023] Open
Abstract
In collective cell migration, directional protrusions orient cells in response to external cues, which requires coordinated polarity among the migrating cohort. However, the molecular mechanism has not been well defined. Drosophila border cells (BCs) migrate collectively and invade via the confined space between nurse cells, offering an in vivo model to examine how group polarity is organized. Here, we show that the front/back polarity of BCs requires Rap1, hyperactivation of which disrupts cluster polarity and induces misoriented protrusions and loss of asymmetry in the actin network. Conversely, hypoactive Rap1 causes fewer protrusions and cluster spinning during migration. A forward genetic screen revealed that downregulation of the Hippo (Hpo) pathway core components hpo or mats enhances the Rap1V12-induced migration defect and misdirected protrusions. Mechanistically, association of Rap1V12 with the kinase domain of Hpo suppresses its activity, which releases Hpo signaling-mediated suppression of F-actin elongation, promoting cellular protrusions in collective cell migration.
Collapse
|
104
|
Snigdha K, Gangwani KS, Lapalikar GV, Singh A, Kango-Singh M. Hippo Signaling in Cancer: Lessons From Drosophila Models. Front Cell Dev Biol 2019; 7:85. [PMID: 31231648 PMCID: PMC6558396 DOI: 10.3389/fcell.2019.00085] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/03/2019] [Indexed: 12/19/2022] Open
Abstract
Hippo pathway was initially identified through genetic screens for genes regulating organ size in fruitflies. Recent studies have highlighted the role of Hippo signaling as a key regulator of homeostasis, and in tumorigenesis. Hippo pathway is comprised of genes that act as tumor suppressor genes like hippo (hpo) and warts (wts), and oncogenes like yorkie (yki). YAP and TAZ are two related mammalian homologs of Drosophila Yki that act as effectors of the Hippo pathway. Hippo signaling deficiency can cause YAP- or TAZ-dependent oncogene addiction for cancer cells. YAP and TAZ are often activated in human malignant cancers. These transcriptional regulators may initiate tumorigenic changes in solid tumors by inducing cancer stem cells and proliferation, culminating in metastasis and chemo-resistance. Given the complex mechanisms (e.g., of the cancer microenvironment, and the extrinsic and intrinsic cues) that overpower YAP/TAZ inhibition, the molecular roles of the Hippo pathway in tumor growth and progression remain poorly defined. Here we review recent findings from studies in whole animal model organism like Drosophila on the role of Hippo signaling regarding its connection to inflammation, tumor microenvironment, and other oncogenic signaling in cancer growth and progression.
Collapse
Affiliation(s)
- Kirti Snigdha
- Department of Biology, University of Dayton, Dayton, OH, United States
| | | | - Gauri Vijay Lapalikar
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, United States.,Pre-Medical Programs, University of Dayton, Dayton, OH, United States.,Center for Tissue Regeneration and Engineering at Dayton, University of Dayton, Dayton, OH, United States.,Integrated Science and Engineering Center, University of Dayton, Dayton, OH, United States
| | - Madhuri Kango-Singh
- Department of Biology, University of Dayton, Dayton, OH, United States.,Pre-Medical Programs, University of Dayton, Dayton, OH, United States.,Center for Tissue Regeneration and Engineering at Dayton, University of Dayton, Dayton, OH, United States.,Integrated Science and Engineering Center, University of Dayton, Dayton, OH, United States
| |
Collapse
|
105
|
Oceandy D, Amanda B, Ashari FY, Faizah Z, Azis MA, Stafford N. The Cross-Talk Between the TNF-α and RASSF-Hippo Signalling Pathways. Int J Mol Sci 2019; 20:ijms20092346. [PMID: 31083564 PMCID: PMC6539482 DOI: 10.3390/ijms20092346] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022] Open
Abstract
The regulation of cell death through apoptosis is essential to a number of physiological processes. Defective apoptosis regulation is associated with many abnormalities including anomalies in organ development, altered immune response and the development of cancer. Several signalling pathways are known to regulate apoptosis including the Tumour Necrosis Factor-α (TNF-α) and Hippo signalling pathways. In this paper we review the cross-talk between the TNF-α pathway and the Hippo signalling pathway. Several molecules that tightly regulate the Hippo pathway, such as members of the Ras-association domain family member (RASSF) family proteins, interact and modulate some key proteins within the TNF-α pathway. Meanwhile, TNF-α stimulation also affects the expression and activation of core components of the Hippo pathway. This implies the crucial role of signal integration between these two major pathways in regulating apoptosis.
Collapse
Affiliation(s)
- Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK.
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Bella Amanda
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Faisal Yusuf Ashari
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Zakiyatul Faizah
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - M Aminudin Azis
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Nicholas Stafford
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
106
|
Politano SF, Salemme RR, Ashley J, López-Rivera JA, Bakula TA, Puhalla KA, Quinn JP, Juszczak MJ, Phillip LK, Carrillo RA, Vanderzalm PJ. Tao Negatively Regulates BMP Signaling During Neuromuscular Junction Development in Drosophila. Dev Neurobiol 2019; 79:335-349. [PMID: 31002474 DOI: 10.1002/dneu.22681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 03/15/2019] [Accepted: 04/15/2019] [Indexed: 12/15/2022]
Abstract
The coordinated growth and development of synapses is critical for all aspects of neural circuit function and mutations that disrupt these processes can result in various neurological defects. Several anterograde and retrograde signaling pathways, including the canonical Bone Morphogenic Protein (BMP) pathway, regulate synaptic development in vertebrates and invertebrates. At the Drosophila larval neuromuscular junction (NMJ), the retrograde BMP pathway is a part of the machinery that controls NMJ expansion concurrent with larval growth. We sought to determine whether the conserved Hippo pathway, critical for proportional growth in other tissues, also functions in NMJ development. We found that neuronal loss of the serine-threonine protein kinase Tao, a regulator of the Hippo signaling pathway, results in supernumerary boutons which contain a normal density of active zones. Tao is also required for proper synaptic function, as reduction of Tao results in NMJs with decreased evoked excitatory junctional potentials. Surprisingly, Tao function in NMJ growth is independent of the Hippo pathway. Instead, our experiments suggest that Tao negatively regulates BMP signaling as reduction of Tao leads to an increase in pMad levels in motor neuron nuclei and an increase in BMP target gene expression. Taken together, these results support a role for Tao as a novel inhibitor of BMP signaling in motor neurons during synaptic development and function.
Collapse
Affiliation(s)
- Stephen F Politano
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - Ryan R Salemme
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - James Ashley
- Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, 60637
| | | | - Toren A Bakula
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - Kathryn A Puhalla
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - John P Quinn
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - Madison J Juszczak
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - Lauren K Phillip
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| | - Robert A Carrillo
- Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, 60637
| | - Pamela J Vanderzalm
- Department of Biology, John Carroll University, University Heights, Ohio, 44118
| |
Collapse
|
107
|
Makwana V, Ryan P, Patel B, Dukie SA, Rudrawar S. Essential role of O-GlcNAcylation in stabilization of oncogenic factors. Biochim Biophys Acta Gen Subj 2019; 1863:1302-1317. [PMID: 31034911 DOI: 10.1016/j.bbagen.2019.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
A reversible post-translational protein modification which involves addition of N-acetylglucosamine (GlcNAc) onto hydroxyl groups of serine and/or threonine residues which is known as O-GlcNAcylation, has emerged as a potent competitor of phosphorylation. This glycosyltransfer reaction is catalyzed by the enzyme O-linked β-N-acetylglucosamine transferase (OGT). This enzyme uses uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), the end product of hexosamine biosynthetic pathway, to modify numerous nuclear and cytosolic proteins. O-GlcNAcylation influences cancer cell metabolism in such a way that hyper-O-GlcNAcylation is considered as a prominent trait of many cancers, and is proposed as a major factor enabling cancer cell proliferation and progression. Growing evidence supports a connection between O-GlcNAcylation and major oncogenic factors, including for example, c-MYC, HIF-1α, and NF-κB. A comprehensive study of the roles of O-GlcNAc modification of oncogenic factors is warranted as a thorough understanding may help drive advances in cancer diagnosis and therapy. The focus of this article is to highlight the interplay between oncogenic factors and O-GlcNAcylation along with OGT in cancer cell proliferation and survival. The prospects for OGT inhibitors will also be discussed.
