301
|
Wang P, Chen Y, Li C, Zhao R, Wang F, Lin X, Cao L, Li S, Hu L, Gao Y, Li Y, Wu S. Drosophila eye developmental defect caused by elevation of the activity of the LIM-homeodomain protein, Lmx1a, requires its association with the Co-activator Chip. Biochem Biophys Res Commun 2015; 470:29-34. [PMID: 26718403 DOI: 10.1016/j.bbrc.2015.12.089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/20/2015] [Indexed: 11/18/2022]
Abstract
The LIM-homeodomain (LIM-HD) family member Lmx1a has been successfully used to induce dopaminergic neurons from other cell types, thus showing significant implications in replacement therapies of Parkinson's disease, but the underlying mechanism remains elusive. In this study, we used Drosophila eye as a model system to investigate how forced expression of dLmx1a, the fly homolog of human Lmx1a, alters cell identify. We found that ectopic expression of dLmx1a suppresses the formation of Drosophila eye tissue and identified the LIM and HD as two essential domains. dLmx1a requires and physically binds to Chip, a well-known cofactor of LIM-HD proteins. Chip connects two dLmx1a proteins to form a functional tetrameric complex. In addition, we provide evidence showing that dLmx1a expression results in the suppression of two retina determination gene eyes absent (eya) and string (stg). Taken together, our findings identified Chip as a novel partner of dLmx1a to alter cell differentiation in Drosophila eye through repressing eya and stg expression, and provide an animal model for further understanding the molecular mechanism whereby Lmx1a determines cell fate.
Collapse
Affiliation(s)
- Ping Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yan Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Chaojie Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Runan Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Feng Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Xiaohui Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Lei Cao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Shanshan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Liangchang Hu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yang Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yuanpei Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Shian Wu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
302
|
Yimlamai D, Fowl BH, Camargo FD. Emerging evidence on the role of the Hippo/YAP pathway in liver physiology and cancer. J Hepatol 2015; 63:1491-501. [PMID: 26226451 PMCID: PMC4654680 DOI: 10.1016/j.jhep.2015.07.008] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/30/2015] [Accepted: 07/08/2015] [Indexed: 01/11/2023]
Abstract
The Hippo pathway and its regulatory target, YAP, has recently emerged as an important biochemical signaling pathway that tightly governs epithelial tissue growth. Initially defined in Drosophilia, this pathway has shown remarkable conservation in vertebrate systems with many components of the Hippo/YAP pathway showing biochemical and functional conservation. The liver is particularly sensitive to changes in Hippo/YAP signaling with rapid increases in liver size becoming manifest on the order of days to weeks after perturbation. The first identified direct targets of Hippo/YAP signaling were pro-proliferative and anti-apoptotic gene programs, but recent work has now implicated this pathway in cell fate choice, stem cell maintenance/renewal, epithelial to mesenchymal transition, and oncogenesis. The mechanisms by which Hippo/YAP signaling is changed endogenously are beginning to come to light as well as how this pathway interacts with other signaling pathways, and important details for designing new therapeutic interventions. This review focuses on the known roles for Hippo/YAP signaling in the liver and promising avenues for future study.
Collapse
Affiliation(s)
- Dean Yimlamai
- The Stem Cell Program, Department of Medicine, Boston Children's Hospital, Boston, MA 02115, United States; Division of Gastroenterology and Nutrition, Department of Medicine, Boston Children's Hospital, Boston, MA 02115, United States.
| | - Brendan H Fowl
- The Stem Cell Program, Department of Medicine, Boston Children's Hospital, Boston, MA 02115, United States; Division of Gastroenterology and Nutrition, Department of Medicine, Boston Children's Hospital, Boston, MA 02115, United States
| | - Fernando D Camargo
- The Stem Cell Program, Department of Medicine, Boston Children's Hospital, Boston, MA 02115, United States; Harvard Stem Cell Institute, Cambridge, MA 02138, United States; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, United States.
| |
Collapse
|
303
|
Li S, Cho YS, Yue T, Ip YT, Jiang J. Overlapping functions of the MAP4K family kinases Hppy and Msn in Hippo signaling. Cell Discov 2015; 1:15038. [PMID: 27462435 PMCID: PMC4860773 DOI: 10.1038/celldisc.2015.38] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/16/2015] [Indexed: 01/26/2023] Open
Abstract
The Hippo (Hpo) tumor suppressor pathway is an evolutionarily conserved signaling pathway that controls tissue growth and organ size in species ranging from Drosophila to human, and its malfunction has been implicated in many types of human cancer. In this study, we conducted a kinome screen and identified Happyhour (Hppy)/MAP4K3 as a novel player in the Hpo pathway. Our biochemical study showed that Hppy binds and phosphorylates Wts. Our genetic experiments suggest that Hppy acts in parallel and partial redundantly with Misshapen (Msn)/MAP4K4 to regulate Yki nuclear localization and Hpo target gene expression in Drosophila wing imaginal discs. Furthermore, we showed that cytoskeleton stress restricts Yki nuclear localization through Hppy and Msn when Hpo activity is compromised, thus providing an explanation for the Wts-dependent but Hpo-independent regulation of Yki in certain contexts. Our study has unraveled an additional layer of complexity in the Hpo signaling pathway and laid down a foundation for exploring how different upstream regulators feed into the core Hpo pathway.
Collapse
Affiliation(s)
- Shuangxi Li
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA
| | - Yong Suk Cho
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA
| | - Tao Yue
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA; Center for the genetics and Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Y Tony Ip
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, MA, USA
| | - Jin Jiang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallars, TX, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallars, TX, USA
| |
Collapse
|
304
|
Hirabayashi S, Cagan RL. Salt-inducible kinases mediate nutrient-sensing to link dietary sugar and tumorigenesis in Drosophila. eLife 2015; 4:e08501. [PMID: 26573956 PMCID: PMC4643014 DOI: 10.7554/elife.08501] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/05/2015] [Indexed: 12/26/2022] Open
Abstract
Cancer cells demand excessive nutrients to support their proliferation but how cancer cells sense and promote growth in the nutrient favorable conditions remain incompletely understood. Epidemiological studies have indicated that obesity is a risk factor for various types of cancers. Feeding Drosophila a high dietary sugar was previously demonstrated to not only direct metabolic defects including obesity and organismal insulin resistance, but also transform Ras/Src-activated cells into aggressive tumors. Here we demonstrate that Ras/Src-activated cells are sensitive to perturbations in the Hippo signaling pathway. We provide evidence that nutritional cues activate Salt-inducible kinase, leading to Hippo pathway downregulation in Ras/Src-activated cells. The result is Yorkie-dependent increase in Wingless signaling, a key mediator that promotes diet-enhanced Ras/Src-tumorigenesis in an otherwise insulin-resistant environment. Through this mechanism, Ras/Src-activated cells are positioned to efficiently respond to nutritional signals and ensure tumor growth upon nutrient rich condition including obesity. DOI:http://dx.doi.org/10.7554/eLife.08501.001 Around the world, obesity has become a much more common condition. It is a serious health concern, which can increase a person's risk of developing type 2 diabetes, heart disease and certain types of cancer. People who develop type 2 diabetes become insensitive to a hormone called insulin. This hormone normally helps the body to process sugar, and so insensitivity to insulin causes excess sugar to build up in the blood. The excess sugar may provide the extra nutrients cancer cells need to grow. In 2013, researchers fed a high sugar diet to fruit flies that had been genetically engineered to develop eye tumors to study how obesity caused by a high sugar diet affects tumor growth. The high sugar diet caused the tumors to grow more aggressively. This happened because normal cells became insensitive to insulin, but the tumor cells didn't. This allowed the tumor cells to use the extra sugar to fuel their growth. The experiments showed that the tumor cells had more insulin receptors than normal cells because a molecular switch that controls the receptors was turned on. But it wasn't exactly clear how the cancer genes and excess sugar flipped that switch. Now, Hirabayashi and Cagan—who were both involved in the 2013 work—show that together cancer genes and excess sugar turn on a protein in the flies that senses sugar. This protein, called Salt-inducible kinase, blocks a cellular mechanism that normally limits the growth of cells. With this check on cellular growth blocked, the molecular switch that boosts the number of insulin receptor turns on. This in turn allows the excess sugar to fuel rapid growth of the tumor. In this way, tumor cells know when the sugars are available and make sure they grow in a nutrient-rich condition such as obesity. In the future, scientists may use this new information to develop treatments that help stop the growth of obesity-linked tumors. But first it must be confirmed whether excess sugar and cancer genes behave the same way in humans. DOI:http://dx.doi.org/10.7554/eLife.08501.002
Collapse
Affiliation(s)
- Susumu Hirabayashi
- Metabolism and Cell Growth Group, MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Ross L Cagan
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
305
|
Keder A, Rives-Quinto N, Aerne BL, Franco M, Tapon N, Carmena A. The hippo pathway core cassette regulates asymmetric cell division. Curr Biol 2015; 25:2739-2750. [PMID: 26592338 DOI: 10.1016/j.cub.2015.08.064] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/07/2015] [Accepted: 08/28/2015] [Indexed: 11/23/2022]
Abstract
Asymmetric cell division (ACD) is a crucial process during development, homeostasis, and cancer. Stem and progenitor cells divide asymmetrically, giving rise to two daughter cells, one of which retains the parent cell self-renewal capacity, while the other is committed to differentiation. Any imbalance in this process can induce overgrowth or even a cancer-like state. Here, we show that core components of the Hippo signaling pathway, an evolutionarily conserved organ growth regulator, modulate ACD in Drosophila. Hippo pathway inactivation disrupts the asymmetric localization of ACD regulators, leading to aberrant mitotic spindle orientation and defects in the generation of unequal-sized daughter cells. The Hippo pathway downstream kinase Warts, LATS1-2 in mammals, associates with the ACD modulators Inscuteable and Bazooka in vivo and phosphorylates Canoe, the ortholog of Afadin/AF-6, in vitro. Moreover, phosphosite mutant Canoe protein fails to form apical crescents in dividing neuroblasts in vivo, and the lack of Canoe phosphorylation by Warts leads to failures of Discs Large apical localization in metaphase neuroblasts. Given the relevance of ACD in stem cells during tissue homeostasis, and the well-documented role of the Hippo pathway as a tumor suppressor, these results represent a potential route for perturbations in the Hippo signaling to induce tumorigenesis via aberrant stem cell divisions.
