51
|
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
|
52
|
Li A, Gu K, Wang Q, Chen X, Fu X, Wang Y, Wen Y. Epigallocatechin-3-gallate affects the proliferation, apoptosis, migration and invasion of tongue squamous cell carcinoma through the hippo-TAZ signaling pathway. Int J Mol Med 2018; 42:2615-2627. [PMID: 30106116 PMCID: PMC6192764 DOI: 10.3892/ijmm.2018.3818] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/07/2018] [Indexed: 01/23/2023] Open
Abstract
The purpose of the present study was to investigate the mechanism by which epigallocatechin-3-gallate (EGCG) inhibits the biological behaviors of the tongue squamous cell carcinoma (TSCC) through the Hippo-tafazzin (TAZ) signaling pathway and to provide insights into molecular targeted therapy in TSCC. CAL27 and SCC15 cells were treated with different concentrations of EGCG for 24 h. Cell proliferation was determined using Cell-Counting Kit-8 and EdU assays. Cell apoptosis was evaluated by flow cytometry. Cell migration and invasion were measured using scratch and Transwell assays, respectively. Furthermore, protein levels of associated target genes were detected using a western blot assay. It was demonstrated that EGCG affected biological behaviors of CAL27 and SCC15 cells in concentration- and time-dependent manners. In addition, EGCG decreased the protein levels of TAZ, LATS1, MOB1 and JNK. Overexpression of TAZ alleviated the effect of EGCG on CAL27 cells. Furthermore, the combination of EGCG and simvastatin inhibited the proliferation, migration and invasion, and promoted apoptosis significantly compared with single treatment in CAL27 cells. The results of the present study suggested that EGCG affects proliferation, apoptosis, migration and invasion of TSCC through the Hippo-TAZ signaling pathway.
Collapse
Affiliation(s)
- Aonan Li
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ke Gu
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qi Wang
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiyan Chen
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xucheng Fu
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ying Wang
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yong Wen
- Department of Implantology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
53
|
Foxler DE, Bridge KS, Foster JG, Grevitt P, Curry S, Shah KM, Davidson KM, Nagano A, Gadaleta E, Rhys HI, Kennedy PT, Hermida MA, Chang TY, Shaw PE, Reynolds LE, McKay TR, Wang HW, Ribeiro PS, Plevin MJ, Lagos D, Lemoine NR, Rajan P, Graham TA, Chelala C, Hodivala-Dilke KM, Spendlove I, Sharp TV. A HIF-LIMD1 negative feedback mechanism mitigates the pro-tumorigenic effects of hypoxia. EMBO Mol Med 2018; 10:e8304. [PMID: 29930174 PMCID: PMC6079541 DOI: 10.15252/emmm.201708304] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 05/23/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022] Open
Abstract
The adaptive cellular response to low oxygen tensions is mediated by the hypoxia-inducible factors (HIFs), a family of heterodimeric transcription factors composed of HIF-α and HIF-β subunits. Prolonged HIF expression is a key contributor to cellular transformation, tumorigenesis and metastasis. As such, HIF degradation under hypoxic conditions is an essential homeostatic and tumour-suppressive mechanism. LIMD1 complexes with PHD2 and VHL in physiological oxygen levels (normoxia) to facilitate proteasomal degradation of the HIF-α subunit. Here, we identify LIMD1 as a HIF-1 target gene, which mediates a previously uncharacterised, negative regulatory feedback mechanism for hypoxic HIF-α degradation by modulating PHD2-LIMD1-VHL complex formation. Hypoxic induction of LIMD1 expression results in increased HIF-α protein degradation, inhibiting HIF-1 target gene expression, tumour growth and vascularisation. Furthermore, we report that copy number variation at the LIMD1 locus occurs in 47.1% of lung adenocarcinoma patients, correlates with enhanced expression of a HIF target gene signature and is a negative prognostic indicator. Taken together, our data open a new field of research into the aetiology, diagnosis and prognosis of LIMD1-negative lung cancers.
Collapse
Affiliation(s)
- Daniel E Foxler
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Katherine S Bridge
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - John G Foster
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Paul Grevitt
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Sean Curry
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, UK
| | - Kunal M Shah
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Kathryn M Davidson
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ai Nagano
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Emanuela Gadaleta
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Paul T Kennedy
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Miguel A Hermida
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ting-Yu Chang
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei City, Taiwan
| | - Peter E Shaw
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, UK
| | - Louise E Reynolds
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Tristan R McKay
- School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Hsei-Wei Wang
- Institute of Microbiology and Immunology, National Yang Ming University, Taipei City, Taiwan
| | - Paulo S Ribeiro
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Dimitris Lagos
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, UK
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Prabhakar Rajan
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Trevor A Graham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Ian Spendlove
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, UK
| | - Tyson V Sharp
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
54
|
Abstract
Polyploid cells, which contain multiple copies of the typically diploid genome, are widespread in plants and animals. Polyploidization can be developmentally programmed or stress induced, and arises from either cell-cell fusion or a process known as endoreplication, in which cells replicate their DNA but either fail to complete cytokinesis or to progress through M phase entirely. Polyploidization offers cells several potential fitness benefits, including the ability to increase cell size and biomass production without disrupting cell and tissue structure, and allowing improved cell longevity through higher tolerance to genomic stress and apoptotic signals. Accordingly, recent studies have uncovered crucial roles for polyploidization in compensatory cell growth during tissue regeneration in the heart, liver, epidermis and intestine. Here, we review current knowledge of the molecular pathways that generate polyploidy and discuss how polyploidization is used in tissue repair and regeneration.
Collapse
Affiliation(s)
| | - Bruce A Edgar
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| |
Collapse
|
55
|
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
|
56
|
Identification of raw as a regulator of glial development. PLoS One 2018; 13:e0198161. [PMID: 29813126 PMCID: PMC5973607 DOI: 10.1371/journal.pone.0198161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/15/2018] [Indexed: 12/18/2022] Open
Abstract
Glial cells perform numerous functions to support neuron development and function, including axon wrapping, formation of the blood brain barrier, and enhancement of synaptic transmission. We have identified a novel gene, raw, which functions in glia of the central and peripheral nervous systems in Drosophila. Reducing Raw levels in glia results in morphological defects in the brain and ventral nerve cord, as well as defects in neuron function, as revealed by decreased locomotion in crawling assays. Examination of the number of glia along peripheral nerves reveals a reduction in glial number upon raw knockdown. The reduced number of glia along peripheral nerves occurs as a result of decreased glial proliferation. As Raw has been shown to negatively regulate Jun N-terminal kinase (JNK) signaling in other developmental contexts, we examined the expression of a JNK reporter and the downstream JNK target, matrix metalloproteinase 1 (mmp1), and found that raw knockdown results in increased reporter activity and Mmp1 levels. These results are consistent with previous studies showing increased Mmp levels lead to nerve cord defects similar to those observed upon raw knockdown. In addition, knockdown of puckered, a negative feedback regulator of JNK signaling, also causes a decrease in glial number. Thus, our studies have resulted in the identification of a new regulator of gliogenesis, and demonstrate that increased JNK signaling negatively impacts glial development.
Collapse
|
57
|
Chu CW, Xiang B, Ossipova O, Ioannou A, Sokol SY. The Ajuba family protein Wtip regulates actomyosin contractility during vertebrate neural tube closure. J Cell Sci 2018; 131:jcs.213884. [PMID: 29661847 DOI: 10.1242/jcs.213884] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/06/2018] [Indexed: 12/16/2022] Open
Abstract
Ajuba family proteins are implicated in the assembly of cell junctions and have been reported to antagonize Hippo signaling in response to cytoskeletal tension. To assess the role of these proteins in actomyosin contractility, we examined the localization and function of Wtip, a member of the Ajuba family, in Xenopus early embryos. Targeted in vivo depletion of Wtip inhibited apical constriction in neuroepithelial cells and elicited neural tube defects. Fluorescent protein-tagged Wtip showed predominant punctate localization along the cell junctions in the epidermis and a linear junctional pattern in the neuroectoderm. In cells undergoing Shroom3-induced apical constriction, the punctate distribution was reorganized into a linear pattern. Conversely, the linear junctional pattern of Wtip in neuroectoderm changed to a more punctate distribution in cells with reduced myosin II activity. The C-terminal fragment of Wtip physically associated with Shroom3 and interfered with Shroom3 activity and neural fold formation. We therefore propose that Wtip is a tension-sensitive cytoskeletal adaptor that regulates apical constriction during vertebrate neurulation.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Chih-Wen Chu
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bo Xiang
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Olga Ossipova
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andriani Ioannou
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
58
|
Stuelten CH, Parent CA, Montell DJ. Cell motility in cancer invasion and metastasis: insights from simple model organisms. Nat Rev Cancer 2018; 18:296-312. [PMID: 29546880 PMCID: PMC6790333 DOI: 10.1038/nrc.2018.15] [Citation(s) in RCA: 352] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metastasis remains the greatest challenge in the clinical management of cancer. Cell motility is a fundamental and ancient cellular behaviour that contributes to metastasis and is conserved in simple organisms. In this Review, we evaluate insights relevant to human cancer that are derived from the study of cell motility in non-mammalian model organisms. Dictyostelium discoideum, Caenorhabditis elegans, Drosophila melanogaster and Danio rerio permit direct observation of cells moving in complex native environments and lend themselves to large-scale genetic and pharmacological screening. We highlight insights derived from each of these organisms, including the detailed signalling network that governs chemotaxis towards chemokines; a novel mechanism of basement membrane invasion; the positive role of E-cadherin in collective direction-sensing; the identification and optimization of kinase inhibitors for metastatic thyroid cancer on the basis of work in flies; and the value of zebrafish for live imaging, especially of vascular remodelling and interactions between tumour cells and host tissues. While the motility of tumour cells and certain host cells promotes metastatic spread, the motility of tumour-reactive T cells likely increases their antitumour effects. Therefore, it is important to elucidate the mechanisms underlying all types of cell motility, with the ultimate goal of identifying combination therapies that will increase the motility of beneficial cells and block the spread of harmful cells.
