101
|
Li Q, Nirala NK, Chen HJ, Nie Y, Wang W, Zhang B, Czech MP, Wang Q, Xu L, Mao J, Tony Ip Y. The Misshapen subfamily of Ste20 kinases regulate proliferation in the aging mammalian intestinal epithelium. J Cell Physiol 2019; 234:21925-21936. [PMID: 31042012 PMCID: PMC6711781 DOI: 10.1002/jcp.28756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/01/2019] [Accepted: 04/05/2019] [Indexed: 12/17/2022]
Abstract
The intestinal epithelium has a high rate of cell turn over and is an excellent system to study stem cell-mediated tissue homeostasis. The Misshapen subfamily of the Ste20 kinases in mammals consists of misshapen like kinase 1 (MINK1), mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), and TRAF2 and NCK interacting kinase (TNIK). Recent reports suggest that this subfamily has a novel function equal to the Hippo/MST subfamily as upstream kinases for Warts/Large tumor suppressor kinase (LATS) to suppress tissue growth. To study the in vivo functions of Mink1, Map4k4, and Tnik, we generated a compound knockout of these three genes in the mouse intestinal epithelium. The intestinal epithelia of the mutant animals were phenotypically normal up to approximately 12 months. The older animals then exhibited mildly increased proliferation throughout the lower GI tract. We also observed that the normally spatially organized Paneth cells in the crypt base became dispersed. The expression of one of the YAP pathway target genes Sox9 was increased while other target genes including CTGF did not show a significant change. Therefore, the Misshapen and Hippo subfamilies may have highly redundant functions to regulate growth in the intestinal epithelium, as illustrated in recent tissue culture models.
Collapse
Affiliation(s)
- Qi Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Niraj K. Nirala
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hsi-Ju Chen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yingchao Nie
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Wei Wang
- Guangzhou RiboBio Co., Ltd., Guangzhou 510663, China
| | - Biliang Zhang
- Guangzhou RiboBio Co., Ltd., Guangzhou 510663, China
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Michael P. Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Qi Wang
- Neuroscience Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Lan Xu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Y. Tony Ip
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
102
|
Fearing BV, Jing L, Barcellona MN, Witte SE, Buchowski JM, Zebala LP, Kelly MP, Luhmann S, Gupta MC, Pathak A, Setton LA. Mechanosensitive transcriptional coactivators MRTF-A and YAP/TAZ regulate nucleus pulposus cell phenotype through cell shape. FASEB J 2019; 33:14022-14035. [PMID: 31638828 PMCID: PMC6894097 DOI: 10.1096/fj.201802725rrr] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 09/17/2019] [Indexed: 01/05/2023]
Abstract
Cells of the adult nucleus pulposus (NP) are critically important in maintaining overall disc health and function. NP cells reside in a soft, gelatinous matrix that dehydrates and becomes increasingly fibrotic with age. Such changes result in physical cues of matrix stiffness that may be potent regulators of NP cell phenotype and may contribute to a transition toward a senescent and fibroblastic NP cell with a limited capacity for repair. Here, we investigate the mechanosignaling cues generated from changes in matrix stiffness in directing NP cell phenotype and identify mechanisms that can potentially preserve a biosynthetically active, juvenile NP cell phenotype. Using a laminin-functionalized polyethylene glycol hydrogel, we show that when NP cells form rounded, multicell clusters, they are able to maintain cytosolic localization of myocardin-related transcription factor (MRTF)-A, a coactivator of serum-response factor (SRF), known to promote fibroblast-like behaviors in many cells. Upon preservation of a rounded shape, human NP cells similarly showed cytosolic retention of transcriptional coactivator Yes-associated protein (YAP) and its paralogue PDZ-binding motif (TAZ) with associated decline in activation of its transcription factor TEA domain family member-binding domain (TEAD). When changes in cell shape occur, leading to a more spread, fibrotic morphology associated with stronger F-actin alignment, SRF and TEAD are up-regulated. However, targeted deletion of either cofactor was not sufficient to overcome shape-mediated changes observed in transcriptional activation of SRF or TEAD. Findings show that substrate stiffness-induced promotion of F-actin alignment occurs concomitantly with a flattened, spread morphology, decreased NP marker expression, and reduced biosynthetic activity. This work indicates cell shape is a stronger indicator of SRF and TEAD mechanosignaling pathways than coactivators MRTF-A and YAP/TAZ, respectively, and may play a role in the degeneration-associated loss of NP cellularity and phenotype.-Fearing, B. V., Jing, L., Barcellona, M. N., Witte, S. E., Buchowski, J. M., Zebala, L. P., Kelly, M. P., Luhmann, S., Gupta, M. C., Pathak, A., Setton, L. A. Mechanosensitive transcriptional coactivators MRTF-A and YAP/TAZ regulate nucleus pulposus cell phenotype through cell shape.
Collapse
Affiliation(s)
- Bailey V. Fearing
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Marcos N. Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Savannah Est Witte
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jacob M. Buchowski
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lukas P. Zebala
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael P. Kelly
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Scott Luhmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Munish C. Gupta
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Amit Pathak
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lori A. Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
103
|
Sidor C, Borreguero-Munoz N, Fletcher GC, Elbediwy A, Guillermin O, Thompson BJ. Mask family proteins ANKHD1 and ANKRD17 regulate YAP nuclear import and stability. eLife 2019; 8:e48601. [PMID: 31661072 PMCID: PMC6861002 DOI: 10.7554/elife.48601] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Mask family proteins were discovered in Drosophila to promote the activity of the transcriptional coactivator Yorkie (Yki), the sole fly homolog of mammalian YAP (YAP1) and TAZ (WWTR1). The molecular function of Mask, or its mammalian homologs Mask1 (ANKHD1) and Mask2 (ANKRD17), remains unclear. Mask family proteins contain two ankyrin repeat domains that bind Yki/YAP as well as a conserved nuclear localisation sequence (NLS) and nuclear export sequence (NES), suggesting a role in nucleo-cytoplasmic transport. Here we show that Mask acts to promote nuclear import of Yki, and that addition of an ectopic NLS to Yki is sufficient to bypass the requirement for Mask in Yki-driven tissue growth. Mammalian Mask1/2 proteins also promote nuclear import of YAP, as well as stabilising YAP and driving formation of liquid droplets. Mask1/2 and YAP normally colocalise in a granular fashion in both nucleus and cytoplasm, and are co-regulated during mechanotransduction.
Collapse
Affiliation(s)
- Clara Sidor
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom
| | | | | | - Ahmed Elbediwy
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom
| | - Oriane Guillermin
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom
| | - Barry J Thompson
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUnited Kingdom
- EMBL Australia, ACRF Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical Research, The Australian National UniversityCanberraAustralia
| |
Collapse
|
104
|
Wang HY, Long QY, Tang SB, Xiao Q, Gao C, Zhao QY, Li QL, Ye M, Zhang L, Li LY, Wu M. Histone demethylase KDM3A is required for enhancer activation of hippo target genes in colorectal cancer. Nucleic Acids Res 2019; 47:2349-2364. [PMID: 30649550 PMCID: PMC6412006 DOI: 10.1093/nar/gky1317] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/18/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
Hippo pathway is involved in tumorigenesis, and its regulation in cytosol has been extensively studied, but its regulatory mechanisms in the nuclear are not clear. In the current study, using a FBS-inducing model following serum starvation, we identified KDM3A, a demethylase of histone H3K9me1/2, as a positive regulator for hippo target genes. KDM3A promotes gene expression through two mechanisms, one is to upregulate YAP1 expression, and the other is to facilitate H3K27ac on the enhancers of hippo target genes. H3K27ac upregulation is more relevant with gene activation, but not H3K4me3; and KDM3A depletion caused H3K9me2 upregulation mainly on TEAD1-binding enhancers rather than gene bodies, further resulting in H3K27ac decrease, less TEAD1 binding on enhancers and impaired transcription. Moreover, KDM3A is associated with p300 and required for p300 recruitment to enhancers. KDM3A deficiency delayed cancer cell growth and migration, which was rescued by YAP1 expression. KDM3A expression is correlated with YAP1 and hippo target genes in colorectal cancer patient tissues, and may serve as a potential prognosis mark. Taken together, our study reveals novel mechanisms for hippo signaling and enhancer activation, which is critical for tumorigenesis of colorectal cancer.
Collapse
Affiliation(s)
- Hui-Yi Wang
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Qiao-Yun Long
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shan-Bo Tang
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Qiong Xiao
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Chuan Gao
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Quan-Yi Zhao
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Qing-Lan Li
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Mei Ye
- Division of Gastroenterology, Department of Geriatrics, Hubei Clinical Centre & Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430072, China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Lian-Yun Li
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Min Wu
- Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Intestinal and Colorectal Diseases, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
105
|
Borreguero-Muñoz N, Fletcher GC, Aguilar-Aragon M, Elbediwy A, Vincent-Mistiaen ZI, Thompson BJ. The Hippo pathway integrates PI3K-Akt signals with mechanical and polarity cues to control tissue growth. PLoS Biol 2019; 17:e3000509. [PMID: 31613895 PMCID: PMC6814241 DOI: 10.1371/journal.pbio.3000509] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 10/25/2019] [Accepted: 10/03/2019] [Indexed: 11/19/2022] Open
Abstract
The Hippo signalling pathway restricts cell proliferation in animal tissues by inhibiting Yes-associated protein (YAP or YAP1) and Transcriptional Activator with a PDZ domain (TAZ or WW-domain-containing transcriptional activator [WWTR1]), coactivators of the Scalloped (Sd or TEAD) DNA-binding transcription factor. Drosophila has a single YAP/TAZ homolog named Yorkie (Yki) that is regulated by Hippo pathway signalling in response to epithelial polarity and tissue mechanics during development. Here, we show that Yki translocates to the nucleus to drive Sd-mediated cell proliferation in the ovarian follicle cell epithelium in response to mechanical stretching caused by the growth of the germline. Importantly, mechanically induced Yki nuclear localisation also requires nutritionally induced insulin/insulin-like growth factor 1 (IGF-1) signalling (IIS) via phosphatidyl inositol-3-kinase (PI3K), phosphoinositide-dependent kinase 1 (PDK1 or PDPK1), and protein kinase B (Akt or PKB) in the follicular epithelium. We find similar results in the developing Drosophila wing, where Yki becomes nuclear in the mechanically stretched cells of the wing pouch during larval feeding, which induces IIS, but translocates to the cytoplasm upon cessation of feeding in the third instar stage. Inactivating Akt prevents nuclear Yki localisation in the wing disc, while ectopic activation of the insulin receptor, PI3K, or Akt/PKB is sufficient to maintain nuclear Yki in mechanically stimulated cells of the wing pouch even after feeding ceases. Finally, IIS also promotes YAP nuclear localisation in response to mechanical cues in mammalian skin epithelia. Thus, the Hippo pathway has a physiological function as an integrator of epithelial cell polarity, tissue mechanics, and nutritional cues to control cell proliferation and tissue growth in both Drosophila and mammals.
