1
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
2
|
Kim J, Kim H, Kim J, Cho SY, Moon S, Yoo Y, Kim H, Kim JK, Jeon H, Namkung W, Han G, No KT. Pan-Transcriptional Enhanced Associated Domain Palmitoylation Pocket Covalent Inhibitor. J Med Chem 2024; 67:18957-18968. [PMID: 39487823 DOI: 10.1021/acs.jmedchem.4c01393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
In the Hippo signaling pathway, the palmitoylated transcriptional enhanced associated domain (TEAD) protein interacts with the coactivator Yes-associated protein/PDZ-binding motif, leading to transcriptional upregulation of oncogenes such as Ctgf and Cyr61. Consequently, targeting the palmitoylation sites of TEAD has emerged as a promising strategy for treating TEAD-dependent cancers. Compound 1 was identified using a structure-based drug design approach, leveraging the molecular insights gained from the known TEAD palmitoylation site inhibitor, K-975. Optimization of the initial hit compound resulted in the development of compound 3, a covalent pan-TEAD inhibitor characterized by high potency and oral bioavailability.
Collapse
Affiliation(s)
- Jinhyuk Kim
- The Interdisciplinary Graduate Program in Integrative Biotechnology & Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Hadong Kim
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Jongwan Kim
- Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Seon Yeon Cho
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Sungho Moon
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Youngki Yoo
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Hanseong Kim
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Jin Kwan Kim
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
| | - Hyejin Jeon
- Department of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Wan Namkung
- Department of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Gyoonhee Han
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Department of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
- Postech Biotech Center, 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, Korea
| | - Kyoung Tai No
- Baobab AiBIO Co., Ltd., Incheon 21983, Republic of Korea
- Institute of Convergence Science and Technology, Yonsei University, Incheon 21983, Republic of Korea
| |
Collapse
|
3
|
Moore SM, Jeong E, Zahid M, Gawron J, Arora S, Belin S, Sim F, Poitelon Y, Feltri ML. Loss of YAP in Schwann cells improves HNPP pathophysiology. Glia 2024; 72:1974-1984. [PMID: 38989661 PMCID: PMC11563883 DOI: 10.1002/glia.24592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
Rapid nerve conduction in the peripheral nervous system (PNS) is facilitated by the multilamellar myelin sheath encasing many axons of peripheral nerves. Charcot-Marie-Tooth type 1A (CMT1A), and hereditary neuropathy with liability to pressure palsy (HNPP) are common demyelinating inherited peripheral neuropathies and are caused by mutations in the peripheral myelin protein 22 (PMP22) gene. Duplication of PMP22 leads to its overexpression and causes CMT1A, while its deletion results in PMP22 under expression and causes HNPP. Here, we investigated novel targets for modulating the protein level of PMP22 in HNPP. We found that genetic attenuation of the transcriptional coactivator Yap in Schwann cells reduces p-TAZ levels, increased TAZ activity, and increases PMP22 in peripheral nerves. Based on these findings, we ablated Yap alleles in Schwann cells of the Pmp22-haploinsufficient mouse model of HNPP and identified fewer tomacula on morphological assessment and improved nerve conduction in peripheral nerves. These findings suggest YAP modulation may be a new avenue for treatment of HNPP.