Collapse
Affiliation(s)
- Vivek Makwana
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Philip Ryan
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Bhautikkumar Patel
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Shailendra-Anoopkumar Dukie
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia.
| | - Santosh Rudrawar
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia.
| |
Collapse
|
108
|
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: 249] [Impact Index Per Article: 41.5] [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
|
109
|
Lim S, Hermance N, Mudianto T, Mustaly HM, Mauricio IPM, Vittoria MA, Quinton RJ, Howell BW, Cornils H, Manning AL, Ganem NJ. Identification of the kinase STK25 as an upstream activator of LATS signaling. Nat Commun 2019; 10:1547. [PMID: 30948712 PMCID: PMC6449379 DOI: 10.1038/s41467-019-09597-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 03/20/2019] [Indexed: 02/03/2023] Open
Abstract
The Hippo pathway maintains tissue homeostasis by negatively regulating the oncogenic transcriptional co-activators YAP and TAZ. Though functional inactivation of the Hippo pathway is common in tumors, mutations in core pathway components are rare. Thus, understanding how tumor cells inactivate Hippo signaling remains a key unresolved question. Here, we identify the kinase STK25 as an activator of Hippo signaling. We demonstrate that loss of STK25 promotes YAP/TAZ activation and enhanced cellular proliferation, even under normally growth-suppressive conditions both in vitro and in vivo. Notably, STK25 activates LATS by promoting LATS activation loop phosphorylation independent of a preceding phosphorylation event at the hydrophobic motif, which represents a form of Hippo activation distinct from other kinase activators of LATS. STK25 is significantly focally deleted across a wide spectrum of human cancers, suggesting STK25 loss may represent a common mechanism by which tumor cells functionally impair the Hippo tumor suppressor pathway.
Collapse
Affiliation(s)
- Sanghee Lim
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Nicole Hermance
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | - Tenny Mudianto
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Hatim M Mustaly
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ian Paolo Morelos Mauricio
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Marc A Vittoria
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Ryan J Quinton
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Brian W Howell
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | | | - Amity L Manning
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, 01605, USA
| | - Neil J Ganem
- The Laboratory of Cancer Cell Biology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA.
- Division of Hematology and Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
110
|
Turunen SP, von Nandelstadh P, Öhman T, Gucciardo E, Seashore-Ludlow B, Martins B, Rantanen V, Li H, Höpfner K, Östling P, Varjosalo M, Lehti K. FGFR4 phosphorylates MST1 to confer breast cancer cells resistance to MST1/2-dependent apoptosis. Cell Death Differ 2019; 26:2577-2593. [PMID: 30903103 PMCID: PMC7224384 DOI: 10.1038/s41418-019-0321-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/18/2019] [Accepted: 03/07/2019] [Indexed: 01/01/2023] Open
Abstract
Cancer cells balance with the equilibrium of cell death and growth to expand and metastasize. The activity of mammalian sterile20-like kinases (MST1/2) has been linked to apoptosis and tumor suppression via YAP/Hippo pathway-independent and -dependent mechanisms. Using a kinase substrate screen, we identified here MST1 and MST2 among the top substrates for fibroblast growth factor receptor 4 (FGFR4). In COS-1 cells, MST1 was phosphorylated at Y433 residue in an FGFR4 kinase activity-dependent manner, as assessed by mass spectrometry. Blockade of this phosphorylation by Y433F mutation induced MST1 activation, as indicated by increased threonine phosphorylation of MST1/2, and the downstream substrate MOB1, in FGFR4-overexpressing T47D and MDA-MB-231 breast cancer cells. Importantly, the specific knockdown or short-term inhibition of FGFR4 in endogenous models of human HER2+ breast cancer cells likewise led to increased MST1/2 activation, in conjunction with enhanced MST1 nuclear localization and generation of N-terminal cleaved and autophosphorylated MST1. Unexpectedly, MST2 was also essential for this MST1/N activation and coincident apoptosis induction, although these two kinases, as well as YAP, were differentially regulated in the breast cancer models analyzed. Moreover, pharmacological FGFR4 inhibition specifically sensitized the HER2+ MDA-MB-453 breast cancer cells, not only to HER2/EGFR and AKT/mTOR inhibitors, but also to clinically relevant apoptosis modulators. In TCGA cohort, FGFR4 overexpression correlated with abysmal HER2+ breast carcinoma patient outcome. Therefore, our results uncover a clinically relevant, targetable mechanism of FGFR4 oncogenic activity via suppression of the stress-associated MST1/2-induced apoptosis machinery in tumor cells with prominent HER/ERBB and FGFR4 signaling-driven proliferation.
Collapse
Affiliation(s)
- S Pauliina Turunen
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Pernilla von Nandelstadh
- Research Programs Unit, Genome-Scale Biology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Tiina Öhman
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, FI-00014, Finland
| | - Erika Gucciardo
- Research Programs Unit, Genome-Scale Biology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Brinton Seashore-Ludlow
- Department of Oncology and Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Beatriz Martins
- Research Programs Unit, Genome-Scale Biology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Ville Rantanen
- Research Programs Unit, Genome-Scale Biology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Huini Li
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Katrin Höpfner
- Research Programs Unit, Genome-Scale Biology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland
| | - Päivi Östling
- Department of Oncology and Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, FI-00014, Finland
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, SE-171 77, Sweden. .,Research Programs Unit, Genome-Scale Biology, Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, FI-00014, Finland.
| |
Collapse
|
111
|
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
|
112
|
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
|
113
|
Matsui Y, Zhang Y, Paulson RF, Lai ZC. Dual Role of a C-Terminally Truncated Isoform of Large Tumor Suppressor Kinase 1 in the Regulation of Hippo Signaling and Tissue Growth. DNA Cell Biol 2018; 38:91-106. [PMID: 30461308 DOI: 10.1089/dna.2018.4340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The considerable amount of experimental evidence has defined the Hippo pathway as a tumor suppressive pathway and increased expression and/or activity of its oncogenic effectors is frequently observed in cancer. However, clinical studies have failed to attribute cancer development and progression to mutations in the pathway. In explaining this conundrum, we investigated the expression and functions of a C-terminally truncated isoform of large tumor suppressor kinase 1 (LATS1) called short LATS1 (sLATS1) in human cell lines and Drosophila. Intriguingly, through overexpression of sLATS1, we demonstrated that sLATS1 either activates or suppresses the activity of Yes-associated protein (YAP), one of the effectors of the Hippo pathway, in a cell type-specific manner. The activation is mediated through inhibition of full-length LATS1, whereas suppression of YAP is accomplished through sLATS1-YAP interaction. In HEK293T cells, the former mechanism may affect the cellular response more dominantly, whereas in U2OS cells and developing tissues in Drosophila, the latter mechanism may be solely carried out. Finally, to find the clinical relevance of this molecule, we examined the expression of sLATS1 in breast cancer patients. The transcriptome analysis showed that the ratio of sLATS1 to LATS1 was increased in tumor tissues comparing to their adjacent normal tissues.
Collapse
Affiliation(s)
- Yurika Matsui
- 1 Intercollege Graduate Degree Program in Molecular, Cellular, and Integrative Biosciences, The Pennsylvania State University, University Park , Pennsylvania
| | - Yifan Zhang
- 2 Department of Biology, The Pennsylvania State University , University Park, Pennsylvania
| | - Robert F Paulson
- 3 Department of Veterinary and Biomedical Sciences, and The Pennsylvania State University , University Park, Pennsylvania
| | - Zhi-Chun Lai
- 1 Intercollege Graduate Degree Program in Molecular, Cellular, and Integrative Biosciences, The Pennsylvania State University, University Park , Pennsylvania.,2 Department of Biology, The Pennsylvania State University , University Park, Pennsylvania.,4 Department of Biochemistry and Molecular Biology, The Pennsylvania State University , University Park, Pennsylvania
| |
Collapse
|
114
|
Poon CLC, Liu W, Song Y, Gomez M, Kulaberoglu Y, Zhang X, Xu W, Veraksa A, Hergovich A, Ghabrial A, Harvey KF. A Hippo-like Signaling Pathway Controls Tracheal Morphogenesis in Drosophila melanogaster. Dev Cell 2018; 47:564-575.e5. [PMID: 30458981 DOI: 10.1016/j.devcel.2018.09.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 08/26/2018] [Accepted: 09/28/2018] [Indexed: 11/29/2022]
Abstract
Hippo-like pathways are ancient signaling modules first identified in yeasts. The best-defined metazoan module forms the core of the Hippo pathway, which regulates organ size and cell fate. Hippo-like kinase modules consist of a Sterile 20-like kinase, an NDR kinase, and non-catalytic protein scaffolds. In the Hippo pathway, the upstream kinase Hippo can be activated by another kinase, Tao-1. Here, we delineate a related Hippo-like signaling module that Tao-1 regulates to control tracheal morphogenesis in Drosophila melanogaster. Tao-1 activates the Sterile 20-like kinase GckIII by phosphorylating its activation loop, a mode of regulation that is conserved in humans. Tao-1 and GckIII act upstream of the NDR kinase Tricornered to ensure proper tube formation in trachea. Our study reveals that Tao-1 activates two related kinase modules to control both growth and morphogenesis. The Hippo-like signaling pathway we have delineated has a potential role in the human vascular disease cerebral cavernous malformation.