Collapse
Affiliation(s)
| | - Georgina C. Fletcher
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Mario Aguilar-Aragon
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ahmed Elbediwy
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Barry J. Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
- EMBL Australia, Department of Cancer Biology & Therapeutics, The John Curtin School of Medical Research, The Australian National University, Acton, Australia
- * E-mail:
| |
Collapse
|
106
|
Seo J, Kim MH, Hong H, Cho H, Park S, Kim SK, Kim J. MK5 Regulates YAP Stability and Is a Molecular Target in YAP-Driven Cancers. Cancer Res 2019; 79:6139-6152. [DOI: 10.1158/0008-5472.can-19-1339] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/21/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
|
107
|
Yee WB, Delaney PM, Vanderzalm PJ, Ramachandran S, Fehon RG. The CAF-1 complex couples Hippo pathway target gene expression and DNA replication. Mol Biol Cell 2019; 30:2929-2942. [PMID: 31553691 PMCID: PMC6822585 DOI: 10.1091/mbc.e19-07-0387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Hippo signaling pathway regulates tissue growth and organ development in many animals, including humans. Pathway activity leads to inactivation of Yorkie (Yki), a transcriptional coactivator that drives expression of growth-promoting genes. In addition, Yki has been shown to recruit chromatin modifiers that enhance chromatin accessibility and thereby enhance Yki function. Here, we asked whether changes in chromatin accessibility that occur during DNA replication could also affect Yki function. We found that depletion of the chromatin assembly complex-1 (CAF-1) complex, a histone chaperone that is required for nucleosome assembly after DNA replication, in the wing imaginal epithelium leads to increased Hippo pathway target gene expression but does not affect expression of other genes. Yki shows greater association with target sites when CAF-1 is depleted and misregulation of target gene expression is Yki-dependent, suggesting that nucleosome assembly competes with Yki for pathway targets post-DNA replication. Consistent with this idea, increased target gene expression is DNA replication dependent and newly replicated chromatin at target sites shows marked nucleosome depletion when CAF-1 function is reduced. These observations suggest a connection between cell cycle progression and Hippo pathway target expression, providing insights into functions of the Hippo pathway in normal and abnormal tissue growth.
Collapse
Affiliation(s)
- William B Yee
- Department of Molecular Genetics and Cell Biology.,Graduate Program in Cell and Molecular Biology, and
| | | | - Pamela J Vanderzalm
- Department of Molecular Genetics and Cell Biology.,Department of Biology, John Carroll University, University Heights, OH 44118
| | - Srinivas Ramachandran
- RNA Bioscience Initiative and.,Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology.,Graduate Program in Cell and Molecular Biology, and
| |
Collapse
|
108
|
Walsh T. Editor’s Pick: Systemic Sclerosis: The Role of YAP/TAZ in Disease Pathogenesis. EUROPEAN MEDICAL JOURNAL 2019. [DOI: 10.33590/emj/10310340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Systemic sclerosis (SSc) is a systemic autoimmune condition of unknown cause. Yes-Associated Protein/Tafazzin (YAP/TAZ) are transcriptional coactivators previously demonstrated to be involved in cellular stretch biology, and form the principal effector molecules of the Hippo signalling pathway. The association between YAP/TAZ and stretch is contingent upon their cytoplasmic localisation (with nuclear translocation, the cell adopts a relaxed state). The author weighs the evidence for a central role for YAP/TAZ signalling in scleroderma spanning the major clinical features of the condition. Several of the features unique to SSc are mediated by cytoplasmic localisation of YAP/TAZ, including the stretch phenotype (through binding to NF-2), arterial lumenal obliteration (through their binding to angiomotin), the promotion of hypergammaglobulinaemia (via feedback to the upstream Hippo signalling molecule Mammalian Ste20-like Kinase 1), and the induction of B-Lymphocyte-Induced Maturation Protein-1 leading to the adoption of Th2 lineage, prominent in SSc. One observes that the induction of the fibrotic phenotype of scleroderma is mediated through GLI1/GLI2 (the effector molecules of the Hedgehog pathway). GLI1/GLI2 are induced to reciprocally enter the nucleus when YAP/TAZ is intracytoplasmic. The latter explains the characteristically increased connective tissue growth factor 2 and endothelin-1 expression. In this article, the author references some examples of the role of YAP/TAZ in the biophysically similar condition nephrogenic systemic fibrosis and suggests a role of YAP/TAZ cytoplasmic sequestration in programmed cell death protein 1-ligand antagonist-induced scleroderma.
Collapse
Affiliation(s)
- Thomas Walsh
- Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| |
Collapse
|
109
|
Berry KP, Lu QR. Chromatin modification and epigenetic control in functional nerve regeneration. Semin Cell Dev Biol 2019; 97:74-83. [PMID: 31301357 DOI: 10.1016/j.semcdb.2019.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
The repair and functional recovery of the nervous system is a highly regulated process that requires the coordination of many different components including the proper myelination of regenerated axons. Dysmyelination and remyelination failures after injury result in defective nerve conduction, impairing normal nervous system functions. There are many convergent regulatory networks and signaling mechanisms between development and regeneration. For instance, the regulatory mechanisms required for oligodendrocyte lineage progression could potentially play fundamental roles in myelin repair. In recent years, epigenetic chromatin modifications have been implicated in CNS myelination and functional nerve restoration. The pro-regenerative transcriptional program is likely silenced or repressed in adult neural cells including neurons and myelinating cells in the central and peripheral nervous systems limiting the capacity for repair after injury. In this review, we will discuss the roles of epigenetic mechanisms, including histone modifications, chromatin remodeling, and DNA methylation, in the maintenance and establishment of the myelination program during normal oligodendrocyte development and regeneration. We also discuss how these epigenetic processes impact myelination and axonal regeneration, and facilitate the improvement of current preclinical therapeutics for functional nerve regeneration in neurodegenerative disorders or after injury.
Collapse
Affiliation(s)
- Kalen P Berry
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
110
|
Zheng Y, Pan D. The Hippo Signaling Pathway in Development and Disease. Dev Cell 2019; 50:264-282. [PMID: 31386861 PMCID: PMC6748048 DOI: 10.1016/j.devcel.2019.06.003] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/23/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway regulates diverse physiological processes, and its dysfunction has been implicated in an increasing number of human diseases, including cancer. Here, we provide an updated review of the Hippo pathway; discuss its roles in development, homeostasis, regeneration, and diseases; and highlight outstanding questions for future investigation and opportunities for Hippo-targeted therapies.
Collapse
Affiliation(s)
- Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.
| |
Collapse
|
111
|
Feng X, Wang Z, Wang F, Lu T, Xu J, Ma X, Li J, He L, Zhang W, Li S, Yang W, Zhang S, Ge G, Zhao Y, Hu P, Zhang L. Dual function of VGLL4 in muscle regeneration. EMBO J 2019; 38:e101051. [PMID: 31328806 DOI: 10.15252/embj.2018101051] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/17/2019] [Accepted: 06/28/2019] [Indexed: 01/07/2023] Open
Abstract
VGLL4 has previously been identified as a negative regulator of YAP. Here we show that VGLL4 regulates muscle regeneration in both YAP-dependent and YAP-independent manners at different stages. Knockout of VGLL4 in mice leads to smaller myofiber size and defective muscle contraction force. Furthermore, our studies reveal that knockout of VGLL4 results in increased muscle satellite cells proliferation and impaired myoblast differentiation, which ultimately leads to delayed muscle regeneration. Mechanistically, the results show that VGLL4 works as a conventional repressor of YAP at the proliferation stage of muscle regeneration. At the differentiation stage, VGLL4 acts as a co-activator of TEAD4 to promote MyoG transactivation and facilitate the initiation of differentiation in a YAP-independent manner. Moreover, VGLL4 stabilizes the protein-protein interactions between MyoD and TEAD4 to achieve efficient MyoG transactivation. Our findings define the dual roles of VGLL4 in regulating muscle regeneration at different stages and may open novel therapeutic perspectives for muscle regeneration.