Collapse
Affiliation(s)
- Alyona Keder
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Noemí Rives-Quinto
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Birgit L Aerne
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Maribel Franco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Nicolas Tapon
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Ana Carmena
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
306
|
Scaling the Drosophila Wing: TOR-Dependent Target Gene Access by the Hippo Pathway Transducer Yorkie. PLoS Biol 2015; 13:e1002274. [PMID: 26474042 PMCID: PMC4608745 DOI: 10.1371/journal.pbio.1002274] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/08/2015] [Indexed: 12/19/2022] Open
Abstract
Organ growth is controlled by patterning signals that operate locally (e.g., Wingless/Ints [Wnts], Bone Morphogenetic Proteins [BMPs], and Hedgehogs [Hhs]) and scaled by nutrient-dependent signals that act systemically (e.g., Insulin-like peptides [ILPs] transduced by the Target of Rapamycin [TOR] pathway). How cells integrate these distinct inputs to generate organs of the appropriate size and shape is largely unknown. The transcriptional coactivator Yorkie (Yki, a YES-Associated Protein, or YAP) acts downstream of patterning morphogens and other tissue-intrinsic signals to promote organ growth. Yki activity is regulated primarily by the Warts/Hippo (Wts/Hpo) tumour suppressor pathway, which impedes nuclear access of Yki by a cytoplasmic tethering mechanism. Here, we show that the TOR pathway regulates Yki by a separate and novel mechanism in the Drosophila wing. Instead of controlling Yki nuclear access, TOR signaling governs Yki action after it reaches the nucleus by allowing it to gain access to its target genes. When TOR activity is inhibited, Yki accumulates in the nucleus but is sequestered from its normal growth-promoting target genes—a phenomenon we term “nuclear seclusion.” Hence, we posit that in addition to its well-known role in stimulating cellular metabolism in response to nutrients, TOR also promotes wing growth by liberating Yki from nuclear seclusion, a parallel pathway that we propose contributes to the scaling of wing size with nutrient availability. From dwarves to giants, scaling is a universal property of animal organs, but its mechanistic basis is poorly understood. Here, the authors identify a molecular circuit underlying scaling of the Drosophila wing. What mechanisms control the sizes of animal organs? It is known that organ growth is the product of two systems: an intrinsic system that coordinates cell proliferation with the specification of cell fate (patterning), and an extrinsic system that synchronizes growth with nutrient levels. Developing organs integrate these two inputs to ensure that properly proportioned structures develop which are of the right scale to match overall body size. However, the mechanisms used to integrate these distinct growth control systems have remained largely mysterious. In this study, we have addressed how intrinsic and extrinsic systems combine to drive growth of the Drosophila wing. Focusing on the Target of Rapamycin (TOR) pathway—a major, nutrient-dependent regulator of organ growth—and Yorkie—the transcriptional activator downstream of the Hippo pathway and a key, organ-intrinsic growth regulator—we have identified a circuit in which TOR activity limits Yorkie’s capacity to promote wing growth, in part through a novel mode of transcription factor regulation that we term “nuclear seclusion.” We find that inhibiting TOR leads to the retention of Yorkie in the nucleus but diminishes its transcriptional activity by diverting it away from target genes. We posit that subjugating Yorkie in this way contributes to how fluctuations in TOR activity scale wing size according to nutrient levels.
Collapse
|
307
|
Zhang H, Ramakrishnan SK, Triner D, Centofanti B, Maitra D, Győrffy B, Sebolt-Leopold JS, Dame MK, Varani J, Brenner DE, Fearon ER, Omary MB, Shah YM. Tumor-selective proteotoxicity of verteporfin inhibits colon cancer progression independently of YAP1. Sci Signal 2015; 8:ra98. [PMID: 26443705 DOI: 10.1126/scisignal.aac5418] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Yes-associated protein 1 (YAP1) is a transcriptional coactivator in the Hippo signaling pathway. Increased YAP1 activity promotes the growth of tumors, including that of colorectal cancer (CRC). Verteporfin, a drug that enhances phototherapy to treat neovascular macular degeneration, is an inhibitor of YAP1. We found that verteporfin inhibited tumor growth independently of its effects on YAP1 or the related protein TAZ in genetically or chemically induced mouse models of CRC, in patient-derived xenografts, and in enteroid models of CRC. Instead, verteporfin exhibited in vivo selectivity for killing tumor cells in part by impairing the global clearance of high-molecular weight oligomerized proteins, particularly p62 (a sequestrome involved in autophagy) and STAT3 (signal transducer and activator of transcription 3; a transcription factor). Verteporfin inhibited cytokine-induced STAT3 activity and cell proliferation and reduced the viability of cultured CRC cells. Although verteporfin accumulated to a greater extent in normal cells than in tumor cells in vivo, experiments with cultured cells indicated that the normal cells efficiently cleared verteporfin-induced protein oligomers through autophagic and proteasomal pathways. Culturing CRC cells under hypoxic or nutrient-deprived conditions (modeling a typical CRC microenvironment) impaired the clearance of protein oligomers and resulted in cell death, whereas culturing cells under normoxic or glucose-replete conditions protected cell viability and proliferation in the presence of verteporfin. Furthermore, verteporfin suppressed the proliferation of other cancer cell lines even in the absence of YAP1, suggesting that verteporfin may be effective against multiple types of solid cancers.
Collapse
Affiliation(s)
- Huabing Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sadeesh K Ramakrishnan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Daniel Triner
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brook Centofanti
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dhiman Maitra
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, MTA-SE Pediatrics and Nephrology Research Group, Semmelweis University 2nd Department of Pediatrics, Budapest H-1117, Hungary
| | | | - Michael K Dame
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James Varani
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dean E Brenner
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Eric R Fearon
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA. Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA. Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA. Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA. Department of Veterans Affairs Ann Arbor Health Care System, Ann Arbor, MI 48105, USA
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA. Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
308
|
Li C, Kan L, Chen Y, Zheng X, Li W, Zhang W, Cao L, Lin X, Ji S, Huang S, Zhang G, Liu X, Tao Y, Wu S, Chen D. Ci antagonizes Hippo signaling in the somatic cells of the ovary to drive germline stem cell differentiation. Cell Res 2015; 25:1152-70. [PMID: 26403189 DOI: 10.1038/cr.2015.114] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/02/2015] [Accepted: 08/06/2015] [Indexed: 12/17/2022] Open
Abstract
Many stem cell populations are tightly regulated by their local microenvironment (niche), which comprises distinct types of stromal cells. However, little is known about mechanisms by which niche subgroups coordinately determine the stem cell fate. Here we identify that Yki, the key Hippo pathway component, is essential for escort cell (EC) function in promoting germline differentiation in Drosophila ovary. We found that Hedgehog (Hh) signals emanating primarily from cap cells support the function of ECs, where Cubitus interruptus (Ci), the Hh signaling effector, acts to inhibit Hippo kinase cascade activity. Mechanistically, we found that Ci competitively interacts with Hpo and impairs the Hpo-Wts signaling complex formation, thereby promoting Yki nuclear localization. The actions of Ci ensure effective Yki signaling to antagonize Sd/Tgi/Vg-mediated default repression in ECs. This study uncovers a mechanism explaining how subgroups of niche cells coordinate to determine the stem cell fate via Hh-Hippo signaling crosstalk, and enhances our understanding of mechanistic regulations of the oncogenic Yki/YAP signaling.
Collapse
Affiliation(s)
- Chaoyi Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Lijuan Kan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Yan Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiudeng Zheng
- Centre for Computational and Evolutionary Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weini Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Wenxin Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Lei Cao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaohui Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shanming Ji
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Shoujun Huang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Guoqiang Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| | - Xiaohui Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yi Tao
- Centre for Computational and Evolutionary Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shian Wu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Dahua Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang, Beijing 100101, China
| |
Collapse
|
309
|
Di Cara F, Maile TM, Parsons BD, Magico A, Basu S, Tapon N, King-Jones K. The Hippo pathway promotes cell survival in response to chemical stress. Cell Death Differ 2015; 22:1526-39. [PMID: 26021298 PMCID: PMC4532776 DOI: 10.1038/cdd.2015.10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 12/18/2022] Open
Abstract
Cellular stress defense mechanisms have evolved to maintain homeostasis in response to a broad variety of environmental challenges. Stress signaling pathways activate multiple cellular programs that range from the activation of survival pathways to the initiation of cell death when cells are damaged beyond repair. To identify novel players acting in stress response pathways, we conducted a cell culture RNA interference (RNAi) screen using caffeine as a xenobiotic stress-inducing agent, as this compound is a well-established inducer of detoxification response pathways. Specifically, we examined how caffeine affects cell survival when Drosophila kinases and phosphatases were depleted via RNAi. Using this approach, we identified and validated 10 kinases and 4 phosphatases that are essential for cell survival under caffeine-induced stress both in cell culture and living flies. Remarkably, our screen yielded an enrichment of Hippo pathway components, indicating that this pathway regulates cellular stress responses. Indeed, we show that the Hippo pathway acts as a potent repressor of stress-induced cell death. Further, we demonstrate that Hippo activation is necessary to inhibit a pro-apoptotic program triggered by the interaction of the transcriptional co-activator Yki with the transcription factor p53 in response to a range of stress stimuli. Our in vitro and in vivo loss-of-function data therefore implicate Hippo signaling in the transduction of cellular survival signals in response to chemical stress.
Collapse
Affiliation(s)
- F Di Cara
- Department of Cell Biology, Medical Sciences Building, Faculty of Medicine & Dentistry, University of Alberta, Edmonton T6G 2H7, Alberta, Canada
| | - T M Maile
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - B D Parsons
- Department of Medical Microbiology and Immunology, 6-020 Katz Group Centre, University of Alberta, Edmonton AB T6G 2E1, Alberta, Canada
| | - A Magico
- Department of Pediatrics, Faculty of Medicine & Dentistry, Katz Group Centre, University of Alberta, Edmonton T6G 2H7, Alberta, Canada
| | - S Basu
- Centre for Molecular Oncology, Institute of Cancer, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - N Tapon
- Apoptosis and Proliferation Control Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - K King-Jones
- Department of Biological Sciences, University of Alberta, G-504 Biological Sciences Bldg, Edmonton T6G 2E9, Alberta, Canada
| |
Collapse
|
310
|
Stein C, Bardet AF, Roma G, Bergling S, Clay I, Ruchti A, Agarinis C, Schmelzle T, Bouwmeester T, Schübeler D, Bauer A. YAP1 Exerts Its Transcriptional Control via TEAD-Mediated Activation of Enhancers. PLoS Genet 2015; 11:e1005465. [PMID: 26295846 PMCID: PMC4546604 DOI: 10.1371/journal.pgen.1005465] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/23/2015] [Indexed: 12/30/2022] Open
Abstract
YAP1 is a major effector of the Hippo pathway and a well-established oncogene. Elevated YAP1 activity due to mutations in Hippo pathway components or YAP1 amplification is observed in several types of human cancers. Here we investigated its genomic binding landscape in YAP1-activated cancer cells, as well as in non-transformed cells. We demonstrate that TEAD transcription factors mediate YAP1 chromatin-binding genome-wide, further explaining their dominant role as primary mediators of YAP1-transcriptional activity. Moreover, we show that YAP1 largely exerts its transcriptional control via distal enhancers that are marked by H3K27 acetylation and that YAP1 is necessary for this chromatin mark at bound enhancers and the activity of the associated genes. This work establishes YAP1-mediated transcriptional regulation at distal enhancers and provides an expanded set of target genes resulting in a fundamental source to study YAP1 function in a normal and cancer setting. The YAP1/Hippo signaling pathway is a key regulator of organ size and tissue homeostasis, and its dysregulation is linked to cancer development. Elevated activity of YAP1, a transcriptional coactivator and well-established oncogene has been reported to occur in human cancers. Comprehensive identification of YAP1 regulated genes and its mode of action will be of high importance to uncover YAP1 biology that could be exploited for a therapeutic intervention. To this end, we performed genome-wide analyses to identify YAP1 occupied sites in cancer cell lines representing different YAP1/Hippo pathway tumor etiologies and in non-transformed fibroblasts. Our data demonstrate that YAP1 activity is mediated predominantly via TEAD transcription factors supporting the importance of TEADs as main mediators of YAP1-coactivator activity. We further show that YAP1 and TEAD1 exert their transcriptional control via binding to enhancers, leading to characteristic chromatin changes and distal activation of genes. By linking enhancers to genes, we provide a list of novel YAP1 target genes in an oncogenic setting that we show can readily be exploited in tumor classification and provides a foundation for further investigations.