Collapse
Affiliation(s)
- Christina H. Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Carole A. Parent
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
- Department of Pharmacology, Michigan Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- ;
| | - Denise J. Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA, USA
- ;
| |
Collapse
|
59
|
Abstract
Head and neck squamous cell carcinomas (HNSCCs) arise in the mucosal linings of the upper aerodigestive tract and are unexpectedly heterogeneous in nature. Classical risk factors are smoking and excessive alcohol consumption, and in recent years, the role of human papillomavirus (HPV) has emerged, particularly in oropharyngeal tumours. HPV-induced oropharyngeal tumours are considered a separate disease entity, which recently has manifested in an adapted prognostic staging system while the results of de-intensified treatment trials are awaited. Carcinogenesis caused by HPV in the mucosal linings of the upper aerodigestive tract remains an enigma, but with some recent observations, a model can be proposed. In 2015, The Cancer Genome Atlas (TCGA) consortium published a comprehensive molecular catalogue on HNSCC. Frequent mutations of novel druggable oncogenes were not demonstrated, but the existence of a subgroup of genetically distinct HPV-negative head and neck tumours with favourable prognoses was confirmed. Tumours can be further subclassified based on genomic profiling. However, the amount of molecular data is currently overwhelming and requires detailed biological interpretation. It also became apparent that HNSCC is a disease characterized by frequent mutations that create neoantigens, indicating that immunotherapies might be effective. In 2016, the first results of immunotherapy trials with immune checkpoint inhibitors were published, and these may be considered as a paradigm shift in head and neck oncology.
Collapse
Affiliation(s)
- C René Leemans
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center
| | - Peter J F Snijders
- Department of Pathology, VU University Medical Center, Amsterdam, Netherlands
| | - Ruud H Brakenhoff
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center
| |
Collapse
|
60
|
Abstract
The Hippo signal transduction pathway is an important regulator of organ growth and cell differentiation, and its deregulation contributes to the development of cancer. The activity of the Hippo pathway is strongly dependent on cell junctions, cellular architecture, and the mechanical properties of the microenvironment. In this review, we discuss recent advances in our understanding of how cell junctions transduce signals from the microenvironment and control the activity of the Hippo pathway. We also discuss how these mechanisms may control organ growth during development and regeneration, and how defects in them deregulate Hippo signaling in cancer cells.
Collapse
Affiliation(s)
- Ruchan Karaman
- VIB Center for Cancer Biology, University of Leuven, 3000 Leuven, Belgium.,Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| | - Georg Halder
- VIB Center for Cancer Biology, University of Leuven, 3000 Leuven, Belgium.,Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|
61
|
Drosophila as a Model System to Study Cell Signaling in Organ Regeneration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7359267. [PMID: 29750169 PMCID: PMC5884440 DOI: 10.1155/2018/7359267] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
Regeneration is a fascinating phenomenon that allows organisms to replace or repair damaged organs or tissues. This ability occurs to varying extents among metazoans. The rebuilding of the damaged structure depends on regenerative proliferation that must be accompanied by proper cell fate respecification and patterning. These cellular processes are regulated by the action of different signaling pathways that are activated in response to the damage. The imaginal discs of Drosophila melanogaster have the ability to regenerate and have been extensively used as a model system to study regeneration. Drosophila provides an opportunity to use powerful genetic tools to address fundamental problems about the genetic mechanisms involved in organ regeneration. Different studies in Drosophila have helped to elucidate the genes and signaling pathways that initiate regeneration, promote regenerative growth, and induce cell fate respecification. Here we review the signaling networks involved in regulating the variety of cellular responses that are required for discs regeneration.
Collapse
|
62
|
O'Connell KS, McGregor NW, Lochner C, Emsley R, Warnich L. The genetic architecture of schizophrenia, bipolar disorder, obsessive-compulsive disorder and autism spectrum disorder. Mol Cell Neurosci 2018; 88:300-307. [PMID: 29505902 DOI: 10.1016/j.mcn.2018.02.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/22/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Considerable evidence suggests that autism spectrum disorders (ASD), schizophrenia (SCZ), bipolar disorder (BD) and obsessive-compulsive disorder (OCD) share a common molecular aetiology, despite their unique clinical diagnostic criteria. The aim of this study was therefore to determine and characterise the common and unique molecular architecture of ASD, SCZ, BD and OCD. Gene lists were obtained from previously published studies for ASD, BD, SCZ and for OCD. Genes identified to be common to all disorders, or unique to one specific disorder, were included for enrichment analyses using the web-server tool Enrichr. Ten genes were identified to be commonly associated with the aetiology of ASD, SCZ, BD and OCD. Enrichment analyses determined that these genes are predominantly involved in the dopaminergic and serotonergic pathways, the voltage-gated calcium ion channel gene network, folate metabolism, regulation of the hippo signaling pathway, and the regulation of gene silencing and expression. In addition to well-characterised and previously described pathways, regulation of the hippo signaling pathway was commonly associated with ASD, SCZ, BD and OCD, implicating neural development and neuronal maintenance as key in neuropsychiatric disorders. In contrast, a large number of previously associated genes were shown to be disorder-specific. And unique disorder-specific pathways and biological processes were presented for ASD, BD, SCZ and OCD aetiology. Considering the current global incidence and prevalence rates of mental health disorders, focus should be placed on cross-disorder commonalities in order to realise actionable and translatable results to combat mental health disorders.
Collapse
Affiliation(s)
- Kevin S O'Connell
- System Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa; Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Nathaniel W McGregor
- System Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa; Department of Genetics, Stellenbosch University, Stellenbosch, South Africa.
| | - Christine Lochner
- SU/UCT MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Tygerberg, South Africa
| | - Robin Emsley
- Department of Psychiatry, Stellenbosch University, Tygerberg, South Africa
| | - Louise Warnich
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
63
|
Ibar C, Kirichenko E, Keepers B, Enners E, Fleisch K, Irvine KD. Tension-dependent regulation of mammalian Hippo signaling through LIMD1. J Cell Sci 2018; 131:jcs214700. [PMID: 29440237 PMCID: PMC5897721 DOI: 10.1242/jcs.214700] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/31/2018] [Indexed: 12/26/2022] Open
Abstract
Hippo signaling is regulated by biochemical and biomechanical cues that influence the cytoskeleton, but the mechanisms that mediate this have remained unclear. We show that all three mammalian Ajuba family proteins - AJUBA, LIMD1 and WTIP - exhibit tension-dependent localization to adherens junctions, and that both LATS family proteins, LATS1 and LATS2, exhibit an overlapping tension-dependent junctional localization. This localization of Ajuba and LATS family proteins is also influenced by cell density, and by Rho activation. We establish that junctional localization of LATS kinases requires LIMD1, and that LIMD1 is also specifically required for the regulation of LATS kinases and YAP1 by Rho. Our results identify a biomechanical pathway that contributes to regulation of mammalian Hippo signaling, establish that this occurs through tension-dependent LIMD1-mediated recruitment and inhibition of LATS kinases in junctional complexes, and identify roles for this pathway in both Rho-mediated and density-dependent regulation of Hippo signaling.
Collapse
Affiliation(s)
- Consuelo Ibar
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Elmira Kirichenko
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Benjamin Keepers
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Edward Enners
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Katelyn Fleisch
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| |
Collapse
|
64
|
Dutta S, Mana-Capelli S, Paramasivam M, Dasgupta I, Cirka H, Billiar K, McCollum D. TRIP6 inhibits Hippo signaling in response to tension at adherens junctions. EMBO Rep 2018; 19:337-350. [PMID: 29222344 PMCID: PMC5797958 DOI: 10.15252/embr.201744777] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 11/09/2022] Open
Abstract
The transcriptional co-activator YAP controls cell proliferation, survival, and tissue regeneration in response to changes in the mechanical environment. It is not known how mechanical stimuli such as tension are sensed and how the signal is transduced to control YAP activity. Here, we show that the LIM domain protein TRIP6 acts as part of a mechanotransduction pathway at adherens junctions to promote YAP activity by inhibiting the LATS1/2 kinases. Previous studies showed that vinculin at adherens junctions becomes activated by mechanical tension. We show that vinculin inhibits Hippo signaling by recruiting TRIP6 to adherens junctions and stimulating its binding to and inhibition of LATS1/2 in response to tension. TRIP6 competes with MOB1 for binding to LATS1/2 thereby blocking MOB1 from recruiting the LATS1/2 activating kinases MST1/2. Together, these findings reveal a novel pathway that responds to tension at adherens junctions to control Hippo pathway signaling.