Collapse
Affiliation(s)
- Xue Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zuoyun Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fei Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Tiantian Lu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jinjin Xu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xueyan Ma
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jinhui Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lingli He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenxiang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Sheng Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenjun Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shu Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Gaoxiang Ge
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yun Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ping Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
112
|
Figeac N, Mohamed AD, Sun C, Schönfelder M, Matallanas D, Garcia-Munoz A, Missiaglia E, Collie-Duguid E, De Mello V, Pobbati AV, Pruller J, Jaka O, Harridge SDR, Hong W, Shipley J, Vargesson N, Zammit PS, Wackerhage H. VGLL3 operates via TEAD1, TEAD3 and TEAD4 to influence myogenesis in skeletal muscle. J Cell Sci 2019; 132:jcs.225946. [PMID: 31138678 PMCID: PMC6633393 DOI: 10.1242/jcs.225946] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/03/2019] [Indexed: 12/21/2022] Open
Abstract
VGLL proteins are transcriptional co-factors that bind TEAD family transcription factors to regulate events ranging from wing development in fly, to muscle fibre composition and immune function in mice. Here, we characterise Vgll3 in skeletal muscle. We found that mouse Vgll3 was expressed at low levels in healthy muscle but that its levels increased during hypertrophy or regeneration; in humans, VGLL3 was highly expressed in tissues from patients with various muscle diseases, such as in dystrophic muscle and alveolar rhabdomyosarcoma. Interaction proteomics revealed that VGLL3 bound TEAD1, TEAD3 and TEAD4 in myoblasts and/or myotubes. However, there was no interaction with proteins from major regulatory systems such as the Hippo kinase cascade, unlike what is found for the TEAD co-factors YAP (encoded by YAP1) and TAZ (encoded by WWTR1). Vgll3 overexpression reduced the activity of the Hippo negative-feedback loop, affecting expression of muscle-regulating genes including Myf5, Pitx2 and Pitx3, and genes encoding certain Wnts and IGFBPs. VGLL3 mainly repressed gene expression, regulating similar genes to those regulated by YAP and TAZ. siRNA-mediated Vgll3 knockdown suppressed myoblast proliferation, whereas Vgll3 overexpression strongly promoted myogenic differentiation. However, skeletal muscle was overtly normal in Vgll3-null mice, presumably due to feedback signalling and/or redundancy. This work identifies VGLL3 as a transcriptional co-factor operating with the Hippo signal transduction network to control myogenesis. Summary: VGLL3 interacts with TEAD transcription factors to direct expression of crucial muscle regulatory genes and contribute to the control of skeletal myogenesis.
Collapse
Affiliation(s)
- Nicolas Figeac
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Abdalla D Mohamed
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health, Ingolstaedter Landstrasse 1, D-85764 Munich/Neuherberg, Germany
| | - Congshan Sun
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.,Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Martin Schönfelder
- Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany
| | - David Matallanas
- Systems Biology Ireland, Conway Institute, Belfield; Dublin 4, Ireland
| | | | - Edoardo Missiaglia
- Institute of Pathology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Elaina Collie-Duguid
- University of Aberdeen, Centre for Genome Enabled Biology and Medicine, 23 St Machar Drive, Aberdeen AB24 3RY, UK
| | - Vanessa De Mello
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Ajaybabu V Pobbati
- Institute of Molecular and Cell Biology, A-STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Johanna Pruller
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Oihane Jaka
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Stephen D R Harridge
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A-STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Janet Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Henning Wackerhage
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK .,Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany
| |
Collapse
|
113
|
Xie B, Morton DB, Cook TA. Opposing transcriptional and post-transcriptional roles for Scalloped in binary Hippo-dependent neural fate decisions. Dev Biol 2019; 455:51-59. [PMID: 31265830 DOI: 10.1016/j.ydbio.2019.06.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 01/07/2023]
Abstract
The Hippo tumor suppressor pathway plays many fundamental cell biological roles during animal development. Two central players in controlling Hippo-dependent gene expression are the TEAD transcription factor Scalloped (Sd) and its transcriptional co-activator Yorkie (Yki). Hippo signaling phosphorylates Yki, thereby blocking Yki-dependent transcriptional control. In post-mitotic Drosophila photoreceptors, a bistable negative feedback loop forms between the Hippo-dependent kinase Warts/Lats and Yki to lock in green vs blue-sensitive neuronal subtype choices, respectively. Previous experiments indicate that sd and yki mutants phenocopy each other's functions, both being required for promoting the expression of the blue photoreceptor fate determinant melted (melt) and the blue-sensitive opsin Rh5. Here, we demonstrate that Sd ensures the robustness of this neuronal fate decision via multiple antagonistic gene regulatory roles. In Hippo-positive (green) photoreceptors, Sd directly represses both melt and Rh5 gene expression through defined TEAD binding sites, a mechanism that is antagonized by Yki in Hippo-negative (blue) cells. Additionally, in blue photoreceptors, Sd is required to promote the translation of the Rh5 protein through a 3'UTR-dependent and microRNA-mediated process. Together, these studies reveal that Sd can drive context-dependent cell fate decisions through opposing transcriptional and post-transcriptional mechanisms.
Collapse
Affiliation(s)
- Baotong Xie
- Department of Integrative Biosciences, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - David B Morton
- Department of Integrative Biosciences, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Tiffany A Cook
- Center of Molecular Medicine and Genetics and Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
114
|
Huh HD, Kim DH, Jeong HS, Park HW. Regulation of TEAD Transcription Factors in Cancer Biology. Cells 2019; 8:E600. [PMID: 31212916 PMCID: PMC6628201 DOI: 10.3390/cells8060600] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Transcriptional enhanced associate domain (TEAD) transcription factors play important roles during development, cell proliferation, regeneration, and tissue homeostasis. TEAD integrates with and coordinates various signal transduction pathways including Hippo, Wnt, transforming growth factor beta (TGFβ), and epidermal growth factor receptor (EGFR) pathways. TEAD deregulation affects well-established cancer genes such as KRAS, BRAF, LKB1, NF2, and MYC, and its transcriptional output plays an important role in tumor progression, metastasis, cancer metabolism, immunity, and drug resistance. To date, TEADs have been recognized to be key transcription factors of the Hippo pathway. Therefore, most studies are focused on the Hippo kinases and YAP/TAZ, whereas the Hippo-dependent and Hippo-independent regulators and regulations governing TEAD only emerged recently. Deregulation of the TEAD transcriptional output plays important roles in tumor progression and serves as a prognostic biomarker due to high correlation with clinicopathological parameters in human malignancies. In addition, discovering the molecular mechanisms of TEAD, such as post-translational modifications and nucleocytoplasmic shuttling, represents an important means of modulating TEAD transcriptional activity. Collectively, this review highlights the role of TEAD in multistep-tumorigenesis by interacting with upstream oncogenic signaling pathways and controlling downstream target genes, which provides unprecedented insight and rationale into developing TEAD-targeted anticancer therapeutics.
Collapse
Affiliation(s)
- Hyunbin D Huh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| | - Dong Hyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| | - Han-Sol Jeong
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea.
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| |
Collapse
|
115
|
Hillmer RE, Link BA. The Roles of Hippo Signaling Transducers Yap and Taz in Chromatin Remodeling. Cells 2019; 8:E502. [PMID: 31137701 PMCID: PMC6562424 DOI: 10.3390/cells8050502] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/15/2019] [Accepted: 05/19/2019] [Indexed: 01/04/2023] Open
Abstract
Hippo signaling controls cellular processes that ultimately impact organogenesis and homeostasis. Consequently, disease states including cancer can emerge when signaling is deregulated. The major pathway transducers Yap and Taz require cofactors to impart transcriptional control over target genes. Research into Yap/Taz-mediated epigenetic modifications has revealed their association with chromatin-remodeling complex proteins as a means of altering chromatin structure, therefore affecting accessibility and activity of target genes. Specifically, Yap/Taz have been found to associate with factors of the GAGA, Ncoa6, Mediator, Switch/sucrose nonfermentable (SWI/SNF), and Nucleosome Remodeling and Deacetylase (NuRD) chromatin-remodeling complexes to alter the accessibility of target genes. This review highlights the different mechanisms by which Yap/Taz collaborate with other factors to modify DNA packing at specific loci to either activate or repress target gene transcription.
Collapse
Affiliation(s)
- Ryan E Hillmer
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | - Brian A Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
116
|
Phosphorylation of 14-3-3ζ links YAP transcriptional activation to hypoxic glycolysis for tumorigenesis. Oncogenesis 2019; 8:31. [PMID: 31076568 PMCID: PMC6510816 DOI: 10.1038/s41389-019-0143-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/24/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
Hypoxic microenvironment deregulates metabolic homeostasis in cancer cells albeit the underlying mechanisms involved in this process remain hitherto enigmatic. 14-3-3ζ/Yes-associated protein (YAP) axis plays a principal role in malignant transformation and tumor development. Here, we report that hypoxia disassembles 14-3-3ζ from YAP and thereby promotes YAP nuclear localization mediated by ERK2, which directly binds to the D-site of mitogen-activated protein kinase (MAPK) docking domain in 14-3-3ζ Leu98/100 and phosphorylates 14-3-3ζ at Ser37. When localizing in nucleus, YAP recruits at pyruvate kinase M2 (PKM2) gene promoter with hypoxia-inducible factor 1α (HIF-1α), for which PKM2 transcription is required. 14-3-3ζ Ser37 phosphorylation is instrumental for the hypoxia-induced glucose uptake, lactate production, and clonogenicity of pancreatic ductal adenocarcinoma (PDAC) cells, as well as tumorigenesis in mice. The 14-3-3ζ Ser37 phosphorylation positively correlates with p-ERK1/2 activity and HIF-1α expression in clinical samples from patients with PDAC and predicts unfavorable prognosis. Our findings underscore an appreciable linkage between YAP transcriptional activation and hypoxic glycolysis governed by ERK2-dependent 14-3-3ζ Ser37 phosphorylation for malignant progression of PDAC.