Collapse
Affiliation(s)
- Carole L C Poon
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Weijie Liu
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Yanjun Song
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Marta Gomez
- University College London, Cancer Institute, London WC1E 6BT, UK
| | | | - Xiaomeng Zhang
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Wenjian Xu
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Alexey Veraksa
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | | | - Amin Ghabrial
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology and Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
115
|
Abstract
Organ growth is fundamental to animal development. One of major mechanisms for growth control is mediated by the conserved Hippo signaling pathway initially identified in Drosophila. The core of this pathway in Drosophila consists of a cascade of protein kinases Hippo and Warts that negatively regulate transcriptional coactivator Yorkie (Yki). Activation of Yki promotes cell survival and proliferation to induce organ growth. A key issue in Hippo signaling is to understand how core kinase cascade is activated. Activation of Hippo kinase cascade is regulated in the upstream by at least two transmembrane proteins Crumbs and Fat that act in parallel. These membrane proteins interact with additional factors such as FERM-domain proteins Expanded and Merlin to modulate subcellular localization and function of the Hippo kinase cascade. Hippo signaling is also influenced by cytoskeletal networks and cell tension in epithelia of developing organs. These upstream events in the regulation of Hippo signaling are only partially understood. This review focuses on our current understanding of some upstream processes involved in Hippo signaling in developing Drosophila organs.
Collapse
Affiliation(s)
- Kwang-Wook Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
116
|
Abstract
How the organ size is adjusted to the proper size during development and how organs know that they reach the original size during regeneration remain long-standing questions. Based on studies using multiple model organisms and approaches for over 20 years, a consensus has been established that the Hippo pathway plays crucial roles in controlling organ size and maintaining tissue homeostasis. Given the significance of these processes, the dysregulation of the Hippo pathway has also implicated various diseases, such as tissue degeneration and cancer. By regulating the downstream transcriptional coactivators YAP and TAZ, the Hippo pathway coordinates cell proliferation and apoptosis in response to a variety of signals including cell contact inhibition, polarity, mechanical sensation and soluble factors. Since the core components and their functions of the Hippo pathway are evolutionarily conserved, this pathway serves as a global regulator of organ size control. Therefore, further investigation of the regulatory mechanisms will provide physiological insights to better understand tissue homeostasis. In this review, the historical developments and current understandings of the regulatory mechanism of Hippo signaling pathway are discussed.
Collapse
Affiliation(s)
- Wantae Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Eek-Hoon Jho
- Departement of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
117
|
Kim Y, Jho EH. Regulation of the Hippo signaling pathway by ubiquitin modification. BMB Rep 2018; 51:143-150. [PMID: 29366444 PMCID: PMC5882221 DOI: 10.5483/bmbrep.2018.51.3.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Indexed: 12/27/2022] Open
Abstract
The Hippo signaling pathway plays an essential role in adult tissue homeostasis and organ size control. Abnormal regulation of Hippo signaling can be a cause for multiple types of human cancers. Since the awareness of the importance of the Hippo signaling in a wide range of biological fields has been continually grown, it is also understood that a thorough and well-rounded comprehension of the precise dynamics could provide fundamental insights for therapeutic applications. Several components in the Hippo signaling pathway are known to be targeted for proteasomal degradation via ubiquitination by E3 ligases. β-TrCP is a well-known E3 ligase of YAP/TAZ, which leads to the reduction of YAP/TAZ levels. The Hippo signaling pathway can also be inhibited by the E3 ligases (such as ITCH) which target LATS1/2 for degradation. Regulation via ubiquitination involves not only complex network of E3 ligases but also deubiquitinating enzymes (DUBs), which remove ubiquitin from its targets. Interestingly, non-degradative ubiquitin modifications are also known to play important roles in the regulation of Hippo signaling. Although there has been much advanced progress in the investigation of ubiquitin modifications acting as regulators of the Hippo signaling pathway, research done to date still remains inadequate due to the sheer complexity and diversity of the subject. Herein, we review and discuss recent developments that implicate ubiquitin-mediated regulatory mechanisms at multiple steps of the Hippo signaling pathway. [BMB Reports 2018; 51(3): 143-150].
Collapse
Affiliation(s)
- Youngeun Kim
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
118
|
Activation mechanisms of the Hippo kinase signaling cascade. Biosci Rep 2018; 38:BSR20171469. [PMID: 30038061 PMCID: PMC6131212 DOI: 10.1042/bsr20171469] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 11/21/2022] Open
Abstract
First discovered two decades ago through genetic screens in Drosophila, the Hippo pathway has been shown to be conserved in metazoans and controls organ size and tissue homeostasis through regulating the balance between cell proliferation and apoptosis. Dysregulation of the Hippo pathway leads to aberrant tissue growth and tumorigenesis. Extensive studies in Drosophila and mammals have identified the core components of Hippo signaling, which form a central kinase cascade to ultimately control gene expression. Here, we review recent structural, biochemical, and cellular studies that have revealed intricate phosphorylation-dependent mechanisms in regulating the formation and activation of the core kinase complex in the Hippo pathway. These studies have established the dimerization-mediated activation of the Hippo kinase (mammalian Ste20-like 1 and 2 (MST1/2) in mammals), the dynamic scaffolding and allosteric roles of adaptor proteins in downstream kinase activation, and the importance of multisite linker autophosphorylation by Hippo and MST1/2 in fine-tuning the signaling strength and robustness of the Hippo pathway. We highlight the gaps in our knowledge in this field that will require further mechanistic studies.
Collapse
|
119
|
Mechanoregulation and pathology of YAP/TAZ via Hippo and non-Hippo mechanisms. Clin Transl Med 2018; 7:23. [PMID: 30101371 PMCID: PMC6087706 DOI: 10.1186/s40169-018-0202-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023] Open
Abstract
Yes-associated protein (YAP) and its paralog WW domain containing transcription regulator 1 (TAZ) are important regulators of multiple cellular functions such as proliferation, differentiation, and survival. On the tissue level, YAP/TAZ are essential for embryonic development, organ size control and regeneration, while their deregulation leads to carcinogenesis or other diseases. As an underlying principle for YAP/TAZ-mediated regulation of biological functions, a growing body of research reveals that YAP/TAZ play a central role in delivering information of mechanical environments surrounding cells to the nucleus transcriptional machinery. In this review, we discuss mechanical cue-dependent regulatory mechanisms for YAP/TAZ functions, as well as their clinical significance in cancer progression and treatment.
Collapse
|
120
|
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are two homologous transcriptional coactivators that promote cell proliferation, stem cell maintenance, and tissue homeostasis. Under favorable conditions, YAP and TAZ are active to promote cell growth through a transcriptional program mediated by the TEAD family transcription factors. Given the indispensability of cellular energy and metabolites for survival and growth, YAP and TAZ are inhibited when energy level is low. Indeed, glucose, fatty acids, hormones, and other metabolic factors have been recently revealed to regulate YAP and TAZ. Conversely, YAP and TAZ are also involved in metabolism regulation, such as to promote glycolysis, lipogenesis, and glutaminolysis, suggesting YAP and TAZ as emerging nodes in coordinating nutrient availability with cell growth and tissue homeostasis. In this Review, we summarize recent findings and provide a current overview of YAP and TAZ in metabolism by focusing on the role of YAP and TAZ as integrators for metabolic cues and cell growth.