Collapse
Affiliation(s)
- Shubham Dutta
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sebastian Mana-Capelli
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Murugan Paramasivam
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ishani Dasgupta
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Heather Cirka
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Kris Billiar
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Dannel McCollum
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
65
|
The Hippo pathway as a drug target in gastric cancer. Cancer Lett 2018; 420:14-25. [PMID: 29408652 DOI: 10.1016/j.canlet.2018.01.062] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
Abstract
The Hippo tumor suppressor pathway is critical for balancing cellular differentiation and proliferation in response to cell-cell contact, mechanical signals and diffusible signals such as lysophosphatidic acid. Hippo pathway signaling is frequently dysregulated in gastric cancer (GC), as well as many other kinds of solid tumors, contributing to multiple aspects of malignant progression including unchecked cell division and metastasis. Considering the importance of this Hippo pathway in cancer, its pharmacological disruption may be of huge benefit in the fight against this disease. In this review, we summarize the components of the Hippo pathway, its crosstalk with other major oncogenic signaling pathways, common mechanisms of its dysregulation, as well as potential therapeutic approaches of targeting this pathway for cancer treatment, specifically in a GC context.
Collapse
|
66
|
Yang W, Han W, Qin A, Wang Z, Xu J, Qian Y. The emerging role of Hippo signaling pathway in regulating osteoclast formation. J Cell Physiol 2018; 233:4606-4617. [PMID: 29219182 DOI: 10.1002/jcp.26372] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/01/2017] [Indexed: 12/14/2022]
Abstract
A delicate balance between osteoblastic bone formation and osteoclastic bone resorption is crucial for bone homeostasis. This process is regulated by the Hippo signaling pathway including key regulatory molecules RASSF2, NF2, MST1/2, SAV1, LATS1/2, MOB1, YAP, and TAZ. It is well established that the Hippo signaling pathway plays an important part in regulating osteoblast differentiation, but its role in osteoclast formation and activation remains poorly understood. In this review, we discuss the emerging role of Hippo-signaling pathway in osteoclast formation and bone homeostasis. It is revealed that specific molecules of the Hippo-signaling pathway take part in a stage specific regulation in pre-osteoclast proliferation, osteoclast differentiation and osteoclast apoptosis and survival. Upon activation, MST and LAST, transcriptional co-activators YAP and TAZ bind to the members of the TEA domain (TEAD) family transcription factors, and influence osteoclast differentiation via regulating the expression of downstream target genes such as connective tissue growth factor (CTGF/CCN2) and cysteine-rich protein 61 (CYR61/CCN1). In addition, through interacting or cross talking with RANKL-mediated signaling cascades including NF-κB, MAPKs, AP1, and NFATc1, Hippo-signaling molecules such as YAP/TAZ/TEAD complex, RASSF2, MST2, and Ajuba could also potentially modulate osteoclast differentiation and function. Elucidating the roles of the Hippo-signaling pathway in osteoclast development and specific molecules involved is important for understanding the mechanism of bone homeostasis and diseases.
Collapse
Affiliation(s)
- Wanlei Yang
- Department of Orthopaedics, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, P. R. China
| | - Weiqi Han
- Department of Orthopaedics, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, P. R. China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyi Wang
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Yu Qian
- Department of Orthopaedics, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, P. R. China
| |
Collapse
|
67
|
Shi X, Chen Z, Hu X, Luo M, Sun Z, Li J, Shi S, Feng X, Zhou C, Li Z, Yang W, Li Y, Wang P, Zhou F, Gao Y, He J. AJUBA promotes the migration and invasion of esophageal squamous cell carcinoma cells through upregulation of MMP10 and MMP13 expression. Oncotarget 2017; 7:36407-36418. [PMID: 27172796 PMCID: PMC5095009 DOI: 10.18632/oncotarget.9239] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/22/2016] [Indexed: 12/21/2022] Open
Abstract
The LIM-domain protein AJUBA has been reported to be involved in cell-cell adhesion, proliferation, migration and cell fate decision by acting as a scaffold or adaptor protein. We previously identified AJUBA as a putative cancer gene in esophageal squamous cell carcinoma (ESCC). However, the function and underlying mechanisms of AJUBA in ESCC remain largely unknown. In the present study, we detected AJUBA levels in ESCC tumor tissues and in corresponding adjacent non-tumor tissues by immunohistochemistry (IHC) and investigated the function and mechanism of AJUBA in ESCC cells. The IHC results showed that AJUBA levels were significantly higher in ESCC tissues compared with corresponding adjacent non-tumor tissues (P < 0.001). Both in vitro and in vivo experiments showed that AJUBA promoted cell growth and colony formation, inhibited cisplatin-induced apoptosis of ESCC cells, and promoted ESCC cell migration and invasion. RNA sequencing was used to reveal the oncogenic pathways of AJUBA that were involved, and MMP10 and MMP13 were identified as two of the downstream targets of AJUBA. Thus, AJUBA upregulates the levels of MMP10 and MMP13 by activating ERK1/2. Taken together, these findings revealed that AJUBA serves as oncogenic gene in ESCC and may serve as a new target for ESCC therapy.
Collapse
Affiliation(s)
- Xuejiao Shi
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhaoli Chen
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xueda Hu
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Mei Luo
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zengmiao Sun
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiagen Li
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Susheng Shi
- Department of Pathology, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoli Feng
- Department of Pathology, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chengcheng Zhou
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zitong Li
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Wenhui Yang
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Li
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Pan Wang
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Fang Zhou
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yibo Gao
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
68
|
Bi L, Ma F, Tian R, Zhou Y, Lan W, Song Q, Cheng X. AJUBA increases the cisplatin resistance through hippo pathway in cervical cancer. Gene 2017; 644:148-154. [PMID: 29126926 DOI: 10.1016/j.gene.2017.11.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Though LIM-domain protein AJUBA was identified as a putative oncogene, the function and underlying mechanisms of AJUBA in cervical cancer remain largely unknown. Firstly, AJUBA expression was detected via real-time quantitative PCR in patients' samples. Furthermore, Hela and Siha cells were transfected with AJUBA-overexpressing plasmids, and then exposed to cisplatin, the apoptosis was measured by cytometry assay. In addition, the expression of YAP and TAZ was disclosed through western blot assay. Our results revealed that AJUBA expression was significantly higher in the cervical cancer patients resistant to cisplatin treatment compared with cervical cancer patients sensitive to cisplatin treatment. In addition, overall survival time was significantly shorter in the cervical cancer patients with high AJUBA expression compare with those with low AJUBA expression using kaplan-meier analysis. Hela and Siha cells transfected with AJUBA-expressing plasmids exposed to cisplatin treatment had higher survival rate compared with the cells transfected with empty vector control. Mechanistic studies revealed the AJUBA upregulated the downstream targets YAP and TAZ. These results suggest that high AJUBA level enhances cervical cancer cells drug resistance to cisplatin, also associates with decreased patient survival times.
Collapse
Affiliation(s)
- Lihong Bi
- Department of Gynecology, PKUCare Luzhong Hospital, Zibo, Shandong, China
| | - Feng Ma
- Department of Oncology, PKUCare Luzhong Hospital, Zibo, Shandong, China.
| | - Rui Tian
- Department of Gynecology, PKUCare Luzhong Hospital, Zibo, Shandong, China
| | - Yanli Zhou
- Department of Pharmacy, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Weiguang Lan
- Department of Oncology, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Quanmao Song
- Department of Oncology, PKUCare Luzhong Hospital, Zibo, Shandong, China
| | - Xiankui Cheng
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
69
|
Martín R, Pinal N, Morata G. Distinct regenerative potential of trunk and appendages of Drosophila mediated by JNK signalling. Development 2017; 144:3946-3956. [PMID: 28935711 DOI: 10.1242/dev.155507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 09/11/2017] [Indexed: 12/31/2022]
Abstract
The Drosophila body comprises a central part, the trunk, and outgrowths of the trunk, the appendages. Much is known about appendage regeneration, but little about the trunk. As the wing imaginal disc contains a trunk component, the notum, and a wing appendage, we have investigated the response to ablation of these two components. We find that, in contrast with the strong regenerative response of the wing, the notum does not regenerate. Nevertheless, the elimination of the wing primordium elicits a proliferative response of notum cells, but they do not regenerate wing; they form a notum duplicate. Conversely, the wing cells cannot regenerate an ablated notum; they overproliferate and generate a hinge overgrowth. These results suggest that trunk and appendages cannot be reprogrammed to generate each other. Our experiments demonstrate that the proliferative response is mediated by JNK signalling from dying cells, but JNK functions differently in the trunk and the appendages, which may explain their distinct regenerative potential.