Collapse
Affiliation(s)
- Seth M. Moore
- Department of Biochemistry, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Institute for Myelin and Glia Exploration, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Eunbi Jeong
- Department of Biochemistry, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Institute for Myelin and Glia Exploration, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Muhammad Zahid
- Department of Biological Sciences, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Institute for Myelin and Glia Exploration, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Joseph Gawron
- Department of Biochemistry, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Institute for Myelin and Glia Exploration, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Simar Arora
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, USA
| | - Sophie Belin
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, USA
| | - Fraser Sim
- Department of Pharmacology and Toxicology, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Yannick Poitelon
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, USA
| | - M. Laura Feltri
- Department of Biochemistry, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Institute for Myelin and Glia Exploration, Jacob’s School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
4
|
Zhou C, Sun C, Zhou W, Tian T, Schultz DC, Wu T, Yu M, Wu L, Pi L, Li C. Development of Novel Indole-Based Covalent Inhibitors of TEAD as Potential Antiliver Cancer Agents. J Med Chem 2024; 67:16270-16295. [PMID: 39270302 DOI: 10.1021/acs.jmedchem.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Abnormal activation of the YAP transcriptional signaling pathway drives proliferation in many hepatocellular carcinoma (HCC) and hepatoblastoma (HB) cases. Current treatment options often face resistance and toxicity, highlighting the need for alternative therapies. This article reports the discovery of a hit compound C-3 from docking-based virtual screening targeting TEAD lipid binding pocket, which inhibited TEAD-mediated transcription. Optimization led to the identification of a potent and covalent inhibitor CV-4-26 that exhibited great antitumor activity in HCC and HB cell lines in vitro, xenografted human HCC, and murine HB in vivo. These outcomes signify the potential of a highly promising therapeutic candidate for addressing a subset of HCC and HB cancers. In the cases of current treatment challenges due to high upregulation of YAP-TEAD activity, these findings offer a targeted alternative for more effective interventions against liver cancer.
Collapse
Affiliation(s)
- Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Chunbao Sun
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Wei Zhou
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Tian Tian
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Daniel C Schultz
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Mu Yu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32610, United States
- UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida 32610, United States
- UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
- UF Institute of Genetics, University of Florida, Gainesville, Florida 32610, United States
| | - Liya Pi
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, Louisiana 70112, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
- Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
5
|
Prencipe G, Cerveró-Varona A, Perugini M, Sulcanese L, Iannetta A, Haidar-Montes AA, Stöckl J, Canciello A, Berardinelli P, Russo V, Barboni B. Amphiregulin orchestrates the paracrine immune-suppressive function of amniotic-derived cells through its interplay with COX-2/PGE 2/EP4 axis. iScience 2024; 27:110508. [PMID: 39156643 PMCID: PMC11326934 DOI: 10.1016/j.isci.2024.110508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/10/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
The paracrine crosstalk between amniotic-derived membranes (AMs)/epithelial cells (AECs) and immune cells is pivotal in tissue healing following inflammation. Despite evidence collected to date, gaps in understanding the underlying molecular mechanisms have hindered clinical applications. The present study represents a significant step forward demonstrating that amphiregulin (AREG) orchestrates the native immunomodulatory functions of amniotic derivatives via the COX-2/PGE2/EP4 axis. The results highlight the immunosuppressive efficacy of PGE2-dependent AREG release, dampening PBMCs' activation, and NFAT pathway in Jurkat reporter cells via TGF-β signaling. Moreover, AREG emerges as a key protein mediator by attenuating acute inflammatory response in Tg(lysC:DsRed2) zebrafish larvae. Notably, the interplay of diverse COX-2/PGE2 pathway activators enables AM/AEC to adapt rapidly to external stimuli (LPS and/or stretching) through a responsive positive feedback loop on the AREG/EGFR axis. These findings offer valuable insights for developing innovative cell-free therapies leveraging the potential of amniotic derivatives in immune-mediated diseases and regenerative medicine.
Collapse
Affiliation(s)
- Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Monia Perugini
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Annamaria Iannetta
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Arlette Alina Haidar-Montes
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna 1090, Austria
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
6
|
Zhang H, Mao Z, Yang Z, Nakamura F. Identification of Filamin A Mechanobinding Partner III: SAV1 Specifically Interacts with Filamin A Mechanosensitive Domain 21. Biochemistry 2023; 62:1197-1208. [PMID: 36857526 DOI: 10.1021/acs.biochem.2c00665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Filamin A (FLNA) cross-links actin filaments and mediates mechanotransduction by force-induced conformational changes of its domains. FLNA's mechanosensitive immunoglobulin-like repeats (R) interact with each other to create cryptic binding sites, which can be exposed by physiologically relevant mechanical forces. Using the FLNA mechanosensing domains as an affinity ligand followed by stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, we recently identified smoothelin and fimbacin as FLNA mechanobinding proteins. Here, using the mechanosensing domain as an affinity ligand and two labeled amino acids, we identify salvador homologue 1 (SAV1), a component of the Hippo pathway kinase cascade, as a new FLNA mechanobinding partner. We demonstrate that SAV1 specifically interacts with the cryptic C-D cleft of FLNA R21 and map the FLNA-binding site on SAV1. We show that point mutations on the R21 C strand block the SAV1 interaction and find that SAV1 contains a FLNA-binding motif in the central region (116Phe-124Val). Point mutations F116A and T118A (FT/AA) disrupt the interaction. A proximity ligation assay reveals that their interaction occurs in the cytosol in an actin polymerization-dependent manner. Although SAV1 is typically found in the cytosol, disrupting the interaction between SAV1 and FLNA causes SAV1 to diffuse to the nucleus and YAP1 to diffuse to the cytosol in an inverse relationship. These results suggest that FLNA mediates regulation of the Hippo pathway through actin polymerization-dependent interaction with SAV1.