Collapse
Affiliation(s)
- Claudia Stein
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Anaïs Flore Bardet
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Guglielmo Roma
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Sebastian Bergling
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Ieuan Clay
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Alexandra Ruchti
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Claudia Agarinis
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tobias Schmelzle
- Oncology, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tewis Bouwmeester
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Dirk Schübeler
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- University of Basel, Faculty of Sciences, Basel, Switzerland
- * E-mail: (DS); (AB)
| | - Andreas Bauer
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland
- * E-mail: (DS); (AB)
| |
Collapse
|
311
|
Shan HG, Huang CH, Huang DM. Ezrin promotes invasion and metastasis of gastric cancer by up-regulating YAP protein expression. Shijie Huaren Xiaohua Zazhi 2015; 23:3683-3690. [DOI: 10.11569/wcjd.v23.i23.3683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the interactions of Ezrin and Hippo/Yes-associated protein (YAP) in the invasion and metastasis of gastric cancer.
METHODS: Immunohistochemistry (IHC) was used to determine the protein expression of Ezrin and YAP in 209 gastric cancer tissues. MTT assay was carried out to evaluate the effect of Ezrin overexpression with or without siYAP treatment on cell proliferation. Transwell assays were implemented to assess the impact of Ezrin overexpression with or without siYAP on cell migration and invasion.
RESULTS: IHC staining showed that Ezrin and YAP expression was up-regulated in primary gastric cancer compared with their adjacent normal tissues and positively correlated with tumor occurrence, development and metastasis. MTT assay showed that overexpression of Ezrin increased cell proliferation, and knockdown of YAP inhibited cell proliferation. Transwell assays demonstrated that the ectopic expression of Ezrin in SGC7901 cells promoted cell migration and invasion, and knockdown of YAP inhibited cell migration and invasion.
CONCLUSION: Ezrin functions as an oncogene in gastric cancer and can increase YAP protein expression, which in turn up-regulates the expression of the downstream molecules of the Hippo pathway to promote invasion and metastasis of gastric cancer.
Collapse
|
312
|
Meserve JH, Duronio RJ. Scalloped and Yorkie are required for cell cycle re-entry of quiescent cells after tissue damage. Development 2015; 142:2740-51. [PMID: 26160905 DOI: 10.1242/dev.119339] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 06/29/2015] [Indexed: 12/19/2022]
Abstract
Regeneration of damaged tissues typically requires a population of active stem cells. How damaged tissue is regenerated in quiescent tissues lacking a stem cell population is less well understood. We used a genetic screen in the developing Drosophila melanogaster eye to investigate the mechanisms that trigger quiescent cells to re-enter the cell cycle and proliferate in response to tissue damage. We discovered that Hippo signaling regulates compensatory proliferation after extensive cell death in the developing eye. Scalloped and Yorkie, transcriptional effectors of the Hippo pathway, drive Cyclin E expression to induce cell cycle re-entry in cells that normally remain quiescent in the absence of damage. Ajuba, an upstream regulator of Hippo signaling that functions as a sensor of epithelial integrity, is also required for cell cycle re-entry. Thus, in addition to its well-established role in modulating proliferation during periods of tissue growth, Hippo signaling maintains homeostasis by regulating quiescent cell populations affected by tissue damage.
Collapse
Affiliation(s)
- Joy H Meserve
- Curriculum in Genetics & Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Robert J Duronio
- Curriculum in Genetics & Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA Departments of Biology and Genetics, University of North Carolina, Chapel Hill, NC 27599, USA Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
313
|
The Hippo transducers TAZ and YAP in breast cancer: oncogenic activities and clinical implications. Expert Rev Mol Med 2015; 17:e14. [PMID: 26136233 DOI: 10.1017/erm.2015.12] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Hippo signalling is emerging as a tumour suppressor pathway whose function is regulated by an intricate network of intracellular and extracellular cues. Defects in the signal cascade lead to the activation of the Hippo transducers TAZ and YAP. Compelling preclinical evidence showed that TAZ/YAP are often aberrantly engaged in breast cancer (BC), where their hyperactivation culminates into a variety of tumour-promoting functions such as epithelial-to-mesenchymal transition, cancer stem cell generation and therapeutic resistance. Having acquired a more thorough understanding in the biology of TAZ/YAP, and the molecular outputs they elicit, has prompted a first wave of exploratory, clinically-focused analyses aimed at providing initial hints on the prognostic/predictive significance of their expression. In this review, we discuss oncogenic activities linked with TAZ/YAP in BC, and we propose clinical strategies for investigating their role as biomarkers in the clinical setting. Finally, we address the therapeutic potential of TAZ/YAP targeting and the modalities that, in our opinion, should be pursued in order to further study the biological and clinical consequences of their inhibition.
Collapse
|
314
|
Hippo Stabilises Its Adaptor Salvador by Antagonising the HECT Ubiquitin Ligase Herc4. PLoS One 2015; 10:e0131113. [PMID: 26125558 PMCID: PMC4488328 DOI: 10.1371/journal.pone.0131113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/28/2015] [Indexed: 01/13/2023] Open
Abstract
Signalling through the Hippo (Hpo) pathway involves a kinase cascade, which leads to the phosphorylation and inactivation of the pro-growth transcriptional co-activator Yorkie (Yki). Despite the identification of a large number of pathway members and modulators, our understanding of the molecular events that lead to activation of Hpo and the downstream kinase Warts (Wts) remain incomplete. Recently, targeted degradation of several Hpo pathway components has been demonstrated as a means of regulating pathway activity. In particular, the stability of scaffold protein Salvador (Sav), which is believed to promote Hpo/Wts association, is crucially dependent on its binding partner Hpo. In a cell-based RNAi screen for ubiquitin regulators involved in Sav stability, we identify the HECT domain protein Herc4 (HECT and RLD domain containing E3 ligase) as a Sav E3 ligase. Herc4 expression promotes Sav ubiquitylation and degradation, while Herc4 depletion stabilises Sav. Interestingly, Hpo reduces Sav/Herc4 interaction in a kinase-dependent manner. This suggests the existence of a positive feedback loop, where Hpo stabilises its own positive regulator by antagonising Herc4-mediated degradation of Sav.
Collapse
|
315
|
Dong L, Li J, Huang H, Yin MX, Xu J, Li P, Lu Y, Wu W, Yang H, Zhao Y, Zhang L. Growth suppressor lingerer regulates bantam microRNA to restrict organ size. J Mol Cell Biol 2015; 7:415-28. [PMID: 26117838 DOI: 10.1093/jmcb/mjv045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/27/2015] [Indexed: 01/05/2023] Open
Abstract
The evolutionarily conserved Hippo signaling pathway plays an important role in organ size control by regulating cell proliferation and apoptosis. Here, we identify Lingerer (Lig) as a growth suppressor using RNAi modifying screen in Drosophila melanogaster. Loss of lig increases organ size and upregulates bantam (ban) and the expression of the Hippo pathway target genes, while overexpression of lig results in diminished ban expression and organ size reduction. We demonstrate that Lig C-terminal exhibits dominant-negative function on growth and ban expression, and thus plays an important role in organ size control and ban regulation. In addition, we provide evidence that both Yki and Mad are essential for Lig-induced ban expression. We also show that Lig regulates the expression of the Hippo pathway target genes partially via Yorkie. Moreover, we find that Lig physically interacts with and requires Salvador to restrict cell growth. Taken together, we demonstrate that Lig functions as a critical growth suppressor to control organ size via ban and Hippo signaling.
Collapse
Affiliation(s)
- Liang Dong
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinhui Li
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongling Huang
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Meng-Xin Yin
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinjin Xu
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peixue Li
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenqing Wu
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hang Yang
- Department of Biochemistry and Molecular Biology, USC Health Science Campus, Los Angeles, CA 90033, USA
| | - Yun Zhao
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
316
|
Abstract
The heart is the first organ formed during mammalian development. A properly sized and functional heart is vital throughout the entire lifespan. Loss of cardiomyocytes because of injury or diseases leads to heart failure, which is a major cause of human morbidity and mortality. Unfortunately, regenerative potential of the adult heart is limited. The Hippo pathway is a recently identified signaling cascade that plays an evolutionarily conserved role in organ size control by inhibiting cell proliferation, promoting apoptosis, regulating fates of stem/progenitor cells, and in some circumstances, limiting cell size. Interestingly, research indicates a key role of this pathway in regulation of cardiomyocyte proliferation and heart size. Inactivation of the Hippo pathway or activation of its downstream effector, the Yes-associated protein transcription coactivator, improves cardiac regeneration. Several known upstream signals of the Hippo pathway such as mechanical stress, G-protein-coupled receptor signaling, and oxidative stress are known to play critical roles in cardiac physiology. In addition, Yes-associated protein has been shown to regulate cardiomyocyte fate through multiple transcriptional mechanisms. In this review, we summarize and discuss current findings on the roles and mechanisms of the Hippo pathway in heart development, injury, and regeneration.