Collapse
Affiliation(s)
- Raquel Martín
- Centro de Biología Molecular CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Noelia Pinal
- Centro de Biología Molecular CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ginés Morata
- Centro de Biología Molecular CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
70
|
Lin AYT, Pearson BJ. Yorkie is required to restrict the injury responses in planarians. PLoS Genet 2017; 13:e1006874. [PMID: 28686611 PMCID: PMC5515462 DOI: 10.1371/journal.pgen.1006874] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 07/18/2017] [Accepted: 06/15/2017] [Indexed: 12/30/2022] Open
Abstract
Regeneration requires the precise integration of cues that initiate proliferation, direct differentiation, and ultimately re-pattern tissues to the proper size and scale. Yet how these processes are integrated with wounding responses remains relatively unknown. The freshwater planarian, Schmidtea mediterranea, is an ideal model to study the stereotyped proliferative and transcriptional responses to injury due to its high capacity for regeneration. Here, we characterize the effector of the Hippo signalling cascade, yorkie, during planarian regeneration and its role in restricting early injury responses. In yki(RNAi) regenerating animals, wound responses are hyper-activated such that both stem cell proliferation and the transcriptional wound response program are heighted and prolonged. Using this observation, we also uncovered novel wound-induced genes by RNAseq that were de-repressed in yki(RNAi) animals compared with controls. Additionally, we show that yki(RNAi) animals have expanded epidermal and muscle cell populations, which we hypothesize are the increased sources of wound-induced genes. Finally, we show that in yki(RNAi) animals, the sensing of the size of an injury by eyes or the pharynx is not appropriate, and the brain, gut, and midline cannot remodel or scale correctly to the size of the regenerating fragment. Taken together, our results suggest that yki functions as a key molecule that can integrate multiple aspects of the injury response including proliferation, apoptosis, injury-induced transcription, and patterning. The planarian displays a remarkable ability to regenerate any tissue from mere fragments of its original size. This high capacity to regenerate is attributed to the abundant population of pluripotent adult stem cells. In response to an injury, such as an amputation, stem cells proliferate and replace the lost tissues de novo (epimorphosis), whereas existing tissue must rescale to the correct proportions in relation to the new fragment size (morphallaxis). Currently, the molecules that control either the responses to injury or the ones that mediate size and scaling are not well understood. For instance, how are the injury responses precisely activated and shut down to ensure regenerating tissues are not under- or overgrown? Here, we find that Yki, the effector of the Hippo signalling cascade, is a critical molecule that influences several injury processes during regeneration. Loss of Yki function in regenerating animals resulted in increased and temporally dysregulated expression of wound-induced genes, proliferation, and apoptosis. Genes that are injury induced were mis-expressed in yki(RNAi) animals, which also showed increases in the epidermal and muscle cell populations. Taken together, our findings suggest that the injury responses must be restricted to ensure proper regenerative outcomes of correct scale, and that Yki is a key regulator in these processes.
Collapse
Affiliation(s)
- Alexander Y. T. Lin
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Bret J. Pearson
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
71
|
Jiménez AP, Traum A, Boettger T, Hackstein H, Richter AM, Dammann RH. The tumor suppressor RASSF1A induces the YAP1 target gene ANKRD1 that is epigenetically inactivated in human cancers and inhibits tumor growth. Oncotarget 2017; 8:88437-88452. [PMID: 29179447 PMCID: PMC5687617 DOI: 10.18632/oncotarget.18177] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 05/12/2017] [Indexed: 12/20/2022] Open
Abstract
The Hippo pathway regulates organ size, growth and comprises several tumor related factors, including the oncoprotein YAP1 and the tumor suppressor RASSF1A. RASSF1A is frequently epigenetically inactivated in cancer. In our study, we analyzed the effect of RASSF1A on the function of YAP1. Expression of YAP1 resulted in the downregulation of several tumor suppressor genes and induction of S-phase. Co-expression with RASSF1A normalized the expression levels of these tumor suppressors and induced a G0-G1 arrest and apoptosis. This effect was associated with the reduction of MDM2 and the increase of p53. These data suggest that the tumor suppressor RASSF1A inhibits the oncogenic potential of YAP1. Additionally, we could show that ANKRD1 is a YAP1 target gene that is induced by RASSF1A. Further analysis revealed that ANKRD1 is epigenetically inactivated in human cancer. ANKRD1 expression induced the expression of TP53 as well as BAX and CDKN1A and reduced colony formation of cancer cells. We found that ANKRD1 interacts with p53 and is involved in the destabilization of MDM2. Additionally, our data indicate that the tumor-suppressive effect of ANKRD1 depends on the presence of p53. These results suggest that ANKRD1 is a tumor-suppressive downstream target of the Hippo pathway that is epigenetically silenced in human cancer.
Collapse
Affiliation(s)
- Adriana P Jiménez
- Institute for Genetics, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Annalena Traum
- Institute for Genetics, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Thomas Boettger
- Department I-Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Holger Hackstein
- Clinical Immunology, Biomedizinisches Forschungszentrum Seltersberg, D-35392 Giessen, Germany
| | - Antje M Richter
- Institute for Genetics, Justus-Liebig University Giessen, D-35392 Giessen, Germany
| | - Reinhard H Dammann
- Institute for Genetics, Justus-Liebig University Giessen, D-35392 Giessen, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, D-35392 Giessen, Germany
| |
Collapse
|
72
|
Tsai CR, Anderson AE, Burra S, Jo J, Galko MJ. Yorkie regulates epidermal wound healing in Drosophila larvae independently of cell proliferation and apoptosis. Dev Biol 2017; 427:61-71. [PMID: 28514643 DOI: 10.1016/j.ydbio.2017.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022]
Abstract
Yorkie (Yki), the transcriptional co-activator of the Hippo signaling pathway, has well-characterized roles in balancing apoptosis and cell division during organ growth control. Yki is also required in diverse tissue regenerative contexts. In most cases this requirement reflects its well-characterized roles in balancing apoptosis and cell division. Whether Yki has repair functions outside of the control of cell proliferation, death, and growth is not clear. Here we show that Yki and Scalloped (Sd) are required for epidermal wound closure in the Drosophila larval epidermis. Using a GFP-tagged Yki transgene we show that Yki transiently translocates to some epidermal nuclei upon wounding. Genetic analysis strongly suggests that Yki interacts with the known wound healing pathway, Jun N-terminal kinase (JNK), but not with Platelet Derived Growth Factor/Vascular-Endothelial Growth Factor receptor (Pvr). Yki likely acts downstream of or parallel to JNK signaling and does not appear to regulate either proliferation or apoptosis in the larval epidermis during wound repair. Analysis of actin structures after wounding suggests that Yki and Sd promote wound closure through actin regulation. In sum, we found that Yki regulates an epithelial tissue repair process independently of its previously documented roles in balancing proliferation and apoptosis.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aimee E Anderson
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sirisha Burra
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juyeon Jo
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Genes and Development Graduate Program, Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, TX 77030, USA
| | - Michael J Galko
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Genes and Development Graduate Program, Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, TX 77030, USA.
| |
Collapse
|
73
|
Tissue growth and tumorigenesis in Drosophila: cell polarity and the Hippo pathway. Curr Opin Cell Biol 2017; 48:1-9. [PMID: 28364663 DOI: 10.1016/j.ceb.2017.03.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/17/2022]
Abstract
Cell polarity regulation is critical for defining membrane domains required for the establishment and maintenance of the apical-basal axis in epithelial cells (apico-basal polarity), asymmetric cell divisions, planar organization of tissues (planar cell polarity), and the formation of the front-rear axis in cell migration (front-rear polarity). In the vinegar fly, Drosophila melanogaster, cell polarity regulators also interact with the Hippo tissue growth control signaling pathway. In this review we survey the recent Drosophila literature linking cell polarity regulators with the Hippo pathway in epithelial tissue growth, neural stem cell asymmetric divisions and in cell migration in physiological and tumorigenic settings.
Collapse
|
74
|
Differential Regulation of Cyclin E by Yorkie-Scalloped Signaling in Organ Development. G3-GENES GENOMES GENETICS 2017; 7:1049-1060. [PMID: 28143945 PMCID: PMC5345706 DOI: 10.1534/g3.117.039065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tissue integrity and homeostasis are accomplished through strict spatial and temporal regulation of cell growth and proliferation during development. Various signaling pathways have emerged as major growth regulators across metazoans; yet, how differential growth within a tissue is spatiotemporally coordinated remains largely unclear. Here, we report a role of a growth modulator Yorkie (Yki), the Drosophila homolog of Yes-associated protein (YAP), that differentially regulates its targets in Drosophila wing imaginal discs; whereby Yki interacts with its transcriptional partner, Scalloped (Sd), the homolog of the TEAD/TEF family transcription factor in mammals, to control an essential cell cycle regulator Cyclin E (CycE). Interestingly, when Yki was coexpressed with Fizzy-related (Fzr), a Drosophila endocycle inducer and homolog of Cdh1 in mammals, surrounding hinge cells displayed larger nuclear size than distal pouch cells. The observed size difference is attributable to differential regulation of CycE, a target of Yki and Sd, the latter of which can directly bind to CycE regulatory sequences, and is expressed only in the pouch region of the wing disc starting from the late second-instar larval stage. During earlier stages of larval development, when Sd expression was not detected in the wing disc, coexpression of Fzr and Yki did not cause size differences between cells along the proximal–distal axis of the disc. We show that ectopic CycE promoted cell proliferation and apoptosis, and inhibited transcriptional activity of Yki targets. These findings suggest that spatiotemporal expression of transcription factor Sd induces differential growth regulation by Yki during wing disc development, highlighting coordination between Yki and CycE to control growth and maintain homeostasis.