Collapse
|
117
|
Abstract
The Hippo signaling pathway is involved in tissue size regulation and tumorigenesis. Genetic deletion or aberrant expression of some Hippo pathway genes lead to enhanced cell proliferation, tumorigenesis, and cancer metastasis. Recently, multiple studies have identified a wide range of upstream regulators of the Hippo pathway, including mechanical cues and ligands of G protein-coupled receptors (GPCRs). Through the activation related G proteins and possibly rearrangements of actin cytoskeleton, GPCR signaling can potently modulate the phosphorylation states and activity of YAP and TAZ, two homologous oncogenic transcriptional co-activators, and major effectors of the Hippo pathway. Herein, we summarize the network, regulation, and functions of GPCR-Hippo signaling, and we will also discuss potential anti-cancer therapies targeting GPCR-YAP signaling.
Collapse
|
118
|
Li M, Chen J, Yu X, Xu S, Li D, Zheng Q, Yin Y. Myricetin Suppresses the Propagation of Hepatocellular Carcinoma via Down-Regulating Expression of YAP. Cells 2019; 8:cells8040358. [PMID: 30999669 PMCID: PMC6523269 DOI: 10.3390/cells8040358] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 01/05/2023] Open
Abstract
Myricetin is a naturally occurring flavonoid with protective effects against a variety of cancers. However, the molecular mechanism of myricetin against hepatocellular carcinoma (HCC) has still not been fully elucidated. Previous studies have indicated that YAP is essential for cancer initiation and progression. However, whether YAP contributes to the anti-cancer effects of myricetin remains unclear. Herein, we aimed to investigate the effect of myricetin on HCC, and identify the underlying mechanisms. We report that myricetin induced apoptosis and proliferation inhibition in HepG2 and Huh-7 cells. Myricetin inhibited expression of YAP by promoting its phosphorylation and subsequent degradation. Myricetin inhibited YAP expression by stimulating kinase activation of LATS1/2. Knockdown expression of LATS1/2 by shRNA attenuated myricetin-induced phosphorylation and degradation of YAP. Furthermore, myricetin sensitized HCC cells to cisplatin treatment through inhibiting YAP and its target genes, both in vitro and in vivo. The identification of the LATS1/2-YAP pathway as a target of myricetin may help with the design of novel strategies for human HCC prevention and therapy.
Collapse
Affiliation(s)
- Minjing Li
- Institute of Integrated Medicine, Binzhou Medical University, Yantai 264003, China.
| | - Jinliang Chen
- School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China.
| | - Xiaofei Yu
- Institute of Integrated Medicine, Binzhou Medical University, Yantai 264003, China.
| | - Sen Xu
- Key Laboratory of Tumour Molecular Biology, Department of Clinical Medicine, Binzhou Medical University, Yantai 264003, China.
| | - Defang Li
- Institute of Integrated Medicine, Binzhou Medical University, Yantai 264003, China.
| | - Qiusheng Zheng
- Institute of Integrated Medicine, Binzhou Medical University, Yantai 264003, China.
| | - Yancun Yin
- School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
119
|
Guo P, Lee CH, Lei H, Zheng Y, Pulgar Prieto KD, Pan D. Nerfin-1 represses transcriptional output of Hippo signaling in cell competition. eLife 2019; 8:38843. [PMID: 30901309 PMCID: PMC6430605 DOI: 10.7554/elife.38843] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 02/25/2019] [Indexed: 12/29/2022] Open
Abstract
The Hippo tumor suppressor pathway regulates tissue growth in Drosophila by restricting the activity of the transcriptional coactivator Yorkie (Yki), which normally complexes with the TEF/TEAD family DNA-binding transcription factor Scalloped (Sd) to drive the expression of growth-promoting genes. Given its pivotal role as a central hub in mediating the transcriptional output of Hippo signaling, there is great interest in understanding the molecular regulation of the Sd-Yki complex. In this study, we identify Nerfin-1 as a transcriptional repressor that antagonizes the activity of the Sd-Yki complex by binding to the TEA DNA-binding domain of Sd. Consistent with its biochemical function, ectopic expression of Nerfin-1 results in tissue undergrowth in an Sd-dependent manner. Conversely, loss of Nerfin-1 enhances the ability of winner cells to eliminate loser cells in multiple scenarios of cell competition. We further show that INSM1, the mammalian ortholog of Nerfin-1, plays a conserved role in repressing the activity of the TEAD-YAP complex. These findings reveal a novel regulatory mode converging on the transcriptional output of the Hippo pathway that may be exploited for modulating the YAP oncoprotein in cancer and regenerative medicine. Animals uses a range of mechanisms to stop their organs from growing once they have reached the right shape and size. One of these processes, a set of chemical messages called the Hippo pathway, controls the balance of cell death and cell division. In fruit flies, Hippo works by repressing a complex formed of two proteins, Yorkie and Scalloped, which normally switch genes on to encourage cells to grow. Yorkie is also involved in cell competition, a process in which cells in a tissue compare themselves to each other. Healthier ‘winner’ cells then kill neighboring ‘loser’ cells that are weaker or damaged. This ensures that the tissue keeps working properly. Despite Yorkie and Scalloped being key to control the growth and health of tissues, how the activity of these proteins is regulated was not well understood. To investigate, Guo et al. conducted a series experiments on fruit flies and found that a protein called Nerfin-1 can bind onto Scalloped to stop the Scalloped-Yorkie complex from switching on genes. As a result, flies with too much Nerfin-1 had stunted tissue growth. In addition, Guo et al. confirmed that the Nerfin-1 equivalent in mammals acts in the same way. Further work revealed that Nerfin-1 also plays a role in cell competition: without this protein, ‘winner’ cells became 'super winners', eliminating even more of the loser cells. Besides regulating the size of organs, the Hippo pathway is also involved in stopping cells from dividing uncontrollably and becoming cancerous. Further research may therefore focus on Nerfin-1 and its equivalent in mammals to understand how this protein could contribute to the emergence of cancer.
Collapse
Affiliation(s)
- Pengfei Guo
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chang-Hyun Lee
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Huiyan Lei
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Katiuska Daniela Pulgar Prieto
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
120
|
Chen YA, Lu CY, Cheng TY, Pan SH, Chen HF, Chang NS. WW Domain-Containing Proteins YAP and TAZ in the Hippo Pathway as Key Regulators in Stemness Maintenance, Tissue Homeostasis, and Tumorigenesis. Front Oncol 2019; 9:60. [PMID: 30805310 PMCID: PMC6378284 DOI: 10.3389/fonc.2019.00060] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/21/2019] [Indexed: 12/29/2022] Open
Abstract
The Hippo pathway is a conserved signaling pathway originally defined in Drosophila melanogaster two decades ago. Deregulation of the Hippo pathway leads to significant overgrowth in phenotypes and ultimately initiation of tumorigenesis in various tissues. The major WW domain proteins in the Hippo pathway are YAP and TAZ, which regulate embryonic development, organ growth, tissue regeneration, stem cell pluripotency, and tumorigenesis. Recent reports reveal the novel roles of YAP/TAZ in establishing the precise balance of stem cell niches, promoting the production of induced pluripotent stem cells (iPSCs), and provoking signals for regeneration and cancer initiation. Activation of YAP/TAZ, for example, results in the expansion of progenitor cells, which promotes regeneration after tissue damage. YAP is highly expressed in self-renewing pluripotent stem cells. Overexpression of YAP halts stem cell differentiation and yet maintains the inherent stem cell properties. A success in reprograming iPSCs by the transfection of cells with Oct3/4, Sox2, and Yap expression constructs has recently been shown. In this review, we update the current knowledge and the latest progress in the WW domain proteins of the Hippo pathway in relevance to stem cell biology, and provide a thorough understanding in the tissue homeostasis and identification of potential targets to block tumor development. We also provide the regulatory role of tumor suppressor WWOX in the upstream of TGF-β, Hyal-2, and Wnt signaling that cross talks with the Hippo pathway.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yu Lu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tian-You Cheng
- Department of Optics and Photonics, National Central University, Chungli, Taiwan
| | - Szu-Hua Pan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University, Taipei, Taiwan
| | - Nan-Shan Chang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, United States.,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
121
|
Abstract
The Hippo Pathway comprises a vast network of components that integrate diverse signals including mechanical cues and cell surface or cell-surface-associated molecules to define cellular outputs of growth, proliferation, cell fate, and cell survival on both the cellular and tissue level. Because of the importance of the regulators, core components, and targets of this pathway in human health and disease, individual components were often identified by efforts in mammalian models or for a role in a specific process such as stress response or cell death. However, multiple components were originally discovered in the Drosophila system, and the breakthrough of conceiving that these components worked together in a signaling pathway came from a series of Drosophila genetic screens and fundamental genetic and phenotypic characterization efforts. In this chapter, we will review the original discoveries leading to the conceptual framework of these components as a tumor suppressor network. We will review chronologically the early efforts that established our initial understanding of the core machinery that then launched the growing and vibrant field to be discussed throughout later chapters of this book.
Collapse
Affiliation(s)
- Rewatee Gokhale
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cathie M Pfleger
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
122
|
Abstract
Immunocytochemistry enables determination of cellular localization and relative abundance of proteins. This protocol describes a rapid and cost-effective approach to study the cellular localization of YAP (and TAZ), the transcriptional co activators of the Hippo pathway, in mammalian cells. Cells are seeded onto coated cover slips, cultivated and treated as required. Subsequently, they are chemically fixed, and cellular proteins are fluorescently labeled by means of specific antibodies. Multiplexing antibodies enables ascertaining the subcellular localization of YAP and TAZ and thereby also the activation state of the Hippo pathway in various cell types.