Collapse
Affiliation(s)
- Qi Zhou
- From the Life Sciences Institute, Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, China (Q.Z., B.Z.); Institute of Aging Research, Hangzhou Normal University, Hangzhou, Zhejiang, China (L.L.); and Department of Pharmacology and Moores Cancer Center, University of California at San Diego, La Jolla (K.-L.G.)
| | - Li Li
- From the Life Sciences Institute, Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, China (Q.Z., B.Z.); Institute of Aging Research, Hangzhou Normal University, Hangzhou, Zhejiang, China (L.L.); and Department of Pharmacology and Moores Cancer Center, University of California at San Diego, La Jolla (K.-L.G.)
| | - Bin Zhao
- From the Life Sciences Institute, Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, China (Q.Z., B.Z.); Institute of Aging Research, Hangzhou Normal University, Hangzhou, Zhejiang, China (L.L.); and Department of Pharmacology and Moores Cancer Center, University of California at San Diego, La Jolla (K.-L.G.).
| | - Kun-Liang Guan
- From the Life Sciences Institute, Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, China (Q.Z., B.Z.); Institute of Aging Research, Hangzhou Normal University, Hangzhou, Zhejiang, China (L.L.); and Department of Pharmacology and Moores Cancer Center, University of California at San Diego, La Jolla (K.-L.G.).
| |
Collapse
|
317
|
Pan D. YAPing Hippo Forecasts a New Target for Lung Cancer Prevention and Treatment. J Clin Oncol 2015; 33:2311-3. [PMID: 26056180 DOI: 10.1200/jco.2015.61.2093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Duojia Pan
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
318
|
Cox CM, Mandell EK, Stewart L, Lu R, Johnson DL, McCarter SD, Tavares A, Runyan R, Ghosh S, Wilson JM. Endosomal regulation of contact inhibition through the AMOT:YAP pathway. Mol Biol Cell 2015; 26:2673-84. [PMID: 25995376 PMCID: PMC4501364 DOI: 10.1091/mbc.e15-04-0224] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/11/2015] [Indexed: 11/15/2022] Open
Abstract
It was shown previously that endotubin, an integral membrane protein of endosomes, regulates the trafficking of tight junction proteins between endosomes and the tight junctions. Here it is shown that endotubin regulates YAP localization on endosomes through its interaction with AMOT and thus may play a role in contact inhibition. Contact-mediated inhibition of cell proliferation is an essential part of organ growth control; the transcription coactivator Yes-associated protein (YAP) plays a pivotal role in this process. In addition to phosphorylation-dependent regulation of YAP, the integral membrane protein angiomotin (AMOT) and AMOT family members control YAP through direct binding. Here we report that regulation of YAP activity occurs at the endosomal membrane through a dynamic interaction of AMOT with an endosomal integral membrane protein, endotubin (EDTB). EDTB interacts with both AMOT and occludin and preferentially associates with occludin in confluent cells but with AMOT family members in subconfluent cells. EDTB competes with YAP for binding to AMOT proteins in subconfluent cells. Overexpression of the cytoplasmic domain or full-length EDTB induces translocation of YAP to the nucleus, an overgrowth phenotype, and growth in soft agar. This increase in proliferation is dependent upon YAP activity and is complemented by overexpression of p130-AMOT. Furthermore, overexpression of EDTB inhibits the AMOT:YAP interaction. EDTB and AMOT have a greater association in subconfluent cells compared with confluent cells, and this association is regulated at the endosomal membrane. These data provide a link between the trafficking of tight junction proteins through endosomes and contact-inhibition-regulated cell growth.
Collapse
Affiliation(s)
- Christopher M Cox
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Edward K Mandell
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06511
| | - Lorraine Stewart
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Ruifeng Lu
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Debra L Johnson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Sarah D McCarter
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Andre Tavares
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Ray Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| | - Sourav Ghosh
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06511
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
319
|
Wittkorn E, Sarkar A, Garcia K, Kango-Singh M, Singh A. The Hippo pathway effector Yki downregulates Wg signaling to promote retinal differentiation in the Drosophila eye. Development 2015; 142:2002-13. [PMID: 25977365 DOI: 10.1242/dev.117358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 04/16/2015] [Indexed: 01/22/2023]
Abstract
The evolutionarily conserved Hippo signaling pathway is known to regulate cell proliferation and maintain tissue homeostasis during development. We found that activation of Yorkie (Yki), the effector of the Hippo signaling pathway, causes separable effects on growth and differentiation of the Drosophila eye. We present evidence supporting a role for Yki in suppressing eye fate by downregulation of the core retinal determination genes. Other upstream regulators of the Hippo pathway mediate this effect of Yki on retinal differentiation. Here, we show that, in the developing eye, Yki can prevent retinal differentiation by blocking morphogenetic furrow (MF) progression and R8 specification. The inhibition of MF progression is due to ectopic induction of Wingless (Wg) signaling and Homothorax (Hth), the negative regulators of eye development. Modulating Wg signaling can modify Yki-mediated suppression of eye fate. Furthermore, ectopic Hth induction due to Yki activation in the eye is dependent on Wg. Last, using Cut (Ct), a marker for the antennal fate, we show that suppression of eye fate by hyperactivation of yki does not change the cell fate (from eye to antenna-specific fate). In summary, we provide the genetic mechanism by which yki plays a role in cell fate specification and differentiation - a novel aspect of Yki function that is emerging from multiple model organisms.
Collapse
Affiliation(s)
- Erika Wittkorn
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Ankita Sarkar
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Kristine Garcia
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Madhuri Kango-Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA Premedical Program, University of Dayton, Dayton, OH 45469, USA Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH 45469, USA
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA Premedical Program, University of Dayton, Dayton, OH 45469, USA Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH 45469, USA
| |
Collapse
|
320
|
Plouffe SW, Hong AW, Guan KL. Disease implications of the Hippo/YAP pathway. Trends Mol Med 2015; 21:212-22. [PMID: 25702974 PMCID: PMC4385444 DOI: 10.1016/j.molmed.2015.01.003] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 12/14/2022]
Abstract
The Hippo signaling pathway is important for controlling organ size and tissue homeostasis. Originally identified in Drosophila melanogaster, the core components of the Hippo pathway are highly conserved in mammals. The Hippo pathway can be modulated by a wide range of stimuli, including G protein-coupled receptor (GPCR) signaling, changes in the actin cytoskeleton, cell-cell contact, and cell polarity. When activated, the Hippo pathway functions as a tumor suppressor to limit cell growth. However, dysregulation by genetic inactivation of core pathway components or amplification or gene fusion of its downstream effectors results in increased cell proliferation and decreased apoptosis and differentiation. Unsurprisingly, this can lead to tissue overgrowth, tumorigenesis, and many other diseases.
Collapse
Affiliation(s)
- Steven W Plouffe
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Audrey W Hong
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Kun-Liang Guan
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
321
|
Fletcher GC, Elbediwy A, Khanal I, Ribeiro PS, Tapon N, Thompson BJ. The Spectrin cytoskeleton regulates the Hippo signalling pathway. EMBO J 2015; 34:940-54. [PMID: 25712476 PMCID: PMC4388601 DOI: 10.15252/embj.201489642] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 01/15/2015] [Accepted: 01/21/2015] [Indexed: 12/20/2022] Open
Abstract
The Spectrin cytoskeleton is known to be polarised in epithelial cells, yet its role remains poorly understood. Here, we show that the Spectrin cytoskeleton controls Hippo signalling. In the developing Drosophila wing and eye, loss of apical Spectrins (alpha/beta-heavy dimers) produces tissue overgrowth and mis-regulation of Hippo target genes, similar to loss of Crumbs (Crb) or the FERM-domain protein Expanded (Ex). Apical beta-heavy Spectrin binds to Ex and co-localises with it at the apical membrane to antagonise Yki activity. Interestingly, in both the ovarian follicular epithelium and intestinal epithelium of Drosophila, apical Spectrins and Crb are dispensable for repression of Yki, while basolateral Spectrins (alpha/beta dimers) are essential. Finally, the Spectrin cytoskeleton is required to regulate the localisation of the Hippo pathway effector YAP in response to cell density human epithelial cells. Our findings identify both apical and basolateral Spectrins as regulators of Hippo signalling and suggest Spectrins as potential mechanosensors.
Collapse
Affiliation(s)
- Georgina C Fletcher
- Epithelial Biology Laboratory, Cancer Research UK - London Research Institute, London, UK
| | - Ahmed Elbediwy
- Epithelial Biology Laboratory, Cancer Research UK - London Research Institute, London, UK
| | - Ichha Khanal
- Epithelial Biology Laboratory, Cancer Research UK - London Research Institute, London, UK
| | - Paulo S Ribeiro
- Apoptosis and Cell Proliferation Laboratory, Cancer Research UK - London Research Institute, London, UK Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Nic Tapon
- Apoptosis and Cell Proliferation Laboratory, Cancer Research UK - London Research Institute, London, UK
| | - Barry J Thompson
- Epithelial Biology Laboratory, Cancer Research UK - London Research Institute, London, UK
| |
Collapse
|
322
|
Gokhale RH, Shingleton AW. Size control: the developmental physiology of body and organ size regulation. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:335-56. [PMID: 25808999 DOI: 10.1002/wdev.181] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 01/08/2015] [Accepted: 01/29/2015] [Indexed: 01/04/2023]
Abstract
The developmental regulation of final body and organ size is fundamental to generating a functional and correctly proportioned adult. Research over the last two decades has identified a long list of genes and signaling pathways that, when perturbed, influence final body size. However, body and organ size are ultimately a characteristic of the whole organism, and how these myriad genes and pathways function within a physiological context to control size remains largely unknown. In this review, we first describe the major size-regulatory signaling pathways: the Insulin/IGF-, RAS/RAF/MAPK-, TOR-, Hippo-, and JNK-signaling pathways. We then explore what is known of how these pathways regulate five major aspects of size regulation: growth rate, growth duration, target size, negative growth and growth coordination. While this review is by no means exhaustive, our goal is to provide a conceptual framework for integrating the mechanisms of size control at a molecular-genetic level with the mechanisms of size control at a physiological level.
Collapse
Affiliation(s)
- Rewatee H Gokhale
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Alexander W Shingleton
- Department of Biology, Lake Forest College, Lake Forest, IL, USA.,Department of Zoology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
323
|
Enzo E, Santinon G, Pocaterra A, Aragona M, Bresolin S, Forcato M, Grifoni D, Pession A, Zanconato F, Guzzo G, Bicciato S, Dupont S. Aerobic glycolysis tunes YAP/TAZ transcriptional activity. EMBO J 2015; 34:1349-70. [PMID: 25796446 DOI: 10.15252/embj.201490379] [Citation(s) in RCA: 339] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/26/2015] [Indexed: 12/14/2022] Open
Abstract
Increased glucose metabolism and reprogramming toward aerobic glycolysis are a hallmark of cancer cells, meeting their metabolic needs for sustained cell proliferation. Metabolic reprogramming is usually considered as a downstream consequence of tumor development and oncogene activation; growing evidence indicates, however, that metabolism on its turn can support oncogenic signaling to foster tumor malignancy. Here, we explored how glucose metabolism regulates gene transcription and found an unexpected link with YAP/TAZ, key transcription factors regulating organ growth, tumor cell proliferation and aggressiveness. When cells actively incorporate glucose and route it through glycolysis, YAP/TAZ are fully active; when glucose metabolism is blocked, or glycolysis is reduced, YAP/TAZ transcriptional activity is decreased. Accordingly, glycolysis is required to sustain YAP/TAZ pro-tumorigenic functions, and YAP/TAZ are required for the full deployment of glucose growth-promoting activity. Mechanistically we found that phosphofructokinase (PFK1), the enzyme regulating the first committed step of glycolysis, binds the YAP/TAZ transcriptional cofactors TEADs and promotes their functional and biochemical cooperation with YAP/TAZ. Strikingly, this regulation is conserved in Drosophila, where phosphofructokinase is required for tissue overgrowth promoted by Yki, the fly homologue of YAP. Moreover, gene expression regulated by glucose metabolism in breast cancer cells is strongly associated in a large dataset of primary human mammary tumors with YAP/TAZ activation and with the progression toward more advanced and malignant stages. These findings suggest that aerobic glycolysis endows cancer cells with particular metabolic properties and at the same time sustains transcription factors with potent pro-tumorigenic activities such as YAP/TAZ.