Collapse
Affiliation(s)
- Huaguan Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Zhenfeng Mao
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Ziwei Yang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
7
|
Yang MG, Ling E, Cowley CJ, Greenberg ME, Vierbuchen T. Characterization of sequence determinants of enhancer function using natural genetic variation. eLife 2022; 11:76500. [PMID: 36043696 PMCID: PMC9662815 DOI: 10.7554/elife.76500] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 08/30/2022] [Indexed: 02/04/2023] Open
Abstract
Sequence variation in enhancers that control cell-type-specific gene transcription contributes significantly to phenotypic variation within human populations. However, it remains difficult to predict precisely the effect of any given sequence variant on enhancer function due to the complexity of DNA sequence motifs that determine transcription factor (TF) binding to enhancers in their native genomic context. Using F1-hybrid cells derived from crosses between distantly related inbred strains of mice, we identified thousands of enhancers with allele-specific TF binding and/or activity. We find that genetic variants located within the central region of enhancers are most likely to alter TF binding and enhancer activity. We observe that the AP-1 family of TFs (Fos/Jun) are frequently required for binding of TEAD TFs and for enhancer function. However, many sequence variants outside of core motifs for AP-1 and TEAD also impact enhancer function, including sequences flanking core TF motifs and AP-1 half sites. Taken together, these data represent one of the most comprehensive assessments of allele-specific TF binding and enhancer function to date and reveal how sequence changes at enhancers alter their function across evolutionary timescales.
Collapse
Affiliation(s)
- Marty G Yang
- Department of Neurobiology, Harvard Medical School, Boston, United States.,Program in Neuroscience, Harvard Medical School, Boston, United States
| | - Emi Ling
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | | | | | - Thomas Vierbuchen
- Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, United States.,Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, New York, United States
| |
Collapse
|
8
|
Li J, Jiang X, Xu Y, Kang P, Huang P, Meng N, Wang H, Zheng W, Wang H, Wang Z, Zhong X, Cui Y. YY1-induced DLEU1/miR-149-5p Promotes Malignant Biological Behavior of Cholangiocarcinoma through Upregulating YAP1/TEAD2/SOX2. Int J Biol Sci 2022; 18:4301-4315. [PMID: 35864972 PMCID: PMC9295058 DOI: 10.7150/ijbs.66224] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Cholangiocarcinoma is an extremely malignant cancer with poor prognosis. Finding efficient diagnosis and treatment is the indispensable way to improve the prognosis of CCA patients. Therefore, exploring molecular abnormalities in CCA development is urgently needed. DLEU1 is a potential tumor-related lncRNA and abnormally expressed in multiple cancers. In this study, TCGA data analysis showed upregulation of DLEU1 expression in CCA. Furthermore, we confirmed that DLEU1 expression was increased in CCA tissues and cells compared with corresponding controls. Upregulated DLEU1 was related to poor clinicopathological characteristics. Functionally, silencing DLEU1 inhibited CCA proliferation, invasion, stemness maintenance and chemo-resistance, whereas amplifying DLEU1 promoted malignant biological behavior of CCA cells. Mechanistically, DLEU1 expression was transcriptionally facilitated by transcription factor YY1. Moreover, DLEU1 promoted oncogene YAP1 expression by functioning as a sponge to competitively bind to miR-149-5p. YAP1 promoted CCA proliferation, invasion and stemness maintenance, whereas miR-149-5p inhibited malignant biological behavior of CCA. Rescue experiments confirmed that the cancer-promoting effect of DLEU1 was saved by interfering miR-149-5p or YAP1. Furthermore, YAP1 promoted tumor stemness maintenance partly by acting as a transcriptional coactivator to promote TEAD2-induced SOX2 expression. These findings indicated that YY1-induced DLEU1 played a crucial role in CCA progression via miR-149-5p/YAP1/TEAD2/SOX2 axis.