Collapse
Affiliation(s)
- Ja Hyun Koo
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
121
|
Homeostatic Control of Hpo/MST Kinase Activity through Autophosphorylation-Dependent Recruitment of the STRIPAK PP2A Phosphatase Complex. Cell Rep 2018; 21:3612-3623. [PMID: 29262338 DOI: 10.1016/j.celrep.2017.11.076] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/25/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
The Hippo pathway controls organ size and tissue homeostasis through a kinase cascade leading from the Ste20-like kinase Hpo (MST1/2 in mammals) to the transcriptional coactivator Yki (YAP/TAZ in mammals). Whereas previous studies have uncovered positive and negative regulators of Hpo/MST, how they are integrated to maintain signaling homeostasis remains poorly understood. Here, we identify a self-restricting mechanism whereby autophosphorylation of an unstructured linker in Hpo/MST creates docking sites for the STRIPAK PP2A phosphatase complex to inactivate Hpo/MST. Mutation of the phospho-dependent docking sites in Hpo/MST or deletion of Slmap, the STRIPAK subunit recognizing these docking sites, results in constitutive activation of Hpo/MST in both Drosophila and mammalian cells. In contrast, autophosphorylation of the Hpo/MST linker at distinct sites is known to recruit Mats/MOB1 to facilitate Hippo signaling. Thus, multisite autophosphorylation of Hpo/MST linker provides an evolutionarily conserved built-in molecular platform to maintain signaling homeostasis by coupling antagonistic signaling activities.
Collapse
|
122
|
Assembly and activation of the Hippo signalome by FAT1 tumor suppressor. Nat Commun 2018; 9:2372. [PMID: 29985391 PMCID: PMC6037762 DOI: 10.1038/s41467-018-04590-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/04/2018] [Indexed: 12/13/2022] Open
Abstract
Dysregulation of the Hippo signaling pathway and the consequent YAP1 activation is a frequent event in human malignancies, yet the underlying molecular mechanisms are still poorly understood. A pancancer analysis of core Hippo kinases and their candidate regulating molecules revealed few alterations in the canonical Hippo pathway, but very frequent genetic alterations in the FAT family of atypical cadherins. By focusing on head and neck squamous cell carcinoma (HNSCC), which displays frequent FAT1 alterations (29.8%), we provide evidence that FAT1 functional loss results in YAP1 activation. Mechanistically, we found that FAT1 assembles a multimeric Hippo signaling complex (signalome), resulting in activation of core Hippo kinases by TAOKs and consequent YAP1 inactivation. We also show that unrestrained YAP1 acts as an oncogenic driver in HNSCC, and that targeting YAP1 may represent an attractive precision therapeutic option for cancers harboring genomic alterations in the FAT1 tumor suppressor genes. Dysregulation of the Hippo signaling is a frequent event in human malignancies, but the molecular mechanisms remain unclear. Here the authors show that in head and neck squamous carcinoma, FAT1 interacts with the Hippo signaling complex, resulting in the activation of core Hippo kinases and YAP1 inactivation.
Collapse
|
123
|
Moon S, Yeon Park S, Woo Park H. Regulation of the Hippo pathway in cancer biology. Cell Mol Life Sci 2018; 75:2303-2319. [PMID: 29602952 PMCID: PMC11105795 DOI: 10.1007/s00018-018-2804-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/17/2018] [Accepted: 03/22/2018] [Indexed: 01/23/2023]
Abstract
The Hippo tumor suppressor pathway, which is well conserved from Drosophila to humans, has emerged as the master regulator of organ size, as well as major cellular properties, such as cell proliferation, survival, stemness, and tissue homeostasis. The biological significance and deregulation of the Hippo pathway in tumorigenesis have received a surge of interest in the past decade. In the current review, we present the major discoveries that made substantial contributions to our understanding of the Hippo pathway and discuss how Hippo pathway components contribute to cellular signaling, physiology, and their potential implications in anticancer therapeutics.
Collapse
Affiliation(s)
- Sungho Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - So Yeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
124
|
|
125
|
Li Q, Nirala NK, Nie Y, Chen HJ, Ostroff G, Mao J, Wang Q, Xu L, Ip YT. Ingestion of Food Particles Regulates the Mechanosensing Misshapen-Yorkie Pathway in Drosophila Intestinal Growth. Dev Cell 2018; 45:433-449.e6. [PMID: 29754801 PMCID: PMC7480018 DOI: 10.1016/j.devcel.2018.04.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 03/04/2018] [Accepted: 04/11/2018] [Indexed: 12/12/2022]
Abstract
The intestinal epithelium has a high cell turnover rate and is an excellent system to study stem cell-mediated adaptive growth. In the Drosophila midgut, the Ste20 kinase Misshapen, which is distally related to Hippo, has a niche function to restrict intestinal stem cell activity. We show here that, under low growth conditions, Misshapen is localized near the cytoplasmic membrane, is phosphorylated at the threonine 194 by the upstream kinase Tao, and is more active toward Warts, which in turn inhibits Yorkie. Ingestion of yeast particles causes a midgut distention and a reduction of Misshapen membrane association and activity. Moreover, Misshapen phosphorylation is regulated by the stiffness of cell culture substrate, changing of actin cytoskeleton, and ingestion of inert particles. These results together suggest that dynamic membrane association and Tao phosphorylation of Misshapen are steps that link the mechanosensing of intestinal stretching after food particle ingestion to control adaptive growth.
Collapse
Affiliation(s)
- Qi Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Niraj K Nirala
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yingchao Nie
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hsi-Ju Chen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Gary Ostroff
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Qi Wang
- Neuroscience Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Lan Xu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Y Tony Ip
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
126
|
Nguyen TH, Kugler JM. Ubiquitin-Dependent Regulation of the Mammalian Hippo Pathway: Therapeutic Implications for Cancer. Cancers (Basel) 2018; 10:cancers10040121. [PMID: 29673168 PMCID: PMC5923376 DOI: 10.3390/cancers10040121] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/08/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway serves as a key barrier for oncogenic transformation. It acts by limiting the activity of the proto-oncogenes YAP and TAZ. Reduced Hippo signaling and elevated YAP/TAZ activities are frequently observed in various types of tumors. Emerging evidence suggests that the ubiquitin system plays an important role in regulating Hippo pathway activity. Deregulation of ubiquitin ligases and of deubiquitinating enzymes has been implicated in increased YAP/TAZ activity in cancer. In this article, we review recent insights into the ubiquitin-mediated regulation of the mammalian Hippo pathway, its deregulation in cancer, and possibilities for targeting the Hippo pathway through the ubiquitin system.
Collapse
Affiliation(s)
- Thanh Hung Nguyen
- Institute of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| | - Jan-Michael Kugler
- Institute of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| |
Collapse
|
127
|
Fulford A, Tapon N, Ribeiro PS. Upstairs, downstairs: spatial regulation of Hippo signalling. Curr Opin Cell Biol 2018; 51:22-32. [PMID: 29154163 DOI: 10.1016/j.ceb.2017.10.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/13/2017] [Indexed: 12/31/2022]
Abstract
Cellular signalling lies at the heart of every decision involved in the development and homeostasis of multicellular organisms. The Hippo pathway was discovered nearly two decades ago through seminal work in Drosophila and rapidly emerged as a crucial signalling network implicated in developmental and oncogenic growth, tissue regeneration and stem cell biology. Here, we review recent advances in the field relating to the upstream regulation of Hippo signalling and the intracellular tug-of-war that tightly controls its main target, the transcriptional co-activator Yorkie/YAP.
Collapse
Affiliation(s)
- Alexander Fulford
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Nicolas Tapon
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Paulo S Ribeiro
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
128
|
Cairns L, Tran T, Fowl BH, Patterson A, Kim YJ, Bothner B, Kavran JM. Salvador has an extended SARAH domain that mediates binding to Hippo kinase. J Biol Chem 2018. [PMID: 29519817 DOI: 10.1074/jbc.ra117.000923] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Hippo pathway controls cell proliferation and differentiation through the precisely tuned activity of a core kinase cassette. The activity of Hippo kinase is modulated by interactions between its C-terminal coiled-coil, termed the SARAH domain, and the SARAH domains of either dRassF or Salvador. Here, we wanted to understand the molecular basis of SARAH domain-mediated interactions and their influence on Hippo kinase activity. We focused on Salvador, a positive effector of Hippo activity and the least well-characterized SARAH domain-containing protein. We determined the crystal structure of a complex between Salvador and Hippo SARAH domains from Drosophila This structure provided insight into the organization of the Salvador SARAH domain including a folded N-terminal extension that expands the binding interface with Hippo SARAH domain. We also found that this extension improves the solubility of the Salvador SARAH domain, enhances binding to Hippo, and is unique to Salvador. We therefore suggest expanding the definition of the Salvador SARAH domain to include this extended region. The heterodimeric assembly observed in the crystal was confirmed by cross-linked MS and provided a structural basis for the mutually exclusive interactions of Hippo with either dRassF or Salvador. Of note, Salvador influenced the kinase activity of Mst2, the mammalian Hippo homolog. In co-transfected HEK293T cells, human Salvador increased the levels of Mst2 autophosphorylation and Mst2-mediated phosphorylation of select substrates, whereas Salvador SARAH domain inhibited Mst2 autophosphorylation in vitro These results suggest Salvador enhances the effects of Hippo kinase activity at multiple points in the Hippo pathway.