Collapse
Affiliation(s)
- Elena Enzo
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Giulia Santinon
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Arianna Pocaterra
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | | | - Silvia Bresolin
- Department of Woman and Child Health, University of Padova, Padua, Italy
| | - Mattia Forcato
- Department of Life Sciences, Center for Genome Research University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Grifoni
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | - Annalisa Pession
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | | | - Giulia Guzzo
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research University of Modena and Reggio Emilia, Modena, Italy
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padova, Padua, Italy
| |
Collapse
|
324
|
Santucci M, Vignudelli T, Ferrari S, Mor M, Scalvini L, Bolognesi ML, Uliassi E, Costi MP. The Hippo Pathway and YAP/TAZ-TEAD Protein-Protein Interaction as Targets for Regenerative Medicine and Cancer Treatment. J Med Chem 2015; 58:4857-73. [PMID: 25719868 DOI: 10.1021/jm501615v] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Hippo pathway is an important organ size control signaling network and the major regulatory mechanism of cell-contact inhibition. Yes associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are its targets and terminal effectors: inhibition of the pathway promotes YAP/TAZ translocation to the nucleus, where they interact with transcriptional enhancer associate domain (TEAD) transcription factors and coactivate the expression of target genes, promoting cell proliferation. Defects in the pathway can result in overgrowth phenotypes due to deregulation of stem-cell proliferation and apoptosis; members of the pathway are directly involved in cancer development. The pharmacological regulation of the pathway might be useful in cancer prevention, treatment, and regenerative medicine applications; currently, a few compounds can selectively modulate the pathway. In this review, we present an overview of the Hippo pathway, the sequence and structural analysis of YAP/TAZ, the known pharmacological modulators of the pathway, especially those targeting YAP/TAZ-TEAD interaction.
Collapse
Affiliation(s)
- Matteo Santucci
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Tatiana Vignudelli
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Stefania Ferrari
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| | - Marco Mor
- ‡Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Laura Scalvini
- ‡Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Maria Laura Bolognesi
- §Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Elisa Uliassi
- §Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Paola Costi
- †Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 183, Modena 41125, Italy
| |
Collapse
|
325
|
Bunker BD, Nellimoottil TT, Boileau RM, Classen AK, Bilder D. The transcriptional response to tumorigenic polarity loss in Drosophila. eLife 2015; 4. [PMID: 25719210 PMCID: PMC4369581 DOI: 10.7554/elife.03189] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/25/2015] [Indexed: 12/14/2022] Open
Abstract
Loss of polarity correlates with progression of epithelial cancers, but how plasma membrane misorganization drives oncogenic transcriptional events remains unclear. The polarity regulators of the Drosophila Scribble (Scrib) module are potent tumor suppressors and provide a model for mechanistic investigation. RNA profiling of Scrib mutant tumors reveals multiple signatures of neoplasia, including altered metabolism and dedifferentiation. Prominent among these is upregulation of cytokine-like Unpaired (Upd) ligands, which drive tumor overgrowth. We identified a polarity-responsive enhancer in upd3, which is activated in a coincident manner by both JNK-dependent Fos and aPKC-mediated Yki transcription. This enhancer, and Scrib mutant overgrowth in general, are also sensitive to activity of the Polycomb Group (PcG), suggesting that PcG attenuation upon polarity loss potentiates select targets for activation by JNK and Yki. Our results link epithelial organization to signaling and epigenetic regulators that control tissue repair programs, and provide insight into why epithelial polarity is tumor-suppressive. DOI:http://dx.doi.org/10.7554/eLife.03189.001 The cavities and organs within our body are lined with epithelial cells, which connect to each other to form continuous barriers. These cells have a highly polarized structure in which different components are found at the top and bottom of cells. In the fruit fly and most other animals, three genes known as the Scribble module control the polarity of epithelial cells. If these genes are faulty, the cells lose their polarity, break the epithelial barrier, and grow rapidly to form a tumor. Most malignant tumors that form from epithelial cells have lost normal cell polarity, so understanding how the organization and growth of epithelial cells are linked is a critical question. It is not clear how the loss of cell polarity can drive tumor formation. Here, Bunker et al. used a technique called RNA sequencing to study the expression of genes in tumor cells that have mutations in the Scribble module. Hundreds of genes in the tumor cells had different levels of expression from the levels seen in normal fly cells. One of these is a gene called upd3, which was expressed much more highly in tumor cells than in normal cells. This gene activates a signaling pathway—called the JAK/STAT pathway—that promotes cell growth and division in many animals. Bunker et al. found that experimentally lowering the activity of the JAK/STAT pathway reduced the growth of the tumor cells that had lost normal polarity. Further experiments show that disrupting the layer of epithelial cells activates two other signaling pathways that work together to switch on the upd3 gene when cell polarity is lost. Proteins belonging to the Polycomb Group also control the expression of upd3 and other genes involved in cell growth by altering how genetic material is packaged in cells. The similarities between this response and the response to tissue damage suggest that the loss of polarity drives tumor formation through an unstoppable wound-healing reaction. Therefore, Bunker et al.'s findings link the formation of epithelial tumors to the signaling pathways that control the repair of damaged tissues. DOI:http://dx.doi.org/10.7554/eLife.03189.002
Collapse
Affiliation(s)
- Brandon D Bunker
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Tittu T Nellimoottil
- University of Southern California, Department of Biological Sciences, Los Angeles, United States
| | - Ryan M Boileau
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Anne K Classen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - David Bilder
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
326
|
Hwang B, An Y, Agata K, Umesono Y. Two distinct roles of the yorkie/yap gene during homeostasis in the planarian Dugesia japonica. Dev Growth Differ 2015; 57:209-17. [PMID: 25708270 PMCID: PMC4415594 DOI: 10.1111/dgd.12195] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/18/2014] [Accepted: 12/21/2014] [Indexed: 12/21/2022]
Abstract
Adult planarians possess somatic pluripotent stem cells called neoblasts that give rise to all missing cell types during regeneration and homeostasis. Recent studies revealed that the Yorkie (Yki)/Yes-associated protein (YAP) transcriptional coactivator family plays an important role in the regulation of tissue growth during development and regeneration, and therefore we investigated the role of a planarian yki-related gene (termed Djyki) during regeneration and homeostasis of the freshwater planarian Dugesia japonica. We found that knockdown of the function of Djyki by RNA interference (RNAi) downregulated neoblast proliferation and caused regeneration defects after amputation. In addition, Djyki RNAi caused edema during homeostasis. These seemingly distinct defects induced by Djyki RNAi were rescued by simultaneous RNAi of a planarian mats-related gene (termed Djmats), suggesting an important role of Djmats in the negative regulation of Djyki, in accordance with the conservation of the functional relationship of these two genes during the course of evolution. Interestingly, Djyki RNAi did not prevent normal protonephridial structure, suggesting that Djyki RNAi induced the edema phenotype without affecting the excretory system. Further analyses revealed that increased expression of the D. japonica gene DjaquaporinA (DjaqpA), which belongs to a large gene family that encodes a water channel protein for the regulation of transcellular water flow, promoted the induction of edema, but not defects in neoblast dynamics, in Djyki(RNAi) animals. Thus, we conclude that Djyki plays two distinct roles in the regulation of active proliferation of stem cells and in osmotic water transport across the body surface in D. japonica.
Collapse
Affiliation(s)
- Byulnim Hwang
- Department of Biophysics, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto
| | | | | | | |
Collapse
|
327
|
Yeung B, Yu J, Yang X. Roles of the Hippo pathway in lung development and tumorigenesis. Int J Cancer 2015; 138:533-9. [PMID: 25644176 DOI: 10.1002/ijc.29457] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 01/23/2015] [Indexed: 02/06/2023]
Abstract
Lung cancer is the most commonly diagnosed cancer and accounts for one fifth of all cancer deaths worldwide. Although significant progress has been made toward our understanding of the causes of lung cancer, the 5-year survival is still lower than 15%. Therefore, there is an urgent need for novel lung cancer biomarkers and drug targets. The Hippo signaling pathway is an emerging signaling pathway that regulates various biological processes. Recently, increasing evidence suggests that the Hippo pathway may play important roles in not only lung development but also lung tumorigenesis. In this review article, we will summarize the most recent advances and predict future directions on this new cancer research field.
Collapse
Affiliation(s)
- Benjamin Yeung
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jihang Yu
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
328
|
Su T, Bondar T, Zhou X, Zhang C, He H, Medzhitov R. Two-signal requirement for growth-promoting function of Yap in hepatocytes. eLife 2015; 4:e02948. [PMID: 25667983 PMCID: PMC4363878 DOI: 10.7554/elife.02948] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 02/09/2015] [Indexed: 01/09/2023] Open
Abstract
The transcriptional coactivator Yes-associated protein (Yap) promotes proliferation and inhibits apoptosis, suggesting that Yap functions as an oncogene. Most oncogenes, however, require a combination of at least two signals to promote proliferation. In this study, we present evidence that Yap activation is insufficient to promote growth in the otherwise normal tissue. Using a mosaic mouse model, we demonstrate that Yap overexpression in a fraction of hepatocytes does not lead to their clonal expansion, as proliferation is counterbalanced by increased apoptosis. To shift the activity of Yap towards growth, a second signal provided by tissue damage or inflammation is required. In response to liver injury, Yap drives clonal expansion, suppresses hepatocyte differentiation, and promotes a progenitor phenotype. These results suggest that Yap activation is insufficient to promote growth in the absence of a second signal thus coordinating tissue homeostasis and repair.
Collapse
Affiliation(s)
- Tian Su
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Tanya Bondar
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Xu Zhou
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Cuiling Zhang
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Hang He
- Peking-Yale Joint Center for Plant Molecular Genetics and Agro-Biotechnology, National Laboratory of Protein Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing, China
| | - Ruslan Medzhitov
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
329
|
Affiliation(s)
- Fa-Xing Yu
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
- Children's Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China 200032
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| | - Steven W. Plouffe
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
330
|
Rashidian J, Le Scolan E, Ji X, Zhu Q, Mulvihill MM, Nomura D, Luo K. Ski regulates Hippo and TAZ signaling to suppress breast cancer progression. Sci Signal 2015; 8:ra14. [PMID: 25670202 DOI: 10.1126/scisignal.2005735] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ski, the transforming protein of the avian Sloan-Kettering retrovirus, inhibits transforming growth factor-β (TGF-β)/Smad signaling and displays both pro-oncogenic and anti-oncogenic activities in human cancer. Inhibition of TGF-β signaling is likely responsible for the pro-oncogenic activity of Ski. We investigated the mechanism(s) underlying the tumor suppressor activity of Ski and found that Ski suppressed the activity of the Hippo signaling effectors TAZ and YAP to inhibit breast cancer progression. TAZ and YAP are transcriptional coactivators that can contribute to cancer by promoting proliferation, tumorigenesis, and cancer stem cell expansion. Hippo signaling activates the the Lats family of kinases, which phosphorylate TAZ and YAP, resulting in cytoplasmic retention and degradation and inhibition of their transcriptional activity. We showed that Ski interacted with multiple components of the Hippo pathway to facilitate activation of Lats2, resulting in increased phosphorylation and subsequent degradation of TAZ. Ski also promoted the degradation of a constitutively active TAZ mutant that is not phosphorylated by Lats, suggesting the existence of a Lats2-independent degradation pathway. Finally, we showed that Ski repressed the transcriptional activity of TAZ by binding to the TAZ partner TEAD and recruiting the transcriptional co-repressor NCoR1 to the TEAD-TAZ complex. Ski effectively reversed transformation and epithelial-to-mesenchyme transition in cultured breast cancer cells and metastasis in TAZ-expressing xenografted tumors. Thus, Ski inhibited the function of TAZ through multiple mechanisms in human cancer cells.