Collapse
Affiliation(s)
- Valentina Rausch
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK
| | - Carsten G Hansen
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK.
| |
Collapse
|
123
|
Hu J, Ruan J, Liu X, Xiao C, Xiong J. MicroRNA-301a-3p suppressed the progression of hepatocellular carcinoma via targeting VGLL4. Pathol Res Pract 2018; 214:2039-2045. [PMID: 30322806 DOI: 10.1016/j.prp.2018.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/23/2018] [Accepted: 09/11/2018] [Indexed: 11/28/2022]
Abstract
Dysregulation of microRNAs (miRNAs) is involved in a variety of biological process including tumorigenesis. miR-301a-3p has been reported to be an onco-miRNA in various types of cancer, like breast cancer, malignant melanoma, and pancreatic cancer. However, the role of miR-301a-3p in hepatocellular carcinoma (HCC) remains largely incomplete. In the present study, we found that miR-301a-3p was upregulated in HCC tissues and cell lines, and higher miR-301a-3p expression predicted poor prognosis in HCC patients. We also demonstrated that miR-301a-3p overexpression enhanced the ability of proliferation, invasion, and chemoresistance in HCC cell lines, and conversely, silencing miR-301a-3p expression induced the opposite effects. VGLL4, as the direct target of miR-301a-3p, was predicted by bioinformatic websites and confirmed by western blot, RT-PCR, and luciferase reporter assay. Enforced VGLL4 expression rescued the effects of miR-301a-3p mimics on cell proliferation, invasion and chemoresistance. Moreover, we found miR-301a-3p modulated the transcription activity of TEADs. Collectively, our findings suggested that the miR-301a-3p-VGLL4 signaling axis might be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Jinhua Hu
- Department of Oncology, The People's Hospital of Xinyu City, Xinyu, Jiangxi Province, 338000, China; Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Jiugen Ruan
- Department of Radiology, The People's Hospital of Xinyu City, Xinyu, Jiangxi Province, 338000, China
| | - Xiaohong Liu
- Department of Oncology, The People's Hospital of Xinyu City, Xinyu, Jiangxi Province, 338000, China
| | - Chijin Xiao
- Department of Oncology, The People's Hospital of Xinyu City, Xinyu, Jiangxi Province, 338000, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
124
|
Abstract
Hippo signaling is an evolutionarily conserved network that has a central role in regulating cell proliferation and cell fate to control organ growth and regeneration. It promotes activation of the LATS kinases, which control gene expression by inhibiting the activity of the transcriptional coactivator proteins YAP and TAZ in mammals and Yorkie in Drosophila. Diverse upstream inputs, including both biochemical cues and biomechanical cues, regulate Hippo signaling and enable it to have a key role as a sensor of cells' physical environment and an integrator of growth control signals. Several components of this pathway localize to cell-cell junctions and contribute to regulation of Hippo signaling by cell polarity, cell contacts, and the cytoskeleton. Downregulation of Hippo signaling promotes uncontrolled cell proliferation, impairs differentiation, and is associated with cancer. We review the current understanding of Hippo signaling and highlight progress in the elucidation of its regulatory mechanisms and biological functions.
Collapse
Affiliation(s)
- Jyoti R Misra
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA;
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA;
| |
Collapse
|
125
|
Cheng Y, Chen D. Fruit fly research in China. J Genet Genomics 2018; 45:583-592. [PMID: 30455037 DOI: 10.1016/j.jgg.2018.09.003] [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: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 11/19/2022]
Abstract
Served as a model organism over a century, fruit fly has significantly pushed forward the development of global scientific research, including in China. The high similarity in genomic features between fruit fly and human enables this tiny insect to benefit the biomedical studies of human diseases. In the past decades, Chinese biologists have used fruit fly to make numerous achievements on understanding the fundamental questions in many diverse areas of biology. Here, we review some of the recent fruit fly studies in China, and mainly focus on those studies in the fields of stem cell biology, cancer therapy and regeneration medicine, neurological disorders and epigenetics.
Collapse
Affiliation(s)
- Ying Cheng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dahua Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
126
|
Wu A, Wu Q, Deng Y, Liu Y, Lu J, Liu L, Li X, Liao C, Zhao B, Song H. Loss of VGLL4 suppresses tumor PD-L1 expression and immune evasion. EMBO J 2018; 38:embj.201899506. [PMID: 30396996 DOI: 10.15252/embj.201899506] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 01/07/2023] Open
Abstract
Targeting immune checkpoints, such as PD-L1 and its receptor PD-1, has opened a new avenue for treating cancers. Understanding the regulatory mechanism of PD-L1 and PD-1 will improve the clinical response rate and efficacy of PD-1/PD-L1 blockade in cancer patients and the development of combinatorial strategies. VGLL4 inhibits YAP-induced cell proliferation and tumorigenesis through competition with YAP for binding to TEADs. However, whether VGLL4 has a role in anti-tumor immunity is largely unknown. Here, we found that disruption of Vgll4 results in potent T cell-mediated tumor regression in murine syngeneic models. VGLL4 deficiency reduces PD-L1 expression in tumor cells. VGLL4 interacts with IRF2BP2 and promotes its protein stability through inhibiting proteasome-mediated protein degradation. Loss of IRF2BP2 results in persistent binding of IRF2, a transcriptional repressor, to PD-L1 promoter. In addition, YAP inhibits IFNγ-inducible PD-L1 expression partially through suppressing the expression of VGLL4 and IRF1 by YAP target gene miR-130a. Our study identifies VGLL4 as an important regulator of PD-L1 expression and highlights a central role of VGLL4 and YAP in the regulation of tumor immunity.
Collapse
Affiliation(s)
- Ailing Wu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Qingzhe Wu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Yujie Deng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Yuning Liu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Jinqiu Lu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Liansheng Liu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Xiaoling Li
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Cheng Liao
- Department of Preclinical Development, Translation Medicine & External Research, Jiangsu Hengrui Medicine CO., LTD., Shanghai, China
| | - Bin Zhao
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Hai Song
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| |
Collapse
|
127
|
The Scalloped and Nerfin-1 Transcription Factors Cooperate to Maintain Neuronal Cell Fate. Cell Rep 2018; 25:1561-1576.e7. [DOI: 10.1016/j.celrep.2018.10.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/14/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023] Open
|
128
|
Pan Y, Alégot H, Rauskolb C, Irvine KD. The dynamics of Hippo signaling during Drosophila wing development. Development 2018; 145:dev165712. [PMID: 30254143 PMCID: PMC6215397 DOI: 10.1242/dev.165712] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
Tissue growth needs to be properly controlled for organs to reach their correct size and shape, but the mechanisms that control growth during normal development are not fully understood. We report here that the activity of the Hippo signaling transcriptional activator Yorkie gradually decreases in the central region of the developing Drosophila wing disc. Spatial and temporal changes in Yorkie activity can be explained by changes in cytoskeletal tension and biomechanical regulators of Hippo signaling. These changes in cellular biomechanics correlate with changes in cell density, and experimental manipulations of cell density are sufficient to alter biomechanical Hippo signaling and Yorkie activity. We also relate the pattern of Yorkie activity in older discs to patterns of cell proliferation. Our results establish that spatial and temporal patterns of Hippo signaling occur during wing development, that these patterns depend upon cell-density modulated tissue mechanics and that they contribute to the regulation of wing cell proliferation.
Collapse
Affiliation(s)
- Yuanwang Pan
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Herve Alégot
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Cordelia Rauskolb
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
129
|
Modulation of the Hippo pathway and organ growth by RNA processing proteins. Proc Natl Acad Sci U S A 2018; 115:10684-10689. [PMID: 30257938 DOI: 10.1073/pnas.1807325115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The Hippo tumor-suppressor pathway regulates organ growth, cell proliferation, and stem cell biology. Defects in Hippo signaling and hyperactivation of its downstream effectors-Yorkie (Yki) in Drosophila and YAP/TAZ in mammals-result in progenitor cell expansion and overgrowth of multiple organs and contribute to cancer development. Deciphering the mechanisms that regulate the activity of the Hippo pathway is key to understanding its function and for therapeutic targeting. However, although the Hippo kinase cascade and several other upstream inputs have been identified, the mechanisms that regulate Yki/YAP/TAZ activity are still incompletely understood. To identify new regulators of Yki activity, we screened in Drosophila for suppressors of tissue overgrowth and Yki activation caused by overexpression of atypical protein kinase C (aPKC), a member of the apical cell polarity complex. In this screen, we identified mutations in the heterogeneous nuclear ribonucleoprotein Hrb27C that strongly suppressed the tissue defects induced by ectopic expression of aPKC. Hrb27C was required for aPKC-induced tissue growth and Yki target gene expression but did not affect general gene expression. Genetic and biochemical experiments showed that Hrb27C affects Yki phosphorylation. Other RNA-binding proteins known to interact with Hrb27C for mRNA transport in oocytes were also required for normal Yki activity, although they suppressed Yki output. Based on the known functions of Hrb27C, we conclude that Hrb27C-mediated control of mRNA splicing, localization, or translation is essential for coordinated activity of the Hippo pathway.