Collapse
|
75
|
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
|
76
|
Ito T, Igaki T. Dissecting cellular senescence and SASP in Drosophila. Inflamm Regen 2016; 36:25. [PMID: 29259698 PMCID: PMC5725765 DOI: 10.1186/s41232-016-0031-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
Cellular senescence can act as both tumor suppressor and tumor promoter depending on the cellular contexts. On one hand, premature senescence has been considered as an innate host defense mechanism against carcinogenesis in mammals. In response to various stresses including oxidative stress, DNA damage, and oncogenic stress, suffered cells undergo irreversible cell cycle arrest, leading to tumor suppression. On the other hand, recent studies in mammalian systems have revealed that senescent cells can drive oncogenesis by secreting diverse proteins such as inflammatory cytokines, matrix remodeling factors, and growth factors, the phenomenon called senescence-associated secretory phenotype (SASP). However, the mechanisms by which these contradictory effects regulate tumor growth and metastasis in vivo have been elusive. Here, we review the recent discovery of cellular senescence in Drosophila and the mechanisms underlying senescence-mediated tumor regulation dissected by Drosophila genetics.
Collapse
Affiliation(s)
- Takao Ito
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
77
|
Jia H, Song L, Cong Q, Wang J, Xu H, Chu Y, Li Q, Zhang Y, Zou X, Zhang C, Chin YE, Zhang X, Li Z, Zhu K, Wang B, Peng H, Hou Z. The LIM protein AJUBA promotes colorectal cancer cell survival through suppression of JAK1/STAT1/IFIT2 network. Oncogene 2016; 36:2655-2666. [DOI: 10.1038/onc.2016.418] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 09/12/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
|
78
|
The Hippo signalling pathway coordinates organ growth and limits developmental variability by controlling dilp8 expression. Nat Commun 2016; 7:13505. [PMID: 27874005 PMCID: PMC5121414 DOI: 10.1038/ncomms13505] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/11/2016] [Indexed: 12/16/2022] Open
Abstract
Coordination of organ growth during development is required to generate fit individuals with fixed proportions. We recently identified Drosophila Dilp8 as a key hormone in coupling organ growth with animal maturation. In addition, dilp8 mutant flies exhibit elevated fluctuating asymmetry (FA) demonstrating a function for Dilp8 in ensuring developmental stability. The signals regulating Dilp8 activity during normal development are not yet known. Here, we show that the transcriptional co-activators of the Hippo (Hpo) pathway, Yorkie (Yki, YAP/TAZ) and its DNA-binding partner Scalloped (Sd), directly regulate dilp8 expression through a Hpo-responsive element (HRE) in the dilp8 promoter. We further demonstrate that mutation of the HRE by genome-editing results in animals with increased FA, thereby mimicking full dilp8 loss of function. Therefore, our results indicate that growth coordination of organs is connected to their growth status through a feedback loop involving Hpo and Dilp8 signalling pathways. The hormone Dipl8 regulates organ growth in Drosophila whereas the Hippo pathway controls organ size. Here, the authors show that the expression of Dipl8 is regulated by the Hippo pathway, thus linking organ growth or organ size in Drosophila.
Collapse
|
79
|
Clavier A, Rincheval-Arnold A, Baillet A, Mignotte B, Guénal I. Two different specific JNK activators are required to trigger apoptosis or compensatory proliferation in response to Rbf1 in Drosophila. Cell Cycle 2016; 15:283-94. [PMID: 26825229 DOI: 10.1080/15384101.2015.1100776] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The Jun Kinase (JNK) signaling pathway responds to diverse stimuli by appropriate and specific cellular responses such as apoptosis, differentiation or proliferation. The mechanisms that mediate this specificity remain largely unknown. The core of this signaling pathway, composed of a JNK protein and a JNK kinase (JNKK), can be activated by various putative JNKK kinases (JNKKK) which are themselves downstream of different adaptor proteins. A proposed hypothesis is that the JNK pathway specific response lies in the combination of a JNKKK and an adaptor protein upstream of the JNKK. We previously showed that the Drosophila homolog of pRb (Rbf1) and a mutant form of Rbf1 (Rbf1(D253A)) have JNK-dependent pro-apoptotic properties. Rbf1(D253A) is also able to induce a JNK-dependent abnormal proliferation. Here, we show that Rbf1-induced apoptosis triggers proliferation which depends on the JNK pathway activation. Taking advantage of these phenotypes, we investigated the JNK signaling involved in either Rbf1-induced apoptosis or in proliferation in response to Rbf1-induced apoptosis. We demonstrated that 2 different JNK pathways involving different adaptor proteins and kinases are involved in Rbf1-apoptosis (i.e. Rac1-dTak1-dMekk1-JNK pathway) and in proliferation in response to Rbf1-induced apoptosis (i.e., dTRAF1-Slipper-JNK pathway). Using a transient induction of rbf1, we show that Rbf1-induced apoptosis activates a compensatory proliferation mechanism which also depends on Slipper and dTRAF1. Thus, these 2 proteins seem to be key players of compensatory proliferation in Drosophila.
Collapse
Affiliation(s)
- Amandine Clavier
- a Université de Versailles Saint-Quentin-en-Yvelines, Laboratoire de Génétique et Biologie Cellulaire , EA4589 , Montigny-le-Bretonneux ; France.,b Ecole Pratique des Hautes Etudes, Laboratoire de Génétique Moléculaire et Physiologique , Montigny-le-Bretonneux , France
| | - Aurore Rincheval-Arnold
- a Université de Versailles Saint-Quentin-en-Yvelines, Laboratoire de Génétique et Biologie Cellulaire , EA4589 , Montigny-le-Bretonneux ; France
| | - Adrienne Baillet
- a Université de Versailles Saint-Quentin-en-Yvelines, Laboratoire de Génétique et Biologie Cellulaire , EA4589 , Montigny-le-Bretonneux ; France.,b Ecole Pratique des Hautes Etudes, Laboratoire de Génétique Moléculaire et Physiologique , Montigny-le-Bretonneux , France
| | - Bernard Mignotte
- a Université de Versailles Saint-Quentin-en-Yvelines, Laboratoire de Génétique et Biologie Cellulaire , EA4589 , Montigny-le-Bretonneux ; France.,b Ecole Pratique des Hautes Etudes, Laboratoire de Génétique Moléculaire et Physiologique , Montigny-le-Bretonneux , France
| | - Isabelle Guénal
- a Université de Versailles Saint-Quentin-en-Yvelines, Laboratoire de Génétique et Biologie Cellulaire , EA4589 , Montigny-le-Bretonneux ; France
| |
Collapse
|
80
|
AJUBA LIM Proteins Limit Hippo Activity in Proliferating Cells by Sequestering the Hippo Core Kinase Complex in the Cytosol. Mol Cell Biol 2016; 36:2526-42. [PMID: 27457617 PMCID: PMC5038147 DOI: 10.1128/mcb.00136-16] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022] Open
Abstract
The Hippo pathway controls organ growth and is implicated in cancer development. Whether and how Hippo pathway activity is limited to sustain or initiate cell growth when needed is not understood. The members of the AJUBA family of LIM proteins are negative regulators of the Hippo pathway. In mammalian epithelial cells, we found that AJUBA LIM proteins limit Hippo regulation of YAP, in proliferating cells only, by sequestering a cytosolic Hippo kinase complex in which LATS kinase is inhibited. At the plasma membranes of growth-arrested cells, AJUBA LIM proteins do not inhibit or associate with the Hippo kinase complex. The ability of AJUBA LIM proteins to inhibit YAP regulation by Hippo and to associate with the kinase complex directly correlate with their capacity to limit Hippo signaling during Drosophila wing development. AJUBA LIM proteins did not influence YAP activity in response to cell-extrinsic or cell-intrinsic mechanical signals. Thus, AJUBA LIM proteins limit Hippo pathway activity in contexts where cell proliferation is needed.
Collapse
|
81
|
Wang D, Li XR, Dong DJ, Huang H, Wang JX, Zhao XF. The Steroid Hormone 20-Hydroxyecdysone Promotes the Cytoplasmic Localization of Yorkie to Suppress Cell Proliferation and Induce Apoptosis. J Biol Chem 2016; 291:21761-21770. [PMID: 27551043 DOI: 10.1074/jbc.m116.719856] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 08/18/2016] [Indexed: 01/04/2023] Open
Abstract
The transcriptional co-activator Yki (Yorkie), a member of the Hippo pathway, regulates cell proliferation or apoptosis, depending on its nuclear or cytoplasmic location. However, the upstream factors regulating the subcellular localization of Yki are unclear. We found that the steroid hormone 20-hydroxyecdysone (20E) induces phosphorylation of Yki, causing it to remain in the cytoplasm, where it promotes apoptosis in the midgut of the lepidopteran insect Helicoverpa armigera Yki is expressed in various tissues, with an increase in the epidermis and midgut during early metamorphic molting. Yki is localized mainly in the nucleus of feeding larval midgut cells but is mainly localized in the cytoplasm of metamorphic molting larval midgut cells. The knockdown of Yki in the feeding larvae promotes larval-pupal transition, midgut programmed cell death, and repressed IAP1 (inhibitor of apoptosis 1) expression. Knockdown of Yki in the epidermal cell line (HaEpi) induced increased activation of Caspase3/7. Overexpressed Yki in HaEpi cells was mainly localized in the nucleus and induced cell proliferation. 20E promotes the cytoplasmic localization of Yki, reducing the expression of the IAP1, resulting in apoptosis. 20E promotes cytoplasmic retention of Yki by increasing Yki phosphorylation levels and promoting the interaction between Yki and the adaptor protein 14-3-3-ϵ. This regulation of Yki suppresses cell proliferation and induces cell apoptosis.