Collapse
Affiliation(s)
- Leah Cairns
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Thao Tran
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Brendan H Fowl
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Angela Patterson
- the Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, and
| | - Yoo Jin Kim
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Brian Bothner
- the Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, and
| | - Jennifer M Kavran
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215, .,the Departments of Biophysics and Biophysical Chemistry, School of Medicine, and.,Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 20215
| |
Collapse
|
129
|
TAOK1 negatively regulates IL-17-mediated signaling and inflammation. Cell Mol Immunol 2018; 15:794-802. [PMID: 29400705 DOI: 10.1038/cmi.2017.158] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/18/2017] [Accepted: 11/19/2017] [Indexed: 12/31/2022] Open
Abstract
Interleukin 17 (IL-17) is an important cytokine that can induce tissue inflammation and is involved in the pathogenesis of numerous autoimmune diseases. However, the regulation of its signaling transduction has not been well described. In this study, we report that thousand and one kinase 1 (TAOK1) functions as a negative regulator of IL-17-mediated signal transduction and inflammation. TAOK1 knockdown promotes IL-17-induced cytokine and chemokine expression and the activation of mitogen-activated protein kinases and nuclear factor-κB. We further demonstrate that TAOK1 interacts with IL-17 receptor A (IL-17RA) independent of its kinase activity, and TAOK1 dose-dependently prevents the formation of the IL-17R-Act1 (nuclear factor activator 1, also known as tumor necrosis factor receptor-associated factor 3 interacting protein 2) complex. Consistent with this, TAOK1 deficiency exacerbates colitis in the 2,4,6-trinitrobenzenesulfonic acid)-induced experimental model of inflammatory bowel disease, likely by its promotion of the IL-17-mediated signaling pathway. TAOK1 expression is decreased in the colons of ulcerative colitis patients. In conclusion, these findings suggest that TAOK1 is involved in the development of IL-17-related autoimmune disorders.
Collapse
|
130
|
Ardestani A, Maedler K. The Hippo Signaling Pathway in Pancreatic β-Cells: Functions and Regulations. Endocr Rev 2018; 39:21-35. [PMID: 29053790 DOI: 10.1210/er.2017-00167] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
Hippo signaling is an evolutionarily conserved pathway that critically regulates development and homeostasis of various tissues in response to a wide range of extracellular and intracellular signals. As an emerging important player in many diseases, the Hippo pathway is also involved in the pathophysiology of diabetes on the level of the pancreatic islets. Multiple lines of evidence uncover the importance of Hippo signaling in pancreas development as well as in the regulation of β-cell survival, proliferation, and regeneration. Hippo therefore represents a potential target for therapeutic agents designed to improve β-cell function and survival in diabetes. In this review, we summarize recent data on the regulation of the Hippo signaling pathway in the pancreas/in pancreatic islets, its functions on β-cell homeostasis in physiology and pathophysiology, and its contribution toward diabetes progression. The current knowledge related to general mechanisms of action and the possibility of exploiting the Hippo pathway for therapeutic approaches to block β-cell failure in diabetes is highlighted.
Collapse
Affiliation(s)
- Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| |
Collapse
|
131
|
Watt KI, Harvey KF, Gregorevic P. Regulation of Tissue Growth by the Mammalian Hippo Signaling Pathway. Front Physiol 2017; 8:942. [PMID: 29225579 PMCID: PMC5705614 DOI: 10.3389/fphys.2017.00942] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/07/2017] [Indexed: 12/14/2022] Open
Abstract
The integrative control of diverse biological processes such as proliferation, differentiation, apoptosis and metabolism is essential to maintain cellular and tissue homeostasis. Disruption of these underlie the development of many disease states including cancer and diabetes, as well as many of the complications that arise as a consequence of aging. These biological outputs are governed by many cellular signaling networks that function independently, and in concert, to convert changes in hormonal, mechanical and metabolic stimuli into alterations in gene expression. First identified in Drosophila melanogaster as a powerful mediator of cell division and apoptosis, the Hippo signaling pathway is a highly conserved regulator of mammalian organ size and functional capacity in both healthy and diseased tissues. Recent studies have implicated the pathway as an effector of diverse physiological cues demonstrating an essential role for the Hippo pathway as an integrative component of cellular homeostasis. In this review, we will: (a) outline the critical signaling elements that constitute the mammalian Hippo pathway, and how they function to regulate Hippo pathway-dependent gene expression and tissue growth, (b) discuss evidence that shows this pathway functions as an effector of diverse physiological stimuli and (c) highlight key questions in this developing field.
Collapse
Affiliation(s)
- Kevin I Watt
- Muscle Research and Therapeutics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Kieran F Harvey
- Department of Pathology, University of Melbourne, Melbourne, VIC, Australia.,Organogenesis and Cancer Programme, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Paul Gregorevic
- Muscle Research and Therapeutics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, University of Melbourne, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Department of Neurology, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
132
|
Bae JS, Kim SM, Lee H. The Hippo signaling pathway provides novel anti-cancer drug targets. Oncotarget 2017; 8:16084-16098. [PMID: 28035075 PMCID: PMC5362547 DOI: 10.18632/oncotarget.14306] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022] Open
Abstract
The Hippo signaling pathway plays a crucial role in cell proliferation, apoptosis, differentiation, and development. Major effectors of the Hippo signaling pathway include the transcriptional co-activators Yes-associated protein 1 (YAP) and WW domain-containing transcription regulator protein 1 (TAZ). The transcriptional activities of YAP and TAZ are affected by interactions with proteins from many diverse signaling pathways as well as responses to the external environment. High YAP and TAZ activity has been observed in many cancer types, and functional dysregulation of Hippo signaling enhances the oncogenic properties of YAP and TAZ and promotes cancer development. Many biological elements, including mechanical strain on the cell, cell polarity/adhesion molecules, other signaling pathways (e.g., G-protein-coupled receptor, epidermal growth factor receptor, Wnt, Notch, and transforming growth factor β/bone morphogenic protein), and cellular metabolic status, can promote oncogenesis through synergistic association with components of the Hippo signaling pathway. Here, we review the signaling networks that interact with the Hippo signaling pathway and discuss the potential of using drugs that inhibit YAP and TAZ activity for cancer therapy.
Collapse
Affiliation(s)
- June Sung Bae
- Biomolecular Function Research Branch, National Cancer Center, Goyang 10408, Republic of Korea
| | - Sun Mi Kim
- Biomolecular Function Research Branch, National Cancer Center, Goyang 10408, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
133
|
A Kinome RNAi Screen in Drosophila Identifies Novel Genes Interacting with Lgl, aPKC, and Crb Cell Polarity Genes in Epithelial Tissues. G3-GENES GENOMES GENETICS 2017; 7:2497-2509. [PMID: 28611255 PMCID: PMC5555457 DOI: 10.1534/g3.117.043513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In both Drosophila melanogaster and mammalian systems, epithelial structure and underlying cell polarity are essential for proper tissue morphogenesis and organ growth. Cell polarity interfaces with multiple cellular processes that are regulated by the phosphorylation status of large protein networks. To gain insight into the molecular mechanisms that coordinate cell polarity with tissue growth, we screened a boutique collection of RNAi stocks targeting the kinome for their capacity to modify Drosophila “cell polarity” eye and wing phenotypes. Initially, we identified kinase or phosphatase genes whose depletion modified adult eye phenotypes associated with the manipulation of cell polarity complexes (via overexpression of Crb or aPKC). We next conducted a secondary screen to test whether these cell polarity modifiers altered tissue overgrowth associated with depletion of Lgl in the wing. These screens identified Hippo, Jun kinase (JNK), and Notch signaling pathways, previously linked to cell polarity regulation of tissue growth. Furthermore, novel pathways not previously connected to cell polarity regulation of tissue growth were identified, including Wingless (Wg/Wnt), Ras, and lipid/Phospho-inositol-3-kinase (PI3K) signaling pathways. Additionally, we demonstrated that the “nutrient sensing” kinases Salt Inducible Kinase 2 and 3 (SIK2 and 3) are potent modifiers of cell polarity phenotypes and regulators of tissue growth. Overall, our screen has revealed novel cell polarity-interacting kinases and phosphatases that affect tissue growth, providing a platform for investigating molecular mechanisms coordinating cell polarity and tissue growth during development.