Collapse
|
130
|
Zhang W, Xu J, Li J, Guo T, Jiang D, Feng X, Ma X, He L, Wu W, Yin M, Ge L, Wang Z, Ho MS, Zhao Y, Fei Z, Zhang L. The TEA domain family transcription factor TEAD4 represses murine adipogenesis by recruiting the cofactors VGLL4 and CtBP2 into a transcriptional complex. J Biol Chem 2018; 293:17119-17134. [PMID: 30209132 DOI: 10.1074/jbc.ra118.003608] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/15/2018] [Indexed: 12/17/2022] Open
Abstract
The Hippo signaling pathway is known to play an important role in multiple physiological processes, including adipogenesis. However, whether the downstream components of the Hippo pathway are involved in adipogenesis remains unknown. Here we demonstrate that the TEA domain family (TEAD) transcription factors are essential for adipogenesis in murine 3T3-L1 preadipocytes. Knockdown of TEAD1-4 stimulated adipogenesis and increased the expression of adipocyte markers in these cells. Interestingly, we found that the TEAD4 knockdown-mediated adipogenesis proceeded in a Yes-associated protein (YAP)/TAZ (Wwtr1)-independent manner and that adipogenesis suppression in WT cells involved formation of a ternary complex comprising TEAD4 and the transcriptional cofactors C-terminal binding protein 2 (CtBP2) and vestigial-like family member 4 (VGLL4). VGLL4 acted as an adaptor protein that enhanced the interaction between TEAD4 and CtBP2, and this TEAD4-VGLL4-CtBP2 ternary complex dynamically existed at the early stage of adipogenesis. Finally, we verified that TEAD4 directly targets the promoters of major adipogenesis transcription factors such as peroxisome proliferator-activated receptor γ (PPARγ) and adiponectin, C1Q, and collagen domain-containing (Adipoq) during adipogenesis. These findings reveal critical insights into the role of the TEAD4-VGLL4-CtBP2 transcriptional repressor complex in suppression of adipogenesis in murine preadipocytes.
Collapse
Affiliation(s)
- Wenxiang Zhang
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Jinjin Xu
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Jinhui Li
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Tong Guo
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Dan Jiang
- the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xue Feng
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Xueyan Ma
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Lingli He
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Wenqing Wu
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Mengxin Yin
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Ling Ge
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Zuoyun Wang
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Margaret S Ho
- the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yun Zhao
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and.,the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhaoliang Fei
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and
| | - Lei Zhang
- From the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China and .,the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
131
|
Abstract
How the organ size is adjusted to the proper size during development and how organs know that they reach the original size during regeneration remain long-standing questions. Based on studies using multiple model organisms and approaches for over 20 years, a consensus has been established that the Hippo pathway plays crucial roles in controlling organ size and maintaining tissue homeostasis. Given the significance of these processes, the dysregulation of the Hippo pathway has also implicated various diseases, such as tissue degeneration and cancer. By regulating the downstream transcriptional coactivators YAP and TAZ, the Hippo pathway coordinates cell proliferation and apoptosis in response to a variety of signals including cell contact inhibition, polarity, mechanical sensation and soluble factors. Since the core components and their functions of the Hippo pathway are evolutionarily conserved, this pathway serves as a global regulator of organ size control. Therefore, further investigation of the regulatory mechanisms will provide physiological insights to better understand tissue homeostasis. In this review, the historical developments and current understandings of the regulatory mechanism of Hippo signaling pathway are discussed.
Collapse
Affiliation(s)
- Wantae Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Eek-Hoon Jho
- Departement of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
132
|
Xu J, Vanderzalm PJ, Ludwig M, Su T, Tokamov SA, Fehon RG. Yorkie Functions at the Cell Cortex to Promote Myosin Activation in a Non-transcriptional Manner. Dev Cell 2018; 46:271-284.e5. [PMID: 30032991 PMCID: PMC6086586 DOI: 10.1016/j.devcel.2018.06.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
The Hippo signaling pathway is an evolutionarily conserved mechanism that controls organ size in animals. Yorkie is well known as a transcriptional co-activator that functions downstream of the Hippo pathway to positively regulate transcription of genes that promote tissue growth. Recent studies have shown that increased myosin activity activates both Yorkie and its vertebrate orthologue YAP, resulting in increased nuclear localization and tissue growth. Here we show that Yorkie also can accumulate at the cell cortex in the apical junctional region. Moreover, Yorkie functions at the cortex to promote activation of myosin through a myosin regulatory light chain kinase, Stretchin-Mlck. This Yorkie function is not dependent on its transcriptional activity and is required for larval and adult tissues to achieve appropriate size. Based on these results, we suggest that Yorkie functions in a feedforward "amplifier" loop that promotes myosin activation, and thereby greater Yorkie activity, in response to tension.
Collapse
Affiliation(s)
- Jiajie Xu
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Pamela J Vanderzalm
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biology, John Carroll University, University Heights, OH 44118, USA
| | - Michael Ludwig
- Department of Ecology and Evolutionary Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Ting Su
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Sherzod A Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
133
|
Homeostatic Control of Hpo/MST Kinase Activity through Autophosphorylation-Dependent Recruitment of the STRIPAK PP2A Phosphatase Complex. Cell Rep 2018; 21:3612-3623. [PMID: 29262338 DOI: 10.1016/j.celrep.2017.11.076] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/25/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
The Hippo pathway controls organ size and tissue homeostasis through a kinase cascade leading from the Ste20-like kinase Hpo (MST1/2 in mammals) to the transcriptional coactivator Yki (YAP/TAZ in mammals). Whereas previous studies have uncovered positive and negative regulators of Hpo/MST, how they are integrated to maintain signaling homeostasis remains poorly understood. Here, we identify a self-restricting mechanism whereby autophosphorylation of an unstructured linker in Hpo/MST creates docking sites for the STRIPAK PP2A phosphatase complex to inactivate Hpo/MST. Mutation of the phospho-dependent docking sites in Hpo/MST or deletion of Slmap, the STRIPAK subunit recognizing these docking sites, results in constitutive activation of Hpo/MST in both Drosophila and mammalian cells. In contrast, autophosphorylation of the Hpo/MST linker at distinct sites is known to recruit Mats/MOB1 to facilitate Hippo signaling. Thus, multisite autophosphorylation of Hpo/MST linker provides an evolutionarily conserved built-in molecular platform to maintain signaling homeostasis by coupling antagonistic signaling activities.
Collapse
|
134
|
Moon S, Yeon Park S, Woo Park H. Regulation of the Hippo pathway in cancer biology. Cell Mol Life Sci 2018; 75:2303-2319. [PMID: 29602952 PMCID: PMC11105795 DOI: 10.1007/s00018-018-2804-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/17/2018] [Accepted: 03/22/2018] [Indexed: 01/23/2023]
Abstract
The Hippo tumor suppressor pathway, which is well conserved from Drosophila to humans, has emerged as the master regulator of organ size, as well as major cellular properties, such as cell proliferation, survival, stemness, and tissue homeostasis. The biological significance and deregulation of the Hippo pathway in tumorigenesis have received a surge of interest in the past decade. In the current review, we present the major discoveries that made substantial contributions to our understanding of the Hippo pathway and discuss how Hippo pathway components contribute to cellular signaling, physiology, and their potential implications in anticancer therapeutics.
Collapse
Affiliation(s)
- Sungho Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - So Yeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
135
|
Han XY, Liu HZ, Cai CY, Li XQ, Ju YB, Li YY, Zhang ZG. Expression of VGLL4 and YAP protein in gastric carcinoma tissues and tumor prognosis. Minerva Med 2018; 109:429-435. [PMID: 29914241 DOI: 10.23736/s0026-4806.18.05692-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The purpose of this study was to investigate the relationship between expression of VGLL4 and YAP protein in gastric carcinoma and adjacent normal tissues and its relationship with clinicopathological parameters and its significance in prognosis of gastric cancer. METHODS The expression of VGLL4 and YAP protein in gastric carcinoma tissues and adjacent tissues were detected by fluorescence quantitative PCR, Western blot and immunohistochemistry. The relationship between the expression of VGLL4 and YAP protein and the clinicopathological parameters of gastric cancer patients was analyzed to determine its impact on the invasion and metastasis and prognosis of patients. RESULTS The expression of VGLL4 protein in gastric carcinoma was lower than that in adjacent tissues, and it was negatively correlated with infiltration depth, lymph node metastasis and TNM staging; the expression of YAP protein in gastric carcinoma was higher than that in adjacent tissues, and it was positively correlated with age, tumor differentiation, depth of invasion, lymph node metastasis and TNM staging. CONCLUSIONS VGLL4 protein decreased in gastric cancer tissue and was negatively correlated with tumor progression and malignancy degree, suggesting that it is a potential antitumor protein; YAP protein increased in gastric cancer and is a proto-oncogene.
Collapse
Affiliation(s)
- Xin-Ying Han
- Department of Pathology, Cangzhou People's Hospital, Cangzhou, China
| | - Hong-Zheng Liu
- Department of Orthopedics, Cangzhou Hospital of Integrated TCM-WM, Cangzhou, China
| | - Chun-Yi Cai
- Department of Pathology, Cangzhou People's Hospital, Cangzhou, China
| | - Xiu-Qing Li
- Department of Pathology, Cangzhou People's Hospital, Cangzhou, China
| | - Ying-Bo Ju
- Department of Pathology, Cangzhou People's Hospital, Cangzhou, China
| | - Yan-Yan Li
- Prenatal Diagnosis Center, Cangzhou People's Hospital, Cangzhou, China
| | - Zhi-Gang Zhang
- Department of Pathology, Cangzhou People's Hospital, Cangzhou, China -
| |
Collapse
|
136
|
Deng X, Fang L. VGLL4 is a transcriptional cofactor acting as a novel tumor suppressor via interacting with TEADs. Am J Cancer Res 2018; 8:932-943. [PMID: 30034932 PMCID: PMC6048398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/18/2018] [Indexed: 06/08/2023] Open
Abstract
Vestigial Like Family Member 4 (VGLL4) is a transcriptional cofactor of VGLL family, which includes VGLL1-4. Unlike other members of VGLL family, VGLL4 was described as a novel tumor suppressor containing two TDU motifs. VGLL4 executes its biological function through two TDU domains via interacting with TEA domain (TEAD) transcription factors. Lower expression of VGLL4 usually indicates poor survival in many cancers, such as lung cancer, gastric cancer, breast cancer, colorectal cancer, bladder cancer, pancreatic adenocarcinoma and esophageal squamous cancer. In cancer cells, the expression of VGLL4 is lower than that of normal tissues, moreover, expression level of VGLL4 is positively related to survival rate. VGLL4 is found to play an important role in several signal pathways, mainly acts as a tumor suppressor interacting with TEADs. In Hippo signaling pathway, VGLL4 competes with YAP in binding to TEADs and inhibits the downstream of YAP. In Wnt/β-catenin signaling pathway, VGLL4 negatively regulates Wnt/β-catenin signaling pathway via inhibiting β-catenin and TCF (T-cell factor). VGLL4 can also suppress epithelial-mesenchymal transition (EMT) and contribute to apoptosis signaling pathway.