Collapse
Affiliation(s)
- Juliet Rashidian
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Erwan Le Scolan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xiaodan Ji
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Melinda M Mulvihill
- Department of Nutritional Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel Nomura
- Department of Nutritional Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA. Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
331
|
Yu L, Daniels JP, Wu H, Wolf MJ. Cardiac hypertrophy induced by active Raf depends on Yorkie-mediated transcription. Sci Signal 2015; 8:ra13. [PMID: 25650441 DOI: 10.1126/scisignal.2005719] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Organ hypertrophy can result from enlargement of individual cells or from cell proliferation or both. Activating mutations in the serine-threonine kinase Raf cause cardiac hypertrophy and contribute to Noonan syndrome in humans. Cardiac-specific expression of activated Raf also causes hypertrophy in Drosophila melanogaster. We found that Yorkie (Yki), a transcriptional coactivator in the Hippo pathway that regulates organ size, is required for Raf-induced cardiac hypertrophy in flies. Although aberrant activation of Yki orthologs stimulates cardiac hyperplasia in mice, cardiac-specific expression of an activated mutant form of Yki in fruit flies caused cardiac hypertrophy without hyperplasia. Knockdown of Yki caused cardiac dilation without loss of cardiomyocytes and prevented Raf-induced cardiac hypertrophy. In flies, Yki-induced cardiac hypertrophy required the TEA domain-containing transcription factor Scalloped, and, in mammalian cells, expression of mouse Raf(L613V), an activated form of Raf with a Noonan syndrome mutation, increased Yki-induced Scalloped activity. Furthermore, overexpression of Tgi (a Tondu domain-containing Scalloped-binding corepressor) in the fly heart abrogated Yki- or Raf-induced cardiac hypertrophy. Thus, crosstalk between Raf and Yki occurs in the heart and can influence Raf-mediated cardiac hypertrophy.
Collapse
Affiliation(s)
- Lin Yu
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Joseph P Daniels
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Huihui Wu
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Matthew J Wolf
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
332
|
The hippo pathway effector YAP regulates motility, invasion, and castration-resistant growth of prostate cancer cells. Mol Cell Biol 2015; 35:1350-62. [PMID: 25645929 DOI: 10.1128/mcb.00102-15] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Yes-associated protein (YAP) is an effector of the Hippo tumor suppressor pathway. The functional significance of YAP in prostate cancer has remained elusive. In this study, we first show that enhanced expression of YAP is able to transform immortalized prostate epithelial cells and promote migration and invasion in both immortalized and cancerous prostate cells. We found that YAP mRNA was upregulated in androgen-insensitive prostate cancer cells (LNCaP-C81 and LNCaP-C4-2 cells) compared to the level in androgen-sensitive LNCaP cells. Importantly, ectopic expression of YAP activated androgen receptor signaling and was sufficient to promote LNCaP cells from an androgen-sensitive state to an androgen-insensitive state in vitro, and YAP conferred castration resistance in vivo. Accordingly, YAP knockdown greatly reduced the rates of migration and invasion of LNCaP-C4-2 cells and under androgen deprivation conditions largely blocked cell division in LNCaP-C4-2 cells. Mechanistically, we found that extracellular signal-regulated kinase-ribosomal s6 kinase signaling was downstream of YAP for cell survival, migration, and invasion in androgen-insensitive cells. Finally, immunohistochemistry showed significant upregulation and hyperactivation of YAP in castration-resistant prostate tumors compared to their levels in hormone-responsive prostate tumors. Together, our results identify YAP to be a novel regulator in prostate cancer cell motility, invasion, and castration-resistant growth and as a potential therapeutic target for metastatic castration-resistant prostate cancer (CRPC).
Collapse
|
333
|
Sarikaya DP, Extavour CG. The Hippo pathway regulates homeostatic growth of stem cell niche precursors in the Drosophila ovary. PLoS Genet 2015; 11:e1004962. [PMID: 25643260 PMCID: PMC4333732 DOI: 10.1371/journal.pgen.1004962] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/18/2014] [Indexed: 01/15/2023] Open
Abstract
The Hippo pathway regulates organ size, stem cell proliferation and tumorigenesis in adult organs. Whether the Hippo pathway influences establishment of stem cell niche size to accommodate changes in organ size, however, has received little attention. Here, we ask whether Hippo signaling influences the number of stem cell niches that are established during development of the Drosophila larval ovary, and whether it interacts with the same or different effector signaling pathways in different cell types. We demonstrate that canonical Hippo signaling regulates autonomous proliferation of the soma, while a novel hippo-independent activity of Yorkie regulates autonomous proliferation of the germ line. Moreover, we demonstrate that Hippo signaling mediates non-autonomous proliferation signals between germ cells and somatic cells, and contributes to maintaining the correct proportion of these niche precursors. Finally, we show that the Hippo pathway interacts with different growth pathways in distinct somatic cell types, and interacts with EGFR and JAK/STAT pathways to regulate non-autonomous proliferation of germ cells. We thus provide evidence for novel roles of the Hippo pathway in establishing the precise balance of soma and germ line, the appropriate number of stem cell niches, and ultimately regulating adult female reproductive capacity. During development, organ growth must be carefully regulated to make sure that organs achieve the correct final size needed for organ function. In organs that are made of many different types of cells, this growth regulation is likely to be particularly complex, because it is important for organs to have appropriate proportions, or relative numbers, of the different kinds of cells that make up the organ, as well as the correct number of total cells. One method that cells use to regulate organ growth is a signaling pathway called the Hippo pathway. However, Hippo signaling has been studied, to date, primarily in organ systems that are made up of one cell type. In this study, we examine how Hippo signaling can work to regulate the proportions of different types of cells, as well as the total number of cells in an organ. To do this, we used the developing ovary of the fruit fly as a study system. We found that (1) Hippo signaling regulates the proliferation of many different cell types of the ovary; and (2) Hippo signaling activity in one cell type influences proliferation of other cell types, thus ensuring appropriate proportions of different ovarian cell types.
Collapse
Affiliation(s)
- Didem P. Sarikaya
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Cassandra G. Extavour
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
334
|
Abstract
The Hippo and c-Jun N-terminal kinase (JNK) pathway both regulate growth and contribute to tumorigenesis when dysregulated. Whereas the Hippo pathway acts via the transcription coactivator Yki/YAP to regulate target gene expression, JNK signaling, triggered by various modulators including Rho GTPases, activates the transcription factors Jun and Fos. Here, we show that impaired Hippo signaling induces JNK activation through Rho1. Blocking Rho1-JNK signaling suppresses Yki-induced overgrowth in the wing disk, whereas ectopic Rho1 expression promotes tissue growth when apoptosis is prohibited. Furthermore, Yki directly regulates Rho1 transcription via the transcription factor Sd. Thus, our results have identified a novel molecular link between the Hippo and JNK pathways and implicated the essential role of the JNK pathway in Hippo signaling-related tumorigenesis.
Collapse
|
335
|
Wang LH, Baker NE. Salvador-Warts-Hippo pathway in a developmental checkpoint monitoring helix-loop-helix proteins. Dev Cell 2015; 32:191-202. [PMID: 25579975 DOI: 10.1016/j.devcel.2014.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 10/16/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022]
Abstract
The E proteins and Id proteins are, respectively, the positive and negative heterodimer partners for the basic-helix-loop-helix protein family and as such contribute to a remarkably large number of cell-fate decisions. E proteins and Id proteins also function to inhibit or promote cell proliferation and cancer. Using a genetic modifier screen in Drosophila, we show that the Id protein Extramacrochaetae enables growth by suppressing activation of the Salvador-Warts-Hippo pathway of tumor suppressors, activation that requires transcriptional activation of the expanded gene by the E protein Daughterless. Daughterless protein binds to an intronic enhancer in the expanded gene, both activating the SWH pathway independently of the transmembrane protein Crumbs and bypassing the negative feedback regulation that targets the same expanded enhancer. Thus, the Salvador-Warts-Hippo pathway has a cell-autonomous function to prevent inappropriate differentiation due to transcription factor imbalance and monitors the intrinsic developmental status of progenitor cells, distinct from any responses to cell-cell interactions.
Collapse
Affiliation(s)
- Lan-Hsin Wang
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
336
|
Brás-Pereira C, Casares F, Janody F. The retinal determination gene dachshund restricts cell proliferation by limiting the activity of the Homothorax-Yorkie complex. Development 2015; 142:1470-9. [DOI: 10.1242/dev.113340] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 02/20/2015] [Indexed: 12/17/2022]
Abstract
The Drosophila transcriptional co-activator protein Yorkie and its vertebrate orthologs YAP and TAZ are potent oncogenes, whose activity is normally kept in check by the upstream Hippo kinase module. Upon its translocation into the nucleus, Yorkie forms complexes with several tissue-specific DNA-binding partners, which help to define the tissue-specific target genes of Yorkie. In the progenitor cells of the eye imaginal disc, the DNA-binding transcription factor Homothorax is required for Yorkie-promoted proliferation and survival through regulation of the bantam microRNA (miRNA). The transit from proliferating progenitors to cell cycle quiescent precursors is associated with the progressive loss of Homothorax and gain of Dachshund, a nuclear protein related to the Sno/Ski family of co-repressors. We have identified Dachshund as an inhibitor of Homothorax-Yorkie-mediated cell proliferation. Loss of dachshund induces Yorkie-dependent tissue overgrowth. Conversely, overexpressing dachshund inhibits tissue growth, prevents Yorkie or Homothorax-mediated cell proliferation of disc epithelia and restricts the transcriptional activity of the Yorkie-Homothorax complex on the bantam enhancer in Drosophila cells. In addition, Dachshund collaborates with the Decapentaplegic receptor Thickveins to repress Homothorax and Cyclin B expression in quiescent precursors. The antagonistic roles of Homothorax and Dachshund in Yorkie activity, together with their mutual repression, ensure that progenitor and precursor cells are under distinct proliferation regimes. Based on the crucial role of the human dachshund homolog DACH1 in tumorigenesis, our work suggests that DACH1 might prevent cellular transformation by limiting the oncogenic activity of YAP and/or TAZ.