Collapse
|
134
|
Nakatani K, Maehama T, Nishio M, Goto H, Kato W, Omori H, Miyachi Y, Togashi H, Shimono Y, Suzuki A. Targeting the Hippo signalling pathway for cancer treatment. J Biochem 2017; 161:237-244. [PMID: 28003431 DOI: 10.1093/jb/mvw074] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022] Open
Abstract
The Hippo signalling pathway monitors cell-cell contact and external factors that shape tissue structure. In mice, tumourigenesis and developmental abnormalities are common consequences of dysregulated Hippo signalling. Expression of Hippo pathway components is also frequently altered in human tumours and correlates with poor prognosis and reduced patient survival. Thus, the Hippo pathway is an attractive anti-cancer target. Here, we provide an overview of the function and regulation of Hippo signalling components and summarize progress to date on the development of agents able to regulate Hippo signalling for cancer therapy.
Collapse
Affiliation(s)
- Keisuke Nakatani
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| | - Tomohiko Maehama
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Miki Nishio
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| | - Hiroki Goto
- Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| | - Wakako Kato
- Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| | - Hirofumi Omori
- Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| | - Yosuke Miyachi
- Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| | - Hideru Togashi
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yohei Shimono
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Akira Suzuki
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Division of Cancer Genetics, Medical Institute of Bioregulation Kyushu University, Fukuoka, Japan
| |
Collapse
|
135
|
Pfleger CM. The Hippo Pathway: A Master Regulatory Network Important in Development and Dysregulated in Disease. Curr Top Dev Biol 2017; 123:181-228. [PMID: 28236967 DOI: 10.1016/bs.ctdb.2016.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Hippo Pathway is a master regulatory network that regulates proliferation, cell growth, stemness, differentiation, and cell death. Coordination of these processes by the Hippo Pathway throughout development and in mature organisms in response to diverse external and internal cues plays a role in morphogenesis, in controlling organ size, and in maintaining organ homeostasis. Given the importance of these processes, the Hippo Pathway also plays an important role in organismal health and has been implicated in a variety of diseases including eye disease, cardiovascular disease, neurodegeneration, and cancer. This review will focus on Drosophila reports that identified the core components of the Hippo Pathway revealing specific downstream biological outputs of this complicated network. A brief description of mammalian reports will complement review of the Drosophila studies. This review will also survey upstream regulation of the core components with a focus on feedback mechanisms.
Collapse
Affiliation(s)
- Cathie M Pfleger
- The Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
136
|
Plouffe SW, Meng Z, Lin KC, Lin B, Hong AW, Chun JV, Guan KL. Characterization of Hippo Pathway Components by Gene Inactivation. Mol Cell 2016; 64:993-1008. [PMID: 27912098 PMCID: PMC5137798 DOI: 10.1016/j.molcel.2016.10.034] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/20/2016] [Accepted: 10/25/2016] [Indexed: 11/29/2022]
Abstract
The Hippo pathway is important for regulating tissue homeostasis, and its dysregulation has been implicated in human cancer. However, it is not well understood how the Hippo pathway becomes dysregulated because few mutations in core Hippo pathway components have been identified. Therefore, much work in the Hippo field has focused on identifying upstream regulators, and a complex Hippo interactome has been identified. Nevertheless, it is not always clear which components are the most physiologically relevant in regulating YAP/TAZ. To provide an overview of important Hippo pathway components, we created knockout cell lines for many of these components and compared their relative contributions to YAP/TAZ regulation in response to a wide range of physiological signals. By this approach, we provide an overview of the functional importance of many Hippo pathway components and demonstrate NF2 and RHOA as important regulators of YAP/TAZ and TAOK1/3 as direct kinases for LATS1/2.
Collapse
Affiliation(s)
- Steven W Plouffe
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Kimberly C Lin
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Brian Lin
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Audrey W Hong
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Justin V Chun
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
137
|
Abstract
The MST1 and MST2 protein kinases comprise the GCK-II subfamily of protein kinases. In addition to their amino-terminal kinase catalytic domain, related to that of the Saccharomyces cerevisiae protein kinase Ste20, their most characteristic feature is the presence near the carboxy terminus of a unique helical structure called a SARAH domain; this segment allows MST1/MST2 to homodimerize and to heterodimerize with the other polypeptides that contain SARAH domains, the noncatalytic polypeptides RASSF1-6 and Sav1/WW45. Early studies emphasized the potent ability of MST1/MST2 to induce apoptosis upon being overexpressed, as well as the conversion of the endogenous MST1/MST2 polypeptides to constitutively active, caspase-cleaved catalytic fragments during apoptosis initiated by any stimulus. Later, the cleaved, constitutively active form of MST1 was identified in nonapoptotic, quiescent adult hepatocytes as well as in cells undergoing terminal differentiation, where its presence is necessary to maintain those cellular states. The physiologic regulation of full length MST1/MST2 is controlled by the availability of its noncatalytic SARAH domain partners. Interaction with Sav1/WW45 recruits MST1/MST2 into a tumor suppressor pathway, wherein it phosphorylates and activates the Sav1-bound protein kinases Lats1/Lats2, potent inhibitors of the Yap1 and TAZ oncogenic transcriptional regulators. A constitutive interaction with the Rap1-GTP binding protein RASSF5B (Nore1B/RAPL) in T cells recruits MST1 (especially) and MST2 as an effector of Rap1's control of T cell adhesion and migration, a program crucial to immune surveillance and response; loss of function mutation in human MST1 results in profound immunodeficiency. MST1 and MST2 are also regulated by other protein kinases, positively by TAO1 and negatively by Par1, SIK2/3, Akt, and cRaf1. The growing list of candidate MST1/MST2 substrates suggests that the full range of MST1/MST2's physiologic programs and contributions to pathophysiology remains to be elucidated.
Collapse
Affiliation(s)
- Jacob A. Galan
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Diabetes Unit and Medical Services, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joseph Avruch
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Diabetes Unit and Medical Services, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
138
|
Chung HL, Augustine GJ, Choi KW. Drosophila Schip1 Links Expanded and Tao-1 to Regulate Hippo Signaling. Dev Cell 2016; 36:511-24. [PMID: 26954546 DOI: 10.1016/j.devcel.2016.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/26/2016] [Accepted: 02/04/2016] [Indexed: 12/12/2022]
Abstract
Regulation of organ size is essential in animal development, and Hippo (Hpo) signaling is a major conserved mechanism for controlling organ growth. In Drosophila, Hpo and Warts kinases are core components of this pathway and function as tumor suppressors by inhibiting Yorkie (Yki). Expanded (Ex) is a regulator of the Hpo activity, but how they are linked is unknown. Here, we show that Schip1, a Drosophila homolog of the mammalian Schwannomin interacting protein 1 (SCHIP1), provides a link between Ex and Hpo. Ex is required for apical localization of Schip1 in imaginal discs. Schip1 is necessary for promoting membrane localization and phosphorylation of Hpo by recruiting the Hpo kinase Tao-1. Taking these findings together, we conclude that Schip1 directly links Ex to Hpo signaling by recruiting Tao-1. This study provides insights into the mechanism of Tao-1 regulation and a potential growth control function for SCHIP1 in mammals.