Collapse
Affiliation(s)
- Xiaochong Deng
- Department of Thyroid and Breast, Division of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai 200072, People's Republic of China
| | - Lin Fang
- Department of Thyroid and Breast, Division of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai 200072, People's Republic of China
| |
Collapse
|
137
|
Manning SA, Dent LG, Kondo S, Zhao ZW, Plachta N, Harvey KF. Dynamic Fluctuations in Subcellular Localization of the Hippo Pathway Effector Yorkie In Vivo. Curr Biol 2018; 28:1651-1660.e4. [DOI: 10.1016/j.cub.2018.04.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/20/2022]
|
138
|
Watt KI, Goodman CA, Hornberger TA, Gregorevic P. The Hippo Signaling Pathway in the Regulation of Skeletal Muscle Mass and Function. Exerc Sport Sci Rev 2018; 46:92-96. [PMID: 29346163 PMCID: PMC6319272 DOI: 10.1249/jes.0000000000000142] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Hippo signaling pathway regulates the activity of the proteins Yes-associated protein (Yap) and transcriptional co-activator with PDZ-binding motif (Taz) to control tissue growth in many different cell types. Previously, we demonstrated that Yap is a critical regulator of skeletal muscle mass. We hypothesize that alterations in Yap and Taz activity modulate the anabolic adaptations of skeletal muscle to resistance exercise.
Collapse
Affiliation(s)
- Kevin I. Watt
- Baker Heart and Diabetes Institute, Victoria, 3004, Australia
- Department of Diabetes, Monash University, Victoria, 3004, Australia
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Washington, USA
| | - Craig A. Goodman
- College of Health & Biomedicine, Victoria University, Melbourne, Victoria 8001, Australia
- Institute for Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, Victoria, 8001, Australia
- Australian Institute of Musculoskeletal Science (AIMSS),Victoria University, St Albans, Victoria 3021, Australia
| | - Troy A. Hornberger
- Dept of Comparative Bioscience, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Paul Gregorevic
- Dept of Physiology, The University of Melbourne, Victoria, Australia, 3010
- Dept of Biochemistry and Molecular Biology, Monash University, Victoria, Australia, 3800
- Dept of Neurology, The University of Washington School of Medicine, Seattle, Washington, USA 98195
| |
Collapse
|
139
|
Lin KC, Park HW, Guan KL. Deregulation and Therapeutic Potential of the Hippo Pathway in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kimberly C. Lin
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
140
|
Park JH, Shin JE, Park HW. The Role of Hippo Pathway in Cancer Stem Cell Biology. Mol Cells 2018; 41:83-92. [PMID: 29429151 PMCID: PMC5824027 DOI: 10.14348/molcells.2018.2242] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 02/08/2023] Open
Abstract
The biological significance and deregulation of the Hippo pathway during organ growth and tumorigenesis have received a surge of interest in the past decade. The Hippo pathway core kinases, MST1/2 and LATS1/2, are tumor suppressors that inhibit the oncogenic nuclear function of YAP/TAZ and TEAD. In addition to earlier studies that highlight the role of Hippo pathway in organ size control, cell proliferation, and tumor development, recent evidence demonstrates its critical role in cancer stem cell biology, including EMT, drug resistance, and self-renewal. Here we provide a brief overview of the regulatory mechanisms of the Hippo pathway, its role in cancer stem cell biology, and promising therapeutic interventions.
Collapse
Affiliation(s)
- Jae Hyung Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Ji Eun Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| |
Collapse
|
141
|
Elbediwy A, Thompson BJ. Evolution of mechanotransduction via YAP/TAZ in animal epithelia. Curr Opin Cell Biol 2018; 51:117-123. [PMID: 29477107 DOI: 10.1016/j.ceb.2018.02.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Abstract
Mechanical stretch forces can control the growth of epithelial tissues such as mammalian skin, whose surface area is precisely coordinated with body size. In skin keratinocytes cultured in vitro, mechanical forces acting via Integrin adhesions and the actin cytoskeleton have been shown to induce nuclear translocation of YAP/TAZ co-activators to induce cell proliferation. Furthermore, conditional knockouts of both YAP (also called YAP1) and TAZ (also called WWTR1) in mouse skin resemble the phenotype of skin-specific loss of Integrin beta1 (ITGB1), indicating that this signalling mechanism is important in vivo. Curiously, Integrins are dispensable in Drosophila to activate the sole YAP/TAZ homolog Yorkie (Yki), which has lost the C-terminal PDZ-binding motif needed to promote nuclear localization of YAP/TAZ in mammalian cells. Differences in the structure of the epidermis between deuterostomes (e.g.: stratified squamous skin of mammals) and protostomes (e.g.: monolayered columnar epidermis of Drosophila) may explain this evolutionary divergence. Monolayered columnar epithelia feature a well-differentiated apical membrane domain, where proteins such as Crumbs, Expanded, Merlin and Kibra activate the Hippo pathway to repress Drosophila Yki. Stratified squamous epithelia lack an apical domain and thus depend primarily on basal Integrin adhesions to activate YAP/TAZ in basal layer stem cells via multiple postulated signalling mechanisms. Finally, YAP and TAZ retain the ability to sense the apical domain in the columnar epithelial cells lining internal organs such as the lung bronchus, where YAP/TAZ localize to the nucleus in proliferating basal layer stem cells but translocate to the cytoplasm in differentiated columnar cells.
Collapse
Affiliation(s)
- Ahmed Elbediwy
- Epithelial Biology Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, United Kingdom
| | - Barry J Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, United Kingdom.
| |
Collapse
|
142
|
Yu J, Pan D. Validating upstream regulators of Yorkie activity in Hippo signaling through scalloped-based genetic epistasis. Development 2018; 145:145/4/dev157545. [PMID: 29467233 DOI: 10.1242/dev.157545] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 01/19/2018] [Indexed: 12/19/2022]
Abstract
Genetic studies in Drosophila have been instrumental in characterizing the Hippo pathway, which converges on the co-activator Yorkie to regulate target gene transcription. A routinely used strategy to interrogate upstream regulators of Yorkie involves the examination of selected Hippo target genes upon loss or gain of function of a suspected pathway regulator. A caveat with this strategy is that aberrant expression of a given Hippo target per se does not distinguish whether it is caused by changes in Yorkie or Yorkie-independent inputs converging on the same target gene. Building on previous findings that the DNA-binding transcription factor Scalloped mediates both Yorkie overexpression and loss-of-function phenotypes yet is itself dispensable for normal eye development, we describe a simple strategy to distinguish these possibilities by analyzing double-mutant clones of scalloped and a suspected Yorkie regulator. We provide proof of principle that this strategy can be used effectively to validate canonical Yorkie regulators and to exclude proteins that impact target expression independent of Yorkie. The described methodology and reagents should facilitate efforts to assess the expanding repertoire of proteins implicated in regulation of Yorkie activity.
Collapse
Affiliation(s)
- Jianzhong Yu
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.,Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| |
Collapse
|
143
|
A histone deacetylase 3-dependent pathway delimits peripheral myelin growth and functional regeneration. Nat Med 2018; 24:338-351. [PMID: 29431744 DOI: 10.1038/nm.4483] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 01/04/2018] [Indexed: 12/11/2022]
Abstract
Deficits in Schwann cell-mediated remyelination impair functional restoration after nerve damage, contributing to peripheral neuropathies. The mechanisms mediating block of remyelination remain elusive. Here, through small-molecule screening focusing on epigenetic modulators, we identified histone deacetylase 3 (HDAC3; a histone-modifying enzyme) as a potent inhibitor of peripheral myelinogenesis. Inhibition of HDAC3 enhanced myelin growth and regeneration and improved functional recovery after peripheral nerve injury in mice. HDAC3 antagonizes the myelinogenic neuregulin-PI3K-AKT signaling axis. Moreover, genome-wide profiling analyses revealed that HDAC3 represses promyelinating programs through epigenetic silencing while coordinating with p300 histone acetyltransferase to activate myelination-inhibitory programs that include the HIPPO signaling effector TEAD4 to inhibit myelin growth. Schwann cell-specific deletion of either Hdac3 or Tead4 in mice resulted in an elevation of myelin thickness in sciatic nerves. Thus, our findings identify the HDAC3-TEAD4 network as a dual-function switch of cell-intrinsic inhibitory machinery that counters myelinogenic signals and maintains peripheral myelin homeostasis, highlighting the therapeutic potential of transient HDAC3 inhibition for improving peripheral myelin repair.