Collapse
Affiliation(s)
- Catarina Brás-Pereira
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras P-2780-156, Portugal
| | - Fernando Casares
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Seville 41013, Spain
| | - Florence Janody
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras P-2780-156, Portugal
| |
Collapse
|
337
|
Abstract
Over the past decade, discoveries on Hippo signaling have revealed a complex signaling network integrating various signaling pathways to modulate tissue homeostasis, organ size control, tissue repair, and regeneration. Malfunction of the Hippo pathway is associated with tumor and cancer development. Moreover, Hippo signaling has been proposed to act in numerous stem cells in a variety of organisms. Recently, more attention has been paid to define the functions of the Hippo pathway in tissue-specific stem cells, which have great potential to be used in cell-based therapies. Here we provide an overview of its roles in regulating stem cells in epithelial tissues and its potential implications in related cancers.
Collapse
Affiliation(s)
- Meng-Xin Yin
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
338
|
Zhu C, Li L, Zhao B. The regulation and function of YAP transcription co-activator. Acta Biochim Biophys Sin (Shanghai) 2015; 47:16-28. [PMID: 25487920 DOI: 10.1093/abbs/gmu110] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Hippo pathway was initially identified in Drosophila by genetic mosaic screens for tumor suppressor genes. Researches indicated that the Hippo pathway is a key regulator of organ size and is conserved during evolution. Furthermore, studies of mouse models and clinical samples demonstrated the importance of Hippo pathway dysregulation in human cancer development. In addition, the Hippo pathway contributes to progenitor cell and stem cell self-renewal and is thus involved in tissue regeneration. In the Hippo pathway, MST1/2 kinases together with the adaptor protein SAV phosphorylate LATS1/2 kinases. Interaction with an adaptor protein MOB is also important for LATS1/2 activation. Activated LATS1/2 in turn phosphorylate and inhibit Yes-associated protein (YAP). YAP is a key downstream effector of the Hippo pathway, and is a transcriptional co-activator that mainly interacts with TEAD family transcription factors to promote gene expression. Alteration of gene expression by YAP leads to cell proliferation, apoptosis evasion, and also stem cell amplification. In this review, we mainly focus on YAP, discussing its regulation and mechanisms of action in the context of organ size control, tissue regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Chu Zhu
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou 310058, China
| | - Li Li
- Institute of Aging Research, Hangzhou Normal University, Hangzhou 311121, China
| | - Bin Zhao
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
339
|
Imajo M, Ebisuya M, Nishida E. Dual role of YAP and TAZ in renewal of the intestinal epithelium. Nat Cell Biol 2014; 17:7-19. [DOI: 10.1038/ncb3084] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023]
|
340
|
Zhang H, Li C, Chen H, Wei C, Dai F, Wu H, Dui W, Deng WM, Jiao R. SCF(Slmb) E3 ligase-mediated degradation of Expanded is inhibited by the Hippo pathway in Drosophila. Cell Res 2014; 25:93-109. [PMID: 25522691 DOI: 10.1038/cr.2014.166] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/13/2014] [Accepted: 11/18/2014] [Indexed: 12/12/2022] Open
Abstract
Deregulation of the evolutionarily conserved Hippo pathway has been implicated in abnormal development of animals and in several types of cancer. One mechanism of Hippo pathway regulation is achieved by controlling the stability of its regulatory components. However, the executive E3 ligases that are involved in this process, and how the process is regulated, remain poorly defined. In this study, we identify, through a genetic candidate screen, the SCF(Slmb) E3 ligase as a novel negative regulator of the Hippo pathway in Drosophila imaginal tissues via mediation of the degradation of Expanded (Ex). Mechanistic study shows that Slmb-mediated degradation of Ex is inhibited by the Hippo signaling. Considering the fact that Hippo signaling suppresses the transcription of ex, we propose that the Hippo pathway employs a double security mechanism to ensure fine-tuned homeostasis during development.
Collapse
Affiliation(s)
- Hongtao Zhang
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100080, China
| | - Changqing Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China
| | - Hanqing Chen
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100080, China
| | - Chuanxian Wei
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100080, China
| | - Fei Dai
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100080, China
| | - Honggang Wu
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100080, China
| | - Wen Dui
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] University of Chinese Academy of Sciences, Beijing 100080, China
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, Florida 32304-4295, USA
| | - Renjie Jiao
- 1] State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, the Chinese Academy of Sciences, Datun Road 15, Beijing 100101, China [2] Guangzhou Hoffmann Institute of Immunology, School of Basic Sciences, Guangzhou Medical University, Dongfengxi Road 195, Guangzhou, Guangdong 510182, China
| |
Collapse
|
341
|
Piccolo S, Dupont S, Cordenonsi M. The biology of YAP/TAZ: hippo signaling and beyond. Physiol Rev 2014; 94:1287-312. [PMID: 25287865 DOI: 10.1152/physrev.00005.2014] [Citation(s) in RCA: 1286] [Impact Index Per Article: 116.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The transcriptional regulators YAP and TAZ are the focus of intense interest given their remarkable biological properties in development, tissue homeostasis and cancer. YAP and TAZ activity is key for the growth of whole organs, for amplification of tissue-specific progenitor cells during tissue renewal and regeneration, and for cell proliferation. In tumors, YAP/TAZ can reprogram cancer cells into cancer stem cells and incite tumor initiation, progression and metastasis. As such, YAP/TAZ are appealing therapeutic targets in cancer and regenerative medicine. Just like the function of YAP/TAZ offers a molecular entry point into the mysteries of tissue biology, their regulation by upstream cues is equally captivating. YAP/TAZ are well known for being the effectors of the Hippo signaling cascade, and mouse mutants in Hippo pathway components display remarkable phenotypes of organ overgrowth, enhanced stem cell content and reduced cellular differentiation. YAP/TAZ are primary sensors of the cell's physical nature, as defined by cell structure, shape and polarity. YAP/TAZ activation also reflects the cell "social" behavior, including cell adhesion and the mechanical signals that the cell receives from tissue architecture and surrounding extracellular matrix (ECM). At the same time, YAP/TAZ entertain relationships with morphogenetic signals, such as Wnt growth factors, and are also regulated by Rho, GPCRs and mevalonate metabolism. YAP/TAZ thus appear at the centerpiece of a signaling nexus by which cells take control of their behavior according to their own shape, spatial location and growth factor context.
Collapse
Affiliation(s)
- Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy
| | | |
Collapse
|
342
|
Del Re DP. The hippo signaling pathway: implications for heart regeneration and disease. Clin Transl Med 2014; 3:27. [PMID: 26932373 PMCID: PMC4884045 DOI: 10.1186/s40169-014-0027-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/22/2014] [Indexed: 12/12/2022] Open
Abstract
Control of cell number and organ size is critical for appropriate development and tissue homeostasis. Studies in both Drosophila and mammals have established the Hippo signaling pathway as an important modulator of organ size and tumorigenesis. Upon activation, this kinase cascade modulates gene expression through the phosphorylation and inhibition of transcription co-activators that are involved in cell proliferation, differentiation, growth and apoptosis. Hippo signaling serves to limit organ size and suppress malignancies, and has been implicated in tissue regeneration following injury. These outcomes highlight the important role that Hippo signaling plays in regulating both physiologic and pathologic processes. In this review, an overview of the signaling pathway will be discussed as well as recent work that has investigated its role in cardiac development, regeneration and disease.
Collapse
Affiliation(s)
- Dominic P Del Re
- Cardiovascular Research Institute and Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Newark, 07103, NJ, USA.
| |
Collapse
|
343
|
The Hippo pathway as a target of the Drosophila DRE/DREF transcriptional regulatory pathway. Sci Rep 2014; 4:7196. [PMID: 25424907 PMCID: PMC4244634 DOI: 10.1038/srep07196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/04/2014] [Indexed: 01/08/2023] Open
Abstract
The DRE/DREF transcriptional regulatory system has been demonstrated to activate a wide variety of genes with various functions. In Drosophila, the Hippo pathway is known to suppress cell proliferation by inducing apoptosis and cell cycle arrest through inactivation of Yorkie, a transcription co-activator. In the present study, we found that half dose reduction of the hippo (hpo) gene induces ectopic DNA synthesis in eye discs that is suppressed by overexpression of DREF. Half reduction of the hpo gene dose reduced apoptosis in DREF-overexpressing flies. Consistent with these observations, overexpression of DREF increased the levels of hpo and phosphorylated Yorkie in eye discs. Interestingly, the diap1-lacZ reporter was seen to be significantly decreased by overexpression of DREF. Luciferase reporter assays in cultured S2 cells revealed that one of two DREs identified in the hpo gene promoter region was responsible for promoter activity in S2 cells. Furthermore, endogenous hpo mRNA was reduced in DREF knockdown S2 cells, and chromatin immnunoprecipitation assays with anti-DREF antibodies proved that DREF binds specifically to the hpo gene promoter region containing DREs in vivo. Together, these results indicate that the DRE/DREF pathway is required for transcriptional activation of the hpo gene to positively control Hippo pathways.
Collapse
|
344
|
Milton CC, Grusche FA, Degoutin JL, Yu E, Dai Q, Lai EC, Harvey KF. The Hippo pathway regulates hematopoiesis in Drosophila melanogaster. Curr Biol 2014; 24:2673-80. [PMID: 25454587 PMCID: PMC4269548 DOI: 10.1016/j.cub.2014.10.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/26/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022]
Abstract
The Salvador-Warts-Hippo (Hippo) pathway is an evolutionarily conserved regulator of organ growth and cell fate. It performs these functions in epithelial and neural tissues of both insects and mammals, as well as in mammalian organs such as the liver and heart. Despite rapid advances in Hippo pathway research, a definitive role for this pathway in hematopoiesis has remained enigmatic. The hematopoietic compartments of Drosophila melanogaster and mammals possess several conserved features. D. melanogaster possess three types of hematopoietic cells that most closely resemble mammalian myeloid cells: plasmatocytes (macrophage-like cells), crystal cells (involved in wound healing), and lamellocytes (which encapsulate parasites). The proteins that control differentiation of these cells also control important blood lineage decisions in mammals. Here, we define the Hippo pathway as a key mediator of hematopoiesis by showing that it controls differentiation and proliferation of the two major types of D. melanogaster blood cells, plasmatocytes and crystal cells. In animals lacking the downstream Hippo pathway kinase Warts, lymph gland cells overproliferated, differentiated prematurely, and often adopted a mixed lineage fate. The Hippo pathway regulated crystal cell numbers by both cell-autonomous and non-cell-autonomous mechanisms. Yorkie and its partner transcription factor Scalloped were found to regulate transcription of the Runx family transcription factor Lozenge, which is a key regulator of crystal cell fate. Further, Yorkie or Scalloped hyperactivation induced ectopic crystal cells in a non-cell-autonomous and Notch-pathway-dependent fashion.