Collapse
Affiliation(s)
- Di Wang
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Xiang-Ru Li
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Du-Juan Dong
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Hua Huang
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Jin-Xing Wang
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Xiao-Fan Zhao
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| |
Collapse
|
82
|
JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol Mol Biol Rev 2016; 80:793-835. [PMID: 27466283 DOI: 10.1128/mmbr.00043-14] [Citation(s) in RCA: 378] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs), as members of the mitogen-activated protein kinase (MAPK) family, mediate eukaryotic cell responses to a wide range of abiotic and biotic stress insults. JNKs also regulate important physiological processes, including neuronal functions, immunological actions, and embryonic development, via their impact on gene expression, cytoskeletal protein dynamics, and cell death/survival pathways. Although the JNK pathway has been under study for >20 years, its complexity is still perplexing, with multiple protein partners of JNKs underlying the diversity of actions. Here we review the current knowledge of JNK structure and isoforms as well as the partnerships of JNKs with a range of intracellular proteins. Many of these proteins are direct substrates of the JNKs. We analyzed almost 100 of these target proteins in detail within a framework of their classification based on their regulation by JNKs. Examples of these JNK substrates include a diverse assortment of nuclear transcription factors (Jun, ATF2, Myc, Elk1), cytoplasmic proteins involved in cytoskeleton regulation (DCX, Tau, WDR62) or vesicular transport (JIP1, JIP3), cell membrane receptors (BMPR2), and mitochondrial proteins (Mcl1, Bim). In addition, because upstream signaling components impact JNK activity, we critically assessed the involvement of signaling scaffolds and the roles of feedback mechanisms in the JNK pathway. Despite a clarification of many regulatory events in JNK-dependent signaling during the past decade, many other structural and mechanistic insights are just beginning to be revealed. These advances open new opportunities to understand the role of JNK signaling in diverse physiological and pathophysiological states.
Collapse
|
83
|
Schimizzi GV, Maher MT, Loza AJ, Longmore GD. Disruption of the Cdc42/Par6/aPKC or Dlg/Scrib/Lgl Polarity Complex Promotes Epithelial Proliferation via Overlapping Mechanisms. PLoS One 2016; 11:e0159881. [PMID: 27454609 PMCID: PMC4959776 DOI: 10.1371/journal.pone.0159881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 11/18/2022] Open
Abstract
The establishment and maintenance of apical-basal polarity is a defining characteristic and essential feature of functioning epithelia. Apical-basal polarity (ABP) proteins are also tumor suppressors that are targeted for disruption by oncogenic viruses and are commonly mutated in human carcinomas. Disruption of these ABP proteins is an early event in cancer development that results in increased proliferation and epithelial disorganization through means not fully characterized. Using the proliferating Drosophila melanogaster wing disc epithelium, we demonstrate that disruption of the junctional vs. basal polarity complexes results in increased epithelial proliferation via distinct downstream signaling pathways. Disruption of the basal polarity complex results in JNK-dependent proliferation, while disruption of the junctional complex primarily results in p38-dependent proliferation. Surprisingly, the Rho-Rok-Myosin contractility apparatus appears to play opposite roles in the regulation of the proliferative phenotype based on which polarity complex is disrupted. In contrast, non-autonomous Tumor Necrosis Factor (TNF) signaling appears to suppress the proliferation that results from apical-basal polarity disruption, regardless of which complex is disrupted. Finally we demonstrate that disruption of the junctional polarity complex activates JNK via the Rho-Rok-Myosin contractility apparatus independent of the cortical actin regulator, Moesin.
Collapse
Affiliation(s)
- Gregory V. Schimizzi
- ICCE Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Meghan T. Maher
- ICCE Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew J. Loza
- ICCE Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Computational and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gregory D. Longmore
- ICCE Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
84
|
Huang D, Li X, Sun L, Huang P, Ying H, Wang H, Wu J, Song H. Regulation of Hippo signalling by p38 signalling. J Mol Cell Biol 2016; 8:328-37. [PMID: 27402810 PMCID: PMC4991669 DOI: 10.1093/jmcb/mjw036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/05/2016] [Indexed: 11/17/2022] Open
Abstract
The Hippo signalling pathway has a crucial role in growth control during development, and its dysregulation contributes to tumorigenesis. Recent studies uncover multiple upstream regulatory inputs into Hippo signalling, which affects phosphorylation of the transcriptional coactivator Yki/YAP/TAZ by Wts/Lats. Here we identify the p38 mitogen-activated protein kinase (MAPK) pathway as a new upstream branch of the Hippo pathway. In Drosophila, overexpression of MAPKK gene licorne (lic), or MAPKKK gene Mekk1, promotes Yki activity and induces Hippo target gene expression. Loss-of-function studies show that lic regulates Hippo signalling in ovary follicle cells and in the wing disc. Epistasis analysis indicates that Mekk1 and lic affect Hippo signalling via p38b and wts. We further demonstrate that the Mekk1-Lic-p38b cascade inhibits Hippo signalling by promoting F-actin accumulation and Jub phosphorylation. In addition, p38 signalling modulates actin filaments and Hippo signalling in parallel to small GTPases Ras, Rac1, and Rho1. Lastly, we show that p38 signalling regulates Hippo signalling in mammalian cell lines. The Lic homologue MKK3 promotes nuclear localization of YAP via the actin cytoskeleton. Upregulation or downregulation of the p38 pathway regulates YAP-mediated transcription. Our work thus reveals a conserved crosstalk between the p38 MAPK pathway and the Hippo pathway in growth regulation.
Collapse
Affiliation(s)
- Dashun Huang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science & Technology of China, 96 Jin Zhai Road, Hefei 230031, China Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| | - Xiaojiao Li
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| | - Li Sun
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| | - Ping Huang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| | - Hao Ying
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| | - Hui Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| | - Jiarui Wu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science & Technology of China, 96 Jin Zhai Road, Hefei 230031, China
| | - Haiyun Song
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China Key Laboratory of Food Safety Risk Assessment, Ministry of Health, 37 Guang Qu Road, Beijing 100021, China
| |
Collapse
|
85
|
Zhang T, Sheng Z, Du W. Loss of histone deacetylase HDAC1 induces cell death in Drosophila epithelial cells through JNK and Hippo signaling. Mech Dev 2016; 141:4-13. [PMID: 27378074 DOI: 10.1016/j.mod.2016.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/15/2016] [Accepted: 07/01/2016] [Indexed: 12/18/2022]
Abstract
Inactivation of HDAC1 and its homolog HDAC2 or addition of HDAC inhibitors in mammalian systems induces apoptosis, cell cycle arrest, and developmental defects. Although these phenotypes have been extensively characterized, the precise underlying mechanisms remain unclear, particularly in in vivo settings. In this study, we show that inactivation of Rpd3, the only HDAC1 and HDAC2 ortholog in Drosophila, induced apoptosis and clone elimination in the developing eye and wing imaginal discs. Depletion of Rpd3 by RNAi cell-autonomously increased JNK activities and decreased activities of Yki, the nuclear effecter of Hippo signaling pathway. In addition, inhibition of JNK activities largely rescued Rpd3 RNAi-induced apoptosis, but did not affect its inhibition of Yki activities. Conversely, increasing the Yki activities largely rescued Rpd3 RNAi-induced apoptosis, but did not affect its induction of JNK activities. Furthermore, inactivation of Mi-2, a core component of the Rpd3-containing NuRD complex strongly induced JNK activities; while inactivation of Sin3A, a key component of the Rpd3-containing Sin3 complex, significantly inhibited Yki activities. Taken together, these results reveal that inactivation of Rpd3 independently regulates JNK and Yki activities and that both Hippo and JNK signaling pathways contribute to Rpd3 RNAi-induced apoptosis.
Collapse
Affiliation(s)
- Tianyi Zhang
- Ben May Department for Cancer Research, The University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA
| | - Zhentao Sheng
- Ben May Department for Cancer Research, The University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA
| | - Wei Du
- Ben May Department for Cancer Research, The University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
86
|
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
|
87
|
Smith-Bolton R. Drosophila Imaginal Discs as a Model of Epithelial Wound Repair and Regeneration. Adv Wound Care (New Rochelle) 2016; 5:251-261. [PMID: 27274435 DOI: 10.1089/wound.2014.0547] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Significance: The Drosophila larval imaginal discs, which form the adult fly during metamorphosis, are an established model system for the study of epithelial tissue damage. The disc proper is a simple columnar epithelium, but it contains complex patterning and cell-fate specification, and is genetically tractable. These features enable unbiased genetic screens to identify genes involved in all aspects of the wound response, from sensing damage to wound closure, initiation of regeneration, and re-establishment of proper cell fates. Identification of the genes that facilitate epithelial wound closure and regeneration will enable development of more sophisticated wound treatments for clinical use. Recent Advances: Imaginal disc epithelia can be damaged in many different ways, including fragmentation, induction of cell death, and irradiation. Recent work has demonstrated that the tissue's response to damage varies depending on how the wound was induced. Here, we summarize the different responses activated in these epithelial tissues after the different types of damage. Critical Issues: These studies highlight that not all wounds elicit the same response from the surrounding tissue. A complete understanding of the various wound-healing mechanisms in Drosophila will be a first step in understanding how to manage damaged human tissues and optimize healing in different clinical contexts. Future Directions: Further work is necessary to understand the similarities and differences among an epithelial tissue's responses to different insults. Ongoing studies will identify the genes and pathways employed by injured imaginal discs. Thus, work in this genetically tractable system complements work in more conventional wound-healing models.