Collapse
Affiliation(s)
- Hyung-Lok Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, South Korea; Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - George J Augustine
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791, South Korea; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553, Singapore; Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Kwang-Wook Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, South Korea.
| |
Collapse
|
139
|
RAC-LATS1/2 signaling regulates YAP activity by switching between the YAP-binding partners TEAD4 and RUNX3. Oncogene 2016; 36:999-1011. [DOI: 10.1038/onc.2016.266] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 12/14/2022]
|
140
|
Matsuda T, Zhai P, Sciarretta S, Zhang Y, Jeong JI, Ikeda S, Park J, Hsu CP, Tian B, Pan D, Sadoshima J, Del Re DP. NF2 Activates Hippo Signaling and Promotes Ischemia/Reperfusion Injury in the Heart. Circ Res 2016; 119:596-606. [PMID: 27402866 DOI: 10.1161/circresaha.116.308586] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/08/2016] [Indexed: 01/07/2023]
Abstract
RATIONALE NF2 (neurofibromin 2) is an established tumor suppressor that promotes apoptosis and inhibits growth in a variety of cell types, yet its function in cardiomyocytes remains largely unknown. OBJECTIVE We sought to determine the role of NF2 in cardiomyocyte apoptosis and ischemia/reperfusion (I/R) injury in the heart. METHODS AND RESULTS We investigated the function of NF2 in isolated cardiomyocytes and mouse myocardium at baseline and in response to oxidative stress. NF2 was activated in cardiomyocytes subjected to H2O2 and in murine hearts subjected to I/R. Increased NF2 expression promoted the activation of Mst1 (mammalian sterile 20-like kinase 1) and the inhibition of Yap (Yes-associated protein), whereas knockdown of NF2 attenuated these responses after oxidative stress. NF2 increased the apoptosis of cardiomyocytes that appeared dependent on Mst1 activity. Mice deficient for NF2 in cardiomyocytes, NF2 cardiomyocyte-specific knockout (CKO), were protected against global I/R ex vivo and showed improved cardiac functional recovery. Moreover, NF2 cardiomyocyte-specific knockout mice were protected against I/R injury in vivo and showed the upregulation of Yap target gene expression. Mechanistically, we observed nuclear association between NF2 and its activator MYPT-1 (myosin phosphatase target subunit 1) in cardiomyocytes, and a subpopulation of stress-induced nuclear Mst1 was diminished in NF2 CKO hearts. Finally, mice deficient for both NF2 and Yap failed to show protection against I/R indicating that Yap is an important target of NF2 in the adult heart. CONCLUSIONS NF2 is activated by oxidative stress in cardiomyocytes and mouse myocardium and facilitates apoptosis. NF2 promotes I/R injury through the activation of Mst1 and inhibition of Yap, thereby regulating Hippo signaling in the adult heart.
Collapse
Affiliation(s)
- Takahisa Matsuda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Yu Zhang
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Jae Im Jeong
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Shohei Ikeda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Jiyeon Park
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Chiao-Po Hsu
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Bin Tian
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Duojia Pan
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.)
| | - Dominic P Del Re
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers, New Jersey Medical School, Newark (T.M., P.Z., Y.Z., J.I.J., S.I., J.S., D.P.D.R.); Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers, New Jersey Medical School, Newark (J.P., B.T.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli (IS) (S.S.) and the Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Latina, Italy (S.S.); Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); and Howard Hughes Medical Institute and Department of Physiology, UT Southwestern Medical Center, Dallas, TX (D.P.).
| |
Collapse
|
141
|
Virbasius JV, Czech MP. Map4k4 Signaling Nodes in Metabolic and Cardiovascular Diseases. Trends Endocrinol Metab 2016; 27:484-492. [PMID: 27160798 PMCID: PMC4912878 DOI: 10.1016/j.tem.2016.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 12/25/2022]
Abstract
Mitogen-activated kinase kinase kinase kinase 4 (Map4k4), originally identified in small interfering (si)RNA screens and characterized by tissue-specific gene deletions, is emerging as a regulator of glucose homeostasis and cardiovascular health. Recent studies have shown that Map4k4 gene ablation or inhibition of its kinase activity attenuates hyperglycemia and plaque formation in mouse models of insulin resistance and atherosclerosis, and suggest roles for Map4k4 in multiple signaling systems, including NFκB activation, small GTPase regulation, the Hippo cascade, and regulation of cell dynamics by FERM domain proteins. This new and promising area of inquiry raises key questions that need to be addressed, such as defining which of the above or other effectors mediate Map4k4 control of metabolic and vascular functions, and identifying upstream activators of Map4k4.
Collapse
Affiliation(s)
- Joseph V Virbasius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
142
|
Sun S, Irvine KD. Cellular Organization and Cytoskeletal Regulation of the Hippo Signaling Network. Trends Cell Biol 2016; 26:694-704. [PMID: 27268910 DOI: 10.1016/j.tcb.2016.05.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/29/2016] [Accepted: 05/10/2016] [Indexed: 01/12/2023]
Abstract
The Hippo signaling network integrates diverse upstream signals to control cell fate decisions and regulate organ growth. Recent studies have provided new insights into the cellular organization of Hippo signaling, its relationship to cell-cell junctions, and how the cytoskeleton modulates Hippo signaling. Cell-cell junctions serve as platforms for Hippo signaling by localizing scaffolding proteins that interact with core components of the pathway. Interactions of Hippo pathway components with cell-cell junctions and the cytoskeleton also suggest potential mechanisms for the regulation of the pathway by cell contact and cell polarity. As our understanding of the complexity of Hippo signaling increases, a future challenge will be to understand how the diverse inputs into the pathway are integrated and to define their respective contributions in vivo.
Collapse
Affiliation(s)
- Shuguo Sun
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
143
|
Affiliation(s)
- Hyung-Lok Chung
- a Department of Biological Sciences , Korea Advanced Institute of Science and Technology , Daejeon , Korea
| | - Kwang-Wook Choi
- a Department of Biological Sciences , Korea Advanced Institute of Science and Technology , Daejeon , Korea
| |
Collapse
|
144
|
Abstract
In this review, Meng et al. focus on recent developments in our understanding of the molecular actions of the core Hippo kinase cascade and discuss key open questions in Hippo pathway regulation and function. The Hippo pathway was initially identified in Drosophila melanogaster screens for tissue growth two decades ago and has been a subject extensively studied in both Drosophila and mammals in the last several years. The core of the Hippo pathway consists of a kinase cascade, transcription coactivators, and DNA-binding partners. Recent studies have expanded the Hippo pathway as a complex signaling network with >30 components. This pathway is regulated by intrinsic cell machineries, such as cell–cell contact, cell polarity, and actin cytoskeleton, as well as a wide range of signals, including cellular energy status, mechanical cues, and hormonal signals that act through G-protein-coupled receptors. The major functions of the Hippo pathway have been defined to restrict tissue growth in adults and modulate cell proliferation, differentiation, and migration in developing organs. Furthermore, dysregulation of the Hippo pathway leads to aberrant cell growth and neoplasia. In this review, we focus on recent developments in our understanding of the molecular actions of the core Hippo kinase cascade and discuss key open questions in the regulation and function of the Hippo pathway.
Collapse
Affiliation(s)
- Zhipeng Meng
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Toshiro Moroishi
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| | - Kun-Liang Guan
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
145
|
He M, Zhou Z, Shah AA, Hong Y, Chen Q, Wan Y. New insights into posttranslational modifications of Hippo pathway in carcinogenesis and therapeutics. Cell Div 2016; 11:4. [PMID: 27042197 PMCID: PMC4818516 DOI: 10.1186/s13008-016-0013-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/22/2016] [Indexed: 02/05/2023] Open
Abstract
PTMs (posttranslational modifications) such as ubiquitylation, sumoylation, acetylation and protein methylation are pivotal modifiers that determine the activation, deactivation or subcellular localization of signaling proteins, facilitating the initiation, amplification and transduction of signaling. Accumulating evidence suggest that several key signaling molecules in Hippo signaling pathway are tightly regulated by various types of PTMs. Malfunction of these critical signaling modules such as YAP/TAZ, MAT1/2 and LATS1/2 due to deregulated PTMs has been linked to a variety of human diseases such as cancer. In this review article, we summarized the current understanding of the impact of PTMs in regulating Hippo signaling pathway and further discussed the potential therapeutic intervention from the view of PTMs and Hippo pathway.