Collapse
Affiliation(s)
- Claire C Milton
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Felix A Grusche
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Joffrey L Degoutin
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Eefang Yu
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Qi Dai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Eric C Lai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Kieran F Harvey
- Cell Growth and Proliferation Laboratory, Peter MacCallum Cancer Centre, 7 St. Andrews Place, East Melbourne, VIC 3002, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
345
|
Zhou Z, Hu T, Xu Z, Lin Z, Zhang Z, Feng T, Zhu L, Rong Y, Shen H, Luk JM, Zhang X, Qin N. Targeting Hippo pathway by specific interruption of YAP-TEAD interaction using cyclic YAP-like peptides. FASEB J 2014; 29:724-32. [PMID: 25384421 DOI: 10.1096/fj.14-262980] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hippo signaling pathway is emerging as a novel target for anticancer therapy because it plays key roles in organ size control and tumorigenesis. As the downstream effectors, Yes-associated protein (YAP)-transcriptional enhancer activation domain family member (TEAD) association is essential for YAP-driven oncogenic activity, while TEAD is largely dispensable for normal tissue growth. We present the design of YAP-like peptides (17mer) to occupy the interface 3 on TEAD. Introducing cysteines at YAP sites 87 and 96 can induce disulfide formation, as confirmed by crystallography. The engineered peptide significantly improves the potency in disrupting YAP-TEAD interaction in vitro. To confirm that blocking YAP-TEAD complex formation by directly targeting on TEAD is a valid approach, we report a significant reduction in tumor growth rate in a hepatocellular carcinoma xenograft model after introducing the dominant-negative mutation (Y406H) of TEAD1 to abolish YAP-TEAD interaction. Our results suggest that targeting TEAD is a promising strategy against YAP-induced oncogenesis.
Collapse
Affiliation(s)
- Zheng Zhou
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Taishan Hu
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Zhiheng Xu
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Zhaohu Lin
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Zhisen Zhang
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Teng Feng
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Liangcheng Zhu
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Yiping Rong
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Hong Shen
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - John M Luk
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Xiongwen Zhang
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| | - Ning Qin
- *Discovery Technology, Medicinal Chemistry, and Discovery Oncology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai, China
| |
Collapse
|
346
|
Ubiquitin E3 ligase dSmurf is essential for Wts protein turnover and Hippo signaling. Biochem Biophys Res Commun 2014; 454:167-71. [DOI: 10.1016/j.bbrc.2014.10.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 10/13/2014] [Indexed: 01/25/2023]
|
347
|
Ferguson GB, Martinez-Agosto JA. Yorkie and Scalloped signaling regulates Notch-dependent lineage specification during Drosophila hematopoiesis. Curr Biol 2014; 24:2665-72. [PMID: 25454586 DOI: 10.1016/j.cub.2014.09.081] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/25/2014] [Accepted: 09/01/2014] [Indexed: 01/02/2023]
Abstract
Cellular microenvironments established by the spatial and temporal expression of specific signaling molecules are critical for both the maintenance and lineage-specific differentiation of progenitor cells. In Drosophila, a population of hematopoietic progenitors, or prohemocytes, within the larval lymph gland gives rise to three mature cell types: plasmatocytes, lamellocytes, and crystal cells. Removal of the secreted signaling molecules Hedgehog and PVF1 from the posterior signaling center (PSC), which acts as a niche, leads to a loss of progenitors and complete differentiation of the lymph gland. Here, we characterize a novel population of signaling cells within the lymph gland, distinct from the PSC, that are required for lineage-specific differentiation of crystal cells. We provide evidence that Yorkie and Scalloped, the Drosophila homologs of YAP and TEAD, are required in lineage-specifying cells to regulate expression of Serrate, the Notch ligand responsible for the initiation of the crystal cell differentiation program. Genetic manipulation of yorkie and scalloped in the lymph gland specifically alters Serrate expression and crystal cell differentiation. Furthermore, Serrate expression in lineage-specifying cells is eliminated in the lymph gland upon the immune response induced by wasp parasitization to ensure the proper differentiation of lamellocytes at the expense of crystal cells. These findings expand the roles for Yorkie/Scalloped beyond growth to encompass specific cell-fate determination in the context of blood development. Similar regulatory functions may extend to their homologs in vertebrate progenitor cell niches that are required for specifying cell fate.
Collapse
Affiliation(s)
- Gabriel B Ferguson
- Molecular Biology Interdepartmental Ph.D. Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julian A Martinez-Agosto
- Molecular Biology Interdepartmental Ph.D. Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, Jonsson Comprehensive Cancer Center, UCLA Broad Stem Cell Center, and Mattel Children's Hospital UCLA, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
348
|
Kwon HJ, Waghmare I, Verghese S, Singh A, Singh A, Kango-Singh M. Drosophila C-terminal Src kinase regulates growth via the Hippo signaling pathway. Dev Biol 2014; 397:67-76. [PMID: 25446534 DOI: 10.1016/j.ydbio.2014.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 09/15/2014] [Accepted: 10/14/2014] [Indexed: 12/11/2022]
Abstract
The Hippo signaling pathway is involved in regulating tissue size by inhibiting cell proliferation and promoting apoptosis. Aberrant Hippo pathway function is often detected in human cancers and correlates with poor prognosis. The Drosophila C-terminal Src kinase (d-Csk) is a genetic modifier of warts (wts), a tumor-suppressor gene in the Hippo pathway, and interacts with the Src oncogene. Reduction in d-Csk expression and the consequent activation of Src are frequently seen in several cancers including hepatocellular and colorectal tumors. Previous studies show that d-Csk regulates cell proliferation and tissue size during development. Given the similarity in the loss-of-function phenotypes of d-Csk and wts, we have investigated the interactions of d-Csk with the Hippo pathway. Here we present multiple lines of evidence suggesting that d-Csk regulates growth via the Hippo signaling pathway. We show that loss of dCsk caused increased Yki activity, and our genetic epistasis places dCsk downstream of Dachs. Furthermore, dCsk requires Yki for its growth regulatory functions, suggesting that dCsk is another upstream member of the network of genes that interact to regulate Wts and its effector Yki in the Hippo signaling pathway.
Collapse
Affiliation(s)
- Hailey J Kwon
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | | | - Shilpi Verghese
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
| | - Aditi Singh
- Centerville High School, Centerville, OH 45459, USA
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; Center for Tissue Regeneration and Engineering at Dayton, Dayton, OH 45469, USA; Premedical Programs, University of Dayton, Dayton, OH 45469, USA
| | - Madhuri Kango-Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; Center for Tissue Regeneration and Engineering at Dayton, Dayton, OH 45469, USA; Premedical Programs, University of Dayton, Dayton, OH 45469, USA.
| |
Collapse
|
349
|
Hu L, Huang H, Li J, Yin MX, Lu Y, Wu W, Zeng R, Jiang J, Zhao Y, Zhang L. Drosophila casein kinase 2 (CK2) promotes warts protein to suppress Yorkie protein activity for growth control. J Biol Chem 2014; 289:33598-607. [PMID: 25320084 DOI: 10.1074/jbc.m114.580456] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Drosophila Hippo signaling regulates Wts activity to phosphorylate and inhibit Yki in order to control tissue growth. CK2 is widely expressed and involved in a variety of signaling pathways. In this study we report that Drosophila CK2 promotes Wts activity to phosphorylate and inhibit Yki activity, which is independent of Hpo-induced Wts promotion. In vivo, CK2 overexpression suppresses hpo mutant-induced expanded (Ex) up-regulation and overgrowth phenotype, whereas it cannot affect wts mutant. Consistent with this, knockdown of CK2 up-regulates Hpo pathway target expression. We also found that Drosophila CK2 is essential for tissue growth as a cell death inhibitor as knockdown of CK2 in the developing disc induces severe growth defects as well as caspase3 signals. Taken together, our results uncover a dual role of CK2; although its major role is promoting cell survive, it may potentially be a growth inhibitor as well.
Collapse
Affiliation(s)
- Lianxin Hu
- From the State Key Laboratory of Cell Biology and
| | | | - Jinhui Li
- From the State Key Laboratory of Cell Biology and
| | - Meng-Xin Yin
- From the State Key Laboratory of Cell Biology and
| | - Yi Lu
- From the State Key Laboratory of Cell Biology and
| | - Wenqing Wu
- From the State Key Laboratory of Cell Biology and
| | - Rong Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Yue-Yang Road, Shanghai 200031, China and
| | - Jin Jiang
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Yun Zhao
- From the State Key Laboratory of Cell Biology and
| | - Lei Zhang
- From the State Key Laboratory of Cell Biology and
| |
Collapse
|
350
|
Yu W, Qiao Y, Tang X, Ma L, Wang Y, Zhang X, Weng W, Pan Q, Yu Y, Sun F, Wang J. Tumor suppressor long non-coding RNA, MT1DP is negatively regulated by YAP and Runx2 to inhibit FoxA1 in liver cancer cells. Cell Signal 2014; 26:2961-8. [PMID: 25261601 DOI: 10.1016/j.cellsig.2014.09.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/05/2014] [Indexed: 02/08/2023]
Abstract
Recent studies are indicative for strong carcinogenetic roles of Runt related transcription factor 2 (Runx2) and Yes associated protein (YAP) in several cancer types. However, whether and how the interaction between Runx2 and YAP plays a role in liver tumorigenesis still remain illusive. Here, we identified a close relationship between Runx2 and YAP in liver cancer cells. Runx2 had a positive role on YAP expression and vice versa. We also found that Rux2 and YAP were capable of inhibiting long non-coding RNA (lncRNA), Metallothionein 1D, Pseudogene (MT1DP) expression through direct promoter binding. Overexpression of MT1DP resulted in reduced cell proliferation and colony formation in soft agar, but increased apoptosis in liver cancer cells, whereas knockdown of this lncRNA had the opposite effect, indicating that MT1DP acts as a tumor suppressor. Furthermore, MT1DP was revealed as a negative regulator of Alfa-fetoprotein (AFP), a classic liver cancer tumor marker, through inhibiting protein synthesis of Forkhead box A1 (FoxA1), an important transcription factor in liver development and cancer progression. Furthermore, we found that FoxA1 plays a positive role on YAP and Runx2 expression. Specially, opening the compacted chromatin by FoxA1 around CREB binding site within the YAP promoter facilitates CREB-mediated YAP transcription. Finally, MT1DP-inhibited in vivo liver cancer cell growth could be rescued by a combination of overexpression of FoxA1, Runx2 and YAP. Taken together, the close relationship between Rnux2 and YAP plays a pro-carcinogenetic role in liver cancer cells through inhibiting tumor suppressor lncRNA, MT1DP in a FoxA1 dependent manner.
Collapse
Affiliation(s)
- Wenjun Yu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yongxia Qiao
- School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Xun Tang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yulan Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Xiao Zhang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Wenhao Weng
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Qiuhui Pan
- Department of Central Laboratory, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yongchun Yu
- Shanghai Municipal Hospital of Traditional Chinese Medicine affiliated to Shanghai TCM University, Shanghai 200071, China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China.
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China.
| |
Collapse
|