Collapse
Affiliation(s)
- Rachel Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
88
|
Abstract
The Hippo pathway is a signalling cascade conserved from Drosophila melanogaster to mammals. The mammalian core kinase components comprise MST1 and MST2, SAV1, LATS1 and LATS2 and MOB1A and MOB1B. The transcriptional co-activators YAP1 and TAZ are the downstream effectors of the Hippo pathway and regulate target gene expression. Hippo signalling has crucial roles in the control of organ size, tissue homeostasis and regeneration, and dysregulation of the Hippo pathway can lead to uncontrolled cell growth and malignant transformation. Mammalian intestine consists of a stem cell compartment as well as differentiated cells, and its ability to regenerate rapidly after injury makes it an excellent model system to study tissue homeostasis, regeneration and tumorigenesis. Several studies have established the important role of the Hippo pathway in these processes. In addition, crosstalk between Hippo and other signalling pathways provides tight, yet versatile, regulation of tissue homeostasis. In this Review, we summarize studies on the role of the Hippo pathway in the intestine on these physiological processes and the underlying mechanisms responsible, and discuss future research directions and potential therapeutic strategies targeting Hippo signalling in intestinal disease.
Collapse
|
89
|
The TIR/BB-loop mimetic AS-1 attenuates mechanical stress-induced cardiac fibroblast activation and paracrine secretion via modulation of large tumor suppressor kinase 1. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1191-202. [DOI: 10.1016/j.bbadis.2016.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/15/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
|
90
|
Chen X, Stauffer S, Chen Y, Dong J. Ajuba Phosphorylation by CDK1 Promotes Cell Proliferation and Tumorigenesis. J Biol Chem 2016; 291:14761-72. [PMID: 27226586 DOI: 10.1074/jbc.m116.722751] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Indexed: 12/21/2022] Open
Abstract
Recent studies identified the adaptor protein Ajuba as a positive regulator of Yes-associated protein (YAP) oncogenic activity through inhibiting large tumor suppressor (Lats1/2) core kinases of the Hippo pathway, a signaling pathway that plays important roles in cancer. In this study, we define a novel mechanism for phospho-regulation of Ajuba in mitosis and its biological significance in cancer. We found that Ajuba is phosphorylated in vitro and in vivo by cyclin-dependent kinase 1 (CDK1) at Ser(119) and Ser(175) during the G2/M phase of the cell cycle. Mitotic phosphorylation of Ajuba controls the expression of multiple cell cycle regulators; however, it does not affect Hippo signaling activity, nor does it induce epithelial-mesenchymal transition. We further showed that mitotic phosphorylation of Ajuba is sufficient to promote cell proliferation and anchorage-independent growth in vitro and tumorigenesis in vivo Collectively, our discoveries reveal a previously unrecognized mechanism for Ajuba regulation in mitosis and its role in tumorigenesis.
Collapse
Affiliation(s)
- Xingcheng Chen
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Seth Stauffer
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Yuanhong Chen
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and
| | - Jixin Dong
- From the Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center and
| |
Collapse
|
91
|
Elbediwy A, Vincent-Mistiaen ZI, Thompson BJ. YAP and TAZ in epithelial stem cells: A sensor for cell polarity, mechanical forces and tissue damage. Bioessays 2016; 38:644-53. [PMID: 27173018 PMCID: PMC5031209 DOI: 10.1002/bies.201600037] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The YAP/TAZ family of transcriptional co‐activators drives cell proliferation in epithelial tissues and cancers. Yet, how YAP and TAZ are physiologically regulated remains unclear. Here we review recent reports that YAP and TAZ act primarily as sensors of epithelial cell polarity, being inhibited when cells differentiate an apical membrane domain, and being activated when cells contact the extracellular matrix via their basal membrane domain. Apical signalling occurs via the canonical Crumbs/CRB‐Hippo/MST‐Warts/LATS kinase cascade to phosphorylate and inhibit YAP/TAZ. Basal signalling occurs via Integrins and Src family kinases to phosphorylate and activate YAP/TAZ. Thus, YAP/TAZ is localised to the nucleus in basal stem/progenitor cells and cytoplasm in differentiated squamous cells or columnar cells. In addition, other signals such as mechanical forces, tissue damage and possibly receptor tyrosine kinases (RTKs) can influence MST‐LATS or Src family kinase activity to modulate YAP/TAZ activity.
Collapse
Affiliation(s)
- Ahmed Elbediwy
- Epithelial Biology Laboratory, Francis Crick Institute, London, UK
| | | | - Barry J Thompson
- Epithelial Biology Laboratory, Francis Crick Institute, London, UK
| |
Collapse
|
92
|
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
|
93
|
Han MKL, de Rooij J. Converging and Unique Mechanisms of Mechanotransduction at Adhesion Sites. Trends Cell Biol 2016; 26:612-623. [PMID: 27036655 DOI: 10.1016/j.tcb.2016.03.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 12/31/2022]
Abstract
The molecular mechanisms by which physical forces control tissue development are beginning to be elucidated. Sites of adhesion between both cells and the extracellular environment [extracellular matrix (ECM) or neighboring cells] contain protein complexes capable of sensing fluctuations in tensile forces. Tension-dependent changes in the dynamics and composition of these complexes mark the transformation of physical input into biochemical signals that defines mechanotransduction. It is becoming apparent that, although the core constituents of these different adhesions are distinct, principles and proteins involved in mechanotransduction are conserved. Here, we discuss the current knowledge of overlapping and distinct aspects of mechanotransduction between integrin and cadherin adhesion complexes.
Collapse
Affiliation(s)
- Mitchell K L Han
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Stratenum 3.231, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Johan de Rooij
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Stratenum 3.231, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
94
|
Willsey HR, Zheng X, Carlos Pastor-Pareja J, Willsey AJ, Beachy PA, Xu T. Localized JNK signaling regulates organ size during development. eLife 2016; 5. [PMID: 26974344 PMCID: PMC4848088 DOI: 10.7554/elife.11491] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/12/2016] [Indexed: 11/17/2022] Open
Abstract
A fundamental question of biology is what determines organ size. Despite demonstrations that factors within organs determine their sizes, intrinsic size control mechanisms remain elusive. Here we show that Drosophila wing size is regulated by JNK signaling during development. JNK is active in a stripe along the center of developing wings, and modulating JNK signaling within this stripe changes organ size. This JNK stripe influences proliferation in a non-canonical, Jun-independent manner by inhibiting the Hippo pathway. Localized JNK activity is established by Hedgehog signaling, where Ci elevates dTRAF1 expression. As the dTRAF1 homolog, TRAF4, is amplified in numerous cancers, these findings provide a new mechanism for how the Hedgehog pathway could contribute to tumorigenesis, and, more importantly, provides a new strategy for cancer therapies. Finally, modulation of JNK signaling centers in developing antennae and legs changes their sizes, suggesting a more generalizable role for JNK signaling in developmental organ size control. DOI:http://dx.doi.org/10.7554/eLife.11491.001 A key challenge in biology is to understand what determines size. As an animal grows, signals are produced that control the size of its organs. Many of the signaling pathways that regulate size during normal animal development also contribute to the formation of tumors. Therefore, it is important to find out exactly how the signaling molecules that regulate size are linked to those that regulate tumor growth. A protein called JNK activates a signaling pathway that triggers tumor growth. JNK signaling also stimulates cells to multiply in tissues that need repair, but it is not known whether it also regulates the size of organs during animal development. Here, Willsey et al. investigate whether JNK is active in the developing wings of fruit flies, which are commonly used as models of animal development. The experiments show that JNK is active in a stripe across the developing wing and is required for the wing to grow to its proper size. A master signal protein called Hedgehog is responsible for establishing this stripe of JNK activity. Unexpectedly, rather than acting through its usual signaling pathway, JNK activates the Hippo pathway in the wing to control organ size during development. Willsey et al.’s findings highlight potential new targets for cancer therapies. A future challenge will be to find out whether small patches of JNK signaling are found in the developing organs of other animals, and whether they can help explain how size changes between species. DOI:http://dx.doi.org/10.7554/eLife.11491.002
Collapse
Affiliation(s)
- Helen Rankin Willsey
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Xiaoyan Zheng
- Departments of Biochemistry and Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, United States
| | - José Carlos Pastor-Pareja
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - A Jeremy Willsey
- Department of Psychiatry, University of California, San Francisco, San Francisco, United States
| | - Philip A Beachy
- Departments of Biochemistry and Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, United States
| | - Tian Xu
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States.,State Key Laboratory of Genetic Engineering and National Center for International Research, Fudan-Yale Biomedical Research Center, Institute of Developmental Biology and Molecular Medicine, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
95
|
Losick VP, Jun AS, Spradling AC. Wound-Induced Polyploidization: Regulation by Hippo and JNK Signaling and Conservation in Mammals. PLoS One 2016; 11:e0151251. [PMID: 26958853 PMCID: PMC4784922 DOI: 10.1371/journal.pone.0151251] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022] Open
Abstract
Tissue integrity and homeostasis often rely on the proliferation of stem cells or differentiated cells to replace lost, aged, or damaged cells. Recently, we described an alternative source of cell replacement- the expansion of resident, non-dividing diploid cells by wound-induced polyploidization (WIP). Here we show that the magnitude of WIP is proportional to the extent of cell loss using a new semi-automated assay with single cell resolution. Hippo and JNK signaling regulate WIP; unexpectedly however, JNK signaling through AP-1 limits rather than stimulates the level of Yki activation and polyploidization in the Drosophila epidermis. We found that polyploidization also quantitatively compensates for cell loss in a mammalian tissue, mouse corneal endothelium, where increased cell death occurs with age in a mouse model of Fuchs Endothelial Corneal Dystrophy (FECD). Our results suggest that WIP is an evolutionarily conserved homeostatic mechanism that maintains the size and synthetic capacity of adult tissues.