Collapse
Affiliation(s)
- Mingjing He
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Hillman Cancer Center, 5117 Centre Avenue, HCC2.6c, Pittsburgh, PA 15213 USA ; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Peoples' Republic of China
| | - Zhuan Zhou
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Hillman Cancer Center, 5117 Centre Avenue, HCC2.6c, Pittsburgh, PA 15213 USA
| | - Anil A Shah
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Hillman Cancer Center, 5117 Centre Avenue, HCC2.6c, Pittsburgh, PA 15213 USA
| | - Yang Hong
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Hillman Cancer Center, 5117 Centre Avenue, HCC2.6c, Pittsburgh, PA 15213 USA
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Peoples' Republic of China
| | - Yong Wan
- Department of Cell Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Hillman Cancer Center, 5117 Centre Avenue, HCC2.6c, Pittsburgh, PA 15213 USA
| |
Collapse
|
146
|
Buglioni S, Vici P, Sergi D, Pizzuti L, Di Lauro L, Antoniani B, Sperati F, Terrenato I, Carosi M, Gamucci T, Vincenzoni C, Mariani L, Vizza E, Venuti A, Sanguineti G, Gadducci A, Barba M, Natoli C, Vitale I, Mottolese M, De Maria R, Maugeri-Saccà M. Analysis of the hippo transducers TAZ and YAP in cervical cancer and its microenvironment. Oncoimmunology 2016; 5:e1160187. [PMID: 27471633 DOI: 10.1080/2162402x.2016.1160187] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/12/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022] Open
Abstract
Hippo is a tumor-suppressor pathway that negatively regulates the oncoproteins TAZ and YAP. Moreover, Hippo affects the biology of a variety of non-neoplastic cells in the tumor microenvironment, even including immune cells. We herein assessed the predictive role of TAZ and YAP, assessed by immunohistochemistry, in 50 cervical cancer patients prevalently treated with neoadjuvant chemotherapy. Tumors were classified as positive or negative according to the percentage of tumor-expressing cells and cellular localization. TAZ/YAP were also evaluated in non-neoplastic cells, namely endothelial cells, non-lymphocytic stromal cells and tumor-infiltrating lymphocytes (TILs). TAZ expression in cancer cells (TAZ(pos)) was associated with a reduced pathological complete response (pCR) rate (p = 0.041). Conversely, the expression of TAZ and YAP in TILs (TAZ(TIL+) and YAP(TIL+)) seemed to be associated with increased pCRs (p = 0.083 and p = 0.018, respectively). When testing the predictive significance of the concomitant expression of TAZ in cancer cells and its absence in TILs (TAZ(pos)/TAZ(TIL-)), patients with TAZ(pos)/TAZ(TIL-) showed lower pCR rate (p = 0.001), as confirmed in multivariate analysis (TAZ(pos)/TAZ(TIL-): OR 8.67, 95% CI: 2.31-32.52, p = 0.001). Sensitivity analysis carried out in the 41 patients treated with neoadjuvant chemotherapy yielded comparable results (TAZ(pos)/TAZ(TIL-): OR 11.0, 95% CI: 2.42-49.91, p = 0.002). Internal validation carried out with two different procedures confirmed the robustness of this model. Overall, we found evidence on the association between TAZ expression in cervical cancer cells and reduced pCR rate. Conversely, the expression of the Hippo transducers in TILs may predict increased treatment efficacy, possibly mirroring the activation of a non-canonical Hippo/MST pathway necessary for T-cells activation and survival.
Collapse
Affiliation(s)
- Simonetta Buglioni
- Department of Pathology, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Barbara Antoniani
- Department of Pathology, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Francesca Sperati
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Irene Terrenato
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Mariantonia Carosi
- Department of Pathology, "Regina Elena" National Cancer Institute , Rome, Italy
| | | | - Cristina Vincenzoni
- Department of Surgery, Gynecologic Oncology Unit, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Luciano Mariani
- Department of Surgery, Gynecologic Oncology Unit, "Regina Elena" National Cancer Institute, Rome, Italy; HPV-UNIT, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Enrico Vizza
- Department of Surgery, Gynecologic Oncology Unit, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Aldo Venuti
- HPV-UNIT, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Giuseppe Sanguineti
- Department of Radiotherapy, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Angiolo Gadducci
- Department of Experimental and Clinical Medicine, Division of Gynecology and Obstetrics, University of Pisa , Pisa, Italy
| | - Maddalena Barba
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, Rome, Italy; Scientific Direction, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences, and CeSi-MeT, "G. d'Annunzio" University , Chieti, Italy
| | - Ilio Vitale
- Scientific Direction, "Regina Elena" National Cancer Institute, Rome, Italy; Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Marcella Mottolese
- Department of Pathology, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Ruggero De Maria
- Scientific Direction, "Regina Elena" National Cancer Institute , Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, Rome, Italy; Scientific Direction, "Regina Elena" National Cancer Institute, Rome, Italy
| |
Collapse
|
147
|
Poon CLC, Mitchell KA, Kondo S, Cheng LY, Harvey KF. The Hippo Pathway Regulates Neuroblasts and Brain Size in Drosophila melanogaster. Curr Biol 2016; 26:1034-42. [PMID: 26996505 DOI: 10.1016/j.cub.2016.02.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 12/22/2015] [Accepted: 02/02/2016] [Indexed: 12/12/2022]
Abstract
A key question in developmental neurobiology is how neural stem cells regulate their proliferative potential and cellular diversity and thus specify the overall size of the brain. Drosophila melanogaster neural stem cells (neuroblasts) are known to regulate their ability to self-renew by asymmetric cell division and produce different types of neurons and glia through sequential expression of temporal transcription factors [1]. Here, we show that the conserved Hippo pathway, a key regulator of epithelial organ size [2-4], restricts neuroblast proliferative potential and neuronal cell number to regulate brain size. The inhibition of Hippo pathway activity via depletion of the core kinases Tao-1, Hippo, or Warts regulates several key characteristics of neuroblasts during postembryonic neurogenesis. The Hippo pathway is required to maintain timely entry and exit from neurogenesis by regulating both neuroblast reactivation from quiescence and the time at which neuroblasts undergo terminal differentiation. Further, it restricts neuroblast cell-cycle speed, specifies cell size, and alters the proportion of neuron types generated during postembryonic neurogenesis. Collectively, deregulation of Hippo signaling in neuroblasts causes a substantial increase in overall brain size. We show that these effects are mediated via the key downstream transcription co-activator Yorkie and that, indeed, Yorkie overexpression in neuroblasts is sufficient to cause brain overgrowth. These studies reveal a novel mechanism that controls stem cell proliferative potential during postembryonic neurogenesis to regulate brain size.
Collapse
Affiliation(s)
- Carole L C Poon
- Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katrina A Mitchell
- Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shu Kondo
- Laboratory of Invertebrate Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, Japan
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
148
|
Maugeri-Saccà M, De Maria R. Hippo pathway and breast cancer stem cells. Crit Rev Oncol Hematol 2016; 99:115-22. [DOI: 10.1016/j.critrevonc.2015.12.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/16/2015] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
|
149
|
Yu FX, Zhao B, Guan KL. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2016; 163:811-28. [PMID: 26544935 DOI: 10.1016/j.cell.2015.10.044] [Citation(s) in RCA: 1682] [Impact Index Per Article: 186.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Indexed: 12/16/2022]
Abstract
Two decades of studies in multiple model organisms have established the Hippo pathway as a key regulator of organ size and tissue homeostasis. By inhibiting YAP and TAZ transcription co-activators, the Hippo pathway regulates cell proliferation, apoptosis, and stemness in response to a wide range of extracellular and intracellular signals, including cell-cell contact, cell polarity, mechanical cues, ligands of G-protein-coupled receptors, and cellular energy status. Dysregulation of the Hippo pathway exerts a significant impact on cancer development. Further investigation of the functions and regulatory mechanisms of this pathway will help uncovering the mystery of organ size control and identify new targets for cancer treatment.
Collapse
Affiliation(s)
- Fa-Xing Yu
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Bin Zhao
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
150
|
Liu B, Zheng Y, Yin F, Yu J, Silverman N, Pan D. Toll Receptor-Mediated Hippo Signaling Controls Innate Immunity in Drosophila. Cell 2016; 164:406-419. [PMID: 26824654 PMCID: PMC4733248 DOI: 10.1016/j.cell.2015.12.029] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 10/18/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023]
Abstract
The Hippo signaling pathway functions through Yorkie to control tissue growth and homeostasis. How this pathway regulates non-developmental processes remains largely unexplored. Here, we report an essential role for Hippo signaling in innate immunity whereby Yorkie directly regulates the transcription of the Drosophila IκB homolog, Cactus, in Toll receptor-mediated antimicrobial response. Loss of Hippo pathway tumor suppressors or activation of Yorkie in fat bodies, the Drosophila immune organ, leads to elevated cactus mRNA levels, decreased expression of antimicrobial peptides, and vulnerability to infection by Gram-positive bacteria. Furthermore, Gram-positive bacteria acutely activate Hippo-Yorkie signaling in fat bodies via the Toll-Myd88-Pelle cascade through Pelle-mediated phosphorylation and degradation of the Cka subunit of the Hippo-inhibitory STRIPAK PP2A complex. Our studies elucidate a Toll-mediated Hippo signaling pathway in antimicrobial response, highlight the importance of regulating IκB/Cactus transcription in innate immunity, and identify Gram-positive bacteria as extracellular stimuli of Hippo signaling under physiological settings.
Collapse
Affiliation(s)
- Bo Liu
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yonggang Zheng
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feng Yin
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jianzhong Yu
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Neal Silverman
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Duojia Pan
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|