Collapse
|
144
|
Kjærner-Semb E, Ayllon F, Kleppe L, Sørhus E, Skaftnesmo K, Furmanek T, Segafredo FT, Thorsen A, Fjelldal PG, Hansen T, Taranger GL, Andersson E, Schulz RW, Wargelius A, Edvardsen RB. Vgll3 and the Hippo pathway are regulated in Sertoli cells upon entry and during puberty in Atlantic salmon testis. Sci Rep 2018; 8:1912. [PMID: 29382956 PMCID: PMC5789820 DOI: 10.1038/s41598-018-20308-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/16/2018] [Indexed: 01/07/2023] Open
Abstract
Vgll3 is linked to age at maturity in Atlantic salmon (Salmo salar). However, the molecular mechanisms involving Vgll3 in controlling timing of puberty as well as relevant tissue and cell types are currently unknown. Vgll3 and the associated Hippo pathway has been linked to reduced proliferation activity in different tissues. Analysis of gene expression reveals for the first time that vgll3 and several members of the Hippo pathway were down-regulated in salmon testis during onset of puberty and remained repressed in maturing testis. In the gonads, we found expression in Sertoli and granulosa cells in males and females, respectively. We hypothesize that vgll3 negatively regulates Sertoli cell proliferation in testis and therefore acts as an inhibitor of pubertal testis growth. Gonadal expression of vgll3 is located to somatic cells that are in direct contact with germ cells in both sexes, however our results indicate sex-biased regulation of vgll3 during puberty.
Collapse
Affiliation(s)
- Erik Kjærner-Semb
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway. .,Department of Biology, University of Bergen, Bergen, Norway.
| | - Fernando Ayllon
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Lene Kleppe
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Elin Sørhus
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Kai Skaftnesmo
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Tomasz Furmanek
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Frida T Segafredo
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Anders Thorsen
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Per Gunnar Fjelldal
- Institute of Marine research, Matre Aquaculture Research Station, 5984, Matredal, Norway
| | - Tom Hansen
- Institute of Marine research, Matre Aquaculture Research Station, 5984, Matredal, Norway
| | - Geir Lasse Taranger
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Eva Andersson
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Rüdiger W Schulz
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway.,Department Biology, Utrecht University, Science Faculty, Padualaan 8, NL-3584 CH, Utrecht, The Netherlands
| | - Anna Wargelius
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| | - Rolf B Edvardsen
- Institute of Marine Research, P.O. Box 1870, Nordnes, NO-5817, Bergen, Norway
| |
Collapse
|
145
|
Jiao S, Guan J, Chen M, Wang W, Li C, Wang Y, Cheng Y, Zhou Z. Targeting IRF3 as a YAP agonist therapy against gastric cancer. J Exp Med 2018; 215:699-718. [PMID: 29339449 PMCID: PMC5789414 DOI: 10.1084/jem.20171116] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/07/2017] [Accepted: 12/12/2017] [Indexed: 11/25/2022] Open
Abstract
Jiao et al. show that the key player of antiviral immunity IRF3 binds to and promotes the transactivation of the YAP–TEAD4 complex to coregulate transcription of Hippo pathway target genes and that therapeutic targeting of IRF3 suppresses YAP-driven gastric cancer. The Hippo pathway plays a vital role in tissue homeostasis and tumorigenesis. The transcription factor IRF3 is essential for innate antiviral immunity. In this study, we discovered IRF3 as an agonist of Yes-associated protein (YAP). The expression of IRF3 is positively correlated with that of YAP and its target genes in gastric cancer; the expression of both IRF3 and YAP is up-regulated and prognosticates patient survival. IRF3 interacts with both YAP and TEAD4 in the nucleus to enhance their interaction, promoting nuclear translocation and activation of YAP. IRF3 and YAP–TEAD4 are associated genome-wide to cobind and coregulate many target genes of the Hippo pathway. Overexpression of active IRF3 increased, but depletion of IRF3 reduced, the occupancy of YAP on the target genes. Knockdown or pharmacological targeting of IRF3 by Amlexanox, a drug used clinically for antiinflammatory treatment, inhibits gastric tumor growth in a YAP-dependent manner. Collectively, our study identifies IRF3 as a positive regulator for YAP, highlighting a new therapeutic target against YAP-driven cancers.
Collapse
Affiliation(s)
- Shi Jiao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jingmin Guan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Min Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenjia Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chuanchuan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yugong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yunfeng Cheng
- Department of Hematology and Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaocai Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China .,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
146
|
Nyarko A. Differential Binding Affinities and Allosteric Conformational Changes Underlie Interactions of Yorkie and a Multivalent PPxY Partner. Biochemistry 2018; 57:547-556. [DOI: 10.1021/acs.biochem.7b00973] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Afua Nyarko
- Department of Biochemistry
and Biophysics, Oregon State University, Corvallis, Oregon 97331, United States
| |
Collapse
|
147
|
Zhang P, Pei C, Wang X, Xiang J, Sun BF, Cheng Y, Qi X, Marchetti M, Xu JW, Sun YP, Edgar BA, Yuan Z. A Balance of Yki/Sd Activator and E2F1/Sd Repressor Complexes Controls Cell Survival and Affects Organ Size. Dev Cell 2018; 43:603-617.e5. [PMID: 29207260 PMCID: PMC5722641 DOI: 10.1016/j.devcel.2017.10.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/17/2017] [Accepted: 10/29/2017] [Indexed: 01/03/2023]
Abstract
The Hippo/Yki and RB/E2F pathways both regulate tissue growth by affecting cell proliferation and survival, but interactions between these parallel control systems are poorly defined. In this study, we demonstrate that interaction between Drosophila E2F1 and Sd disrupts Yki/Sd complex formation and thereby suppresses Yki target gene expression. RBF modifies these effects by reducing E2F1/Sd interaction. This regulation has significant effects on apoptosis, organ size, and progenitor cell proliferation. Using a combination of DamID-seq and RNA-seq, we identified a set of Yki targets that play a diversity of roles during development and are suppressed by E2F1. Further, we found that human E2F1 competes with YAP for TEAD1 binding, affecting YAP activity, indicating that this mode of cross-regulation is conserved. In sum, our study uncovers a previously unknown mechanism in which RBF and E2F1 modify Hippo signaling responses to modulate apoptosis, organ growth, and homeostasis. RBF/E2F1 regulates the Hippo pathway by modulating formation of Yki/Sd complexes E2F1 releases Yki:Sd association and suppresses a set of Yki target expression Human E2F1 competes with YAP for TEAD1 binding and affects YAP activity
Collapse
Affiliation(s)
- Peng Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; German Cancer Research Center (DKFZ) & Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany; Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Chunli Pei
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; The Brain Science Center, Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Xi Wang
- German Cancer Research Center (DKFZ) & Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany
| | - Jinyi Xiang
- German Cancer Research Center (DKFZ) & Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany
| | - Bao-Fa Sun
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongsheng Cheng
- German Cancer Research Center (DKFZ) & Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany
| | - Xiaolong Qi
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; The Brain Science Center, Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - Marco Marchetti
- German Cancer Research Center (DKFZ) & Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany; Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Jia-Wei Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Ying-Pu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Bruce A Edgar
- German Cancer Research Center (DKFZ) & Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany; Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100069, China.
| |
Collapse
|
148
|
Gibault F, Sturbaut M, Bailly F, Melnyk P, Cotelle P. Targeting Transcriptional Enhanced Associate Domains (TEADs). J Med Chem 2017; 61:5057-5072. [DOI: 10.1021/acs.jmedchem.7b00879] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Floriane Gibault
- JPArc, Centre
de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, UMR-S-1172,
INSERM, CHU Lille, Université de Lille, F-59000 Lille, France
| | - Manon Sturbaut
- JPArc, Centre
de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, UMR-S-1172,
INSERM, CHU Lille, Université de Lille, F-59000 Lille, France
| | - Fabrice Bailly
- JPArc, Centre
de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, UMR-S-1172,
INSERM, CHU Lille, Université de Lille, F-59000 Lille, France
| | - Patricia Melnyk
- JPArc, Centre
de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, UMR-S-1172,
INSERM, CHU Lille, Université de Lille, F-59000 Lille, France
| | - Philippe Cotelle
- JPArc, Centre
de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, UMR-S-1172,
INSERM, CHU Lille, Université de Lille, F-59000 Lille, France
- ENSCL, F-59000 Lille, France
| |
Collapse
|
149
|
Wang SP, Wang LH. Disease implication of hyper-Hippo signalling. Open Biol 2017; 6:rsob.160119. [PMID: 27805903 PMCID: PMC5090056 DOI: 10.1098/rsob.160119] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
The Hippo signalling pathway regulates cellular proliferation, apoptosis and differentiation, thus exerting profound effects on cellular homeostasis. Inhibition of Hippo signalling has been frequently implicated in human cancers, indicating a well-known tumour suppressor function of the Hippo pathway. However, it is less certain whether and how hyperactivation of the Hippo pathway affects biological outcome in living cells. This review describes current knowledge of the regulatory mechanisms of the Hippo pathway, mainly focusing on hyperactivation of the Hippo signalling nexus. The disease implications of hyperactivated Hippo signalling have also been discussed, including arrhythmogenic cardiomyopathy, Sveinsson's chorioretinal atrophy, Alzheimer's disease, amyotrophic lateral sclerosis and diabetes. By highlighting the significance of disease-relevant Hippo signalling activation, this review can offer exciting prospects to address the onset and potential reversal of Hippo-related disorders.
Collapse
Affiliation(s)
- Shu-Ping Wang
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 114, Taiwan
| |
Collapse
|
150
|
Scalloped a member of the Hippo tumor suppressor pathway controls mushroom body size in Drosophila brain by non-canonical regulation of neuroblast proliferation. Dev Biol 2017; 432:203-214. [DOI: 10.1016/j.ydbio.2017.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/16/2017] [Accepted: 10/20/2017] [Indexed: 01/18/2023]
|