Collapse
Affiliation(s)
- Vicki P. Losick
- Department of Embryology, Carnegie Institution for Science, Howard Hughes Medical Institute, 3250 San Martin Dr., Baltimore, MD 21218, United States of America
| | - Albert S. Jun
- Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 N. Broadway, Baltimore, MD 21231, United States of America
| | - Allan C. Spradling
- Department of Embryology, Carnegie Institution for Science, Howard Hughes Medical Institute, 3250 San Martin Dr., Baltimore, MD 21218, United States of America
- * E-mail:
| |
Collapse
|
96
|
Pérez-Garijo A, Steller H. Spreading the word: non-autonomous effects of apoptosis during development, regeneration and disease. Development 2016; 142:3253-62. [PMID: 26443630 DOI: 10.1242/dev.127878] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apoptosis, in contrast to other forms of cell death such as necrosis, was originally regarded as a 'silent' mechanism of cell elimination designed to degrade the contents of doomed cells. However, during the past decade it has become clear that apoptotic cells can produce diverse signals that have a profound impact on neighboring cells and tissues. For example, apoptotic cells can release factors that influence the proliferation and survival of adjacent tissues. Apoptosis can also affect tissue movement and morphogenesis by modifying tissue tension in surrounding cells. As we review here, these findings reveal unexpected roles for apoptosis in tissue remodeling during development, as well as in regeneration and cancer.
Collapse
Affiliation(s)
- Ainhoa Pérez-Garijo
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Hermann Steller
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
97
|
Liu X, Li H, Rajurkar M, Li Q, Cotton JL, Ou J, Zhu LJ, Goel HL, Mercurio AM, Park JS, Davis RJ, Mao J. Tead and AP1 Coordinate Transcription and Motility. Cell Rep 2016; 14:1169-1180. [PMID: 26832411 DOI: 10.1016/j.celrep.2015.12.104] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 11/11/2015] [Accepted: 12/23/2015] [Indexed: 11/25/2022] Open
Abstract
The Tead family transcription factors are the major intracellular mediators of the Hippo-Yap pathway. Despite the importance of Hippo signaling in tumorigenesis, Tead-dependent downstream oncogenic programs and target genes in cancer cells remain poorly understood. Here, we characterize Tead4-mediated transcriptional networks in a diverse range of cancer cells, including neuroblastoma, colorectal, lung, and endometrial carcinomas. By intersecting genome-wide chromatin occupancy analyses of Tead4, JunD, and Fra1/2, we find that Tead4 cooperates with AP1 transcription factors to coordinate target gene transcription. We find that Tead-AP1 interaction is JNK independent but engages the SRC1-3 co-activators to promote downstream transcription. Furthermore, we show that Tead-AP1 cooperation regulates the activity of the Dock-Rac/CDC42 module and drives the expression of a unique core set of target genes, thereby directing cell migration and invasion. Together, our data unveil a critical regulatory mechanism underlying Tead- and AP1-controlled transcriptional and functional outputs in cancer cells.
Collapse
Affiliation(s)
- Xiangfan Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huapeng Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Mihir Rajurkar
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Qi Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jennifer L Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jianhong Ou
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lihua J Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hira L Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Joo-Seop Park
- Divisions of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
98
|
La Fortezza M, Schenk M, Cosolo A, Kolybaba A, Grass I, Classen AK. JAK/STAT signalling mediates cell survival in response to tissue stress. Development 2016; 143:2907-19. [DOI: 10.1242/dev.132340] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Tissue homeostasis relies on the ability of tissues to respond to stress. Tissue regeneration and tumour models in Drosophila have shown that JNK is a prominent stress-response pathway promoting injury-induced apoptosis and compensatory proliferation. A central question remaining unanswered is how both responses are balanced by activation of a single pathway. JAK/STAT signalling, a potential JNK target, is implicated in promoting compensatory proliferation. While we observe JAK/STAT activation in imaginal discs upon damage, our data demonstrates that JAK/STAT and its downstream effector Zfh2 promote survival of JNK-signalling cells instead. The JNK component fos and the pro-apoptotic gene hid are regulated in a JAK/STAT-dependent manner. This molecular pathway restrains JNK-induced apoptosis and spatial propagation of JNK-signalling, thereby limiting the extent of tissue damage, as well as facilitating systemic and proliferative responses to injury. We find that the pro-survival function of JAK/STAT also drives tumour growth under conditions of chronic stress. Our study defines JAK/STAT function in tissue stress and illustrates how crosstalk between conserved signalling pathways establishes an intricate equilibrium between proliferation, apoptosis and survival to restore tissue homeostasis.
Collapse
Affiliation(s)
- Marco La Fortezza
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Madlin Schenk
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Andrea Cosolo
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Addie Kolybaba
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Isabelle Grass
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Anne-Kathrin Classen
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
99
|
Abstract
The mammalian MST kinase family, which is related to the Hippo kinase in Drosophila melanogaster, includes five related proteins: MST1 (also called STK4), MST2 (also called STK3), MST3 (also called STK24), MST4, and YSK1 (also called STK25 or SOK1). MST kinases are emerging as key signaling molecules that influence cell proliferation, organ size, cell migration, and cell polarity. Here we review the regulation and function of these kinases in normal physiology and pathologies, including cancer, endothelial malformations, and autoimmune disease.
Collapse
Affiliation(s)
| | - Erik Sahai
- The Francis Crick Institute, London WC2A 3LY, England, UK
| |
Collapse
|
100
|
Kremer KN, Dudakovic A, Hess AD, Smith BD, Karp JE, Kaufmann SH, Westendorf JJ, van Wijnen AJ, Hedin KE. Histone Deacetylase Inhibitors Target the Leukemic Microenvironment by Enhancing a Nherf1-Protein Phosphatase 1α-TAZ Signaling Pathway in Osteoblasts. J Biol Chem 2015; 290:29478-92. [PMID: 26491017 DOI: 10.1074/jbc.m115.668160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Indexed: 01/20/2023] Open
Abstract
Disrupting the protective signals provided by the bone marrow microenvironment will be critical for more effective combination drug therapies for acute myeloid leukemia (AML). Cells of the osteoblast lineage that reside in the endosteal niche have been implicated in promoting survival of AML cells. Here, we investigated how to prevent this protective interaction. We previously showed that SDF-1, a chemokine abundant in the bone marrow, induces apoptosis of AML cells, unless the leukemic cells receive protective signals provided by differentiating osteoblasts (8, 10). We now identify a novel signaling pathway in differentiating osteoblasts that can be manipulated to disrupt the osteoblast-mediated protection of AML cells. Treating differentiating osteoblasts with histone deacetylase inhibitors (HDACi) abrogated their ability to protect co-cultured AML cells from SDF-1-induced apoptosis. HDACi prominently up-regulated expression of the Nherf1 scaffold protein, which played a major role in preventing osteoblast-mediated protection of AML cells. Protein phosphatase-1α (PP1α) was identified as a novel Nherf1 interacting protein that acts as the downstream mediator of this response by promoting nuclear localization of the TAZ transcriptional modulator. Moreover, independent activation of either PP1α or TAZ was sufficient to prevent osteoblast-mediated protection of AML cells even in the absence of HDACi. Together, these results indicate that HDACi target the AML microenvironment by enhancing activation of the Nherf1-PP1α-TAZ pathway in osteoblasts. Selective drug targeting of this osteoblast signaling pathway may improve treatments of AML by rendering leukemic cells in the bone marrow more susceptible to apoptosis.
Collapse
Affiliation(s)
| | | | - Allan D Hess
- the Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland 21287
| | - B Douglas Smith
- the Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland 21287
| | - Judith E Karp
- the Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland 21287
| | - Scott H Kaufmann
- Oncology and Molecular Pharmacology & Experimental Therapeutics and
| | - Jennifer J Westendorf
- Orthopedic Surgery, the Center of Regenerative Medicine, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota 55905 and
| | - Andre J van Wijnen
- Orthopedic Surgery, the Center of Regenerative Medicine, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota 55905 and
| | | |
Collapse
|