Collapse
Affiliation(s)
- Jinglin Li
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Xingming Jiang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Yi Xu
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Pengcheng Kang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Peng Huang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Nanfeng Meng
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Hang Wang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Wangyang Zheng
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Hao Wang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Zhidong Wang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Xiangyu Zhong
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| | - Yunfu Cui
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 148 Baojian Street, Harbin 150086, Heilongjiang Province, China
| |
Collapse
|
9
|
Liu X, Liu L, Zhao J, Wang H, Li Y. Mechanotransduction regulates inflammation responses of epicardial adipocytes in cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1080383. [PMID: 36589802 PMCID: PMC9800500 DOI: 10.3389/fendo.2022.1080383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is a crucial regulator in maintaining cardiovascular homeostasis by secreting various bioactive products to mediate the physiological function of the cardiovascular system. Accumulating evidence shows that adipose tissue disorders contribute to several kinds of cardiovascular disease (CVD). Furthermore, the adipose tissue would present various biological effects depending on its tissue localization and metabolic statuses, deciding the individual cardiometabolic risk. Crosstalk between adipose and myocardial tissue is involved in the pathophysiological process of arrhythmogenic right ventricular cardiomyopathy (ARVC), cardiac fibrosis, heart failure, and myocardial infarction/atherosclerosis. The abnormal distribution of adipose tissue in the heart might yield direct and/or indirect effects on cardiac function. Moreover, mechanical transduction is critical for adipocytes in differentiation, proliferation, functional maturity, and homeostasis maintenance. Therefore, understanding the features of mechanotransduction pathways in the cellular ontogeny of adipose tissue is vital for underlining the development of adipocytes involved in cardiovascular disorders, which would preliminarily contribute positive implications on a novel therapeutic invention for cardiovascular diseases. In this review, we aim to clarify the role of mechanical stress in cardiac adipocyte homeostasis and its interplay with maintaining cardiac function.
Collapse
Affiliation(s)
- Xiaoliang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfei Zhao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Hua Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| |
Collapse
|
10
|
Hadjittofi C, Feretis M, Martin J, Harper S, Huguet E. Liver regeneration biology: Implications for liver tumour therapies. World J Clin Oncol 2021; 12:1101-1156. [PMID: 35070734 PMCID: PMC8716989 DOI: 10.5306/wjco.v12.i12.1101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/22/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
The liver has remarkable regenerative potential, with the capacity to regenerate after 75% hepatectomy in humans and up to 90% hepatectomy in some rodent models, enabling it to meet the challenge of diverse injury types, including physical trauma, infection, inflammatory processes, direct toxicity, and immunological insults. Current understanding of liver regeneration is based largely on animal research, historically in large animals, and more recently in rodents and zebrafish, which provide powerful genetic manipulation experimental tools. Whilst immensely valuable, these models have limitations in extrapolation to the human situation. In vitro models have evolved from 2-dimensional culture to complex 3 dimensional organoids, but also have shortcomings in replicating the complex hepatic micro-anatomical and physiological milieu. The process of liver regeneration is only partially understood and characterized by layers of complexity. Liver regeneration is triggered and controlled by a multitude of mitogens acting in autocrine, paracrine, and endocrine ways, with much redundancy and cross-talk between biochemical pathways. The regenerative response is variable, involving both hypertrophy and true proliferative hyperplasia, which is itself variable, including both cellular phenotypic fidelity and cellular trans-differentiation, according to the type of injury. Complex interactions occur between parenchymal and non-parenchymal cells, and regeneration is affected by the status of the liver parenchyma, with differences between healthy and diseased liver. Finally, the process of termination of liver regeneration is even less well understood than its triggers. The complexity of liver regeneration biology combined with limited understanding has restricted specific clinical interventions to enhance liver regeneration. Moreover, manipulating the fundamental biochemical pathways involved would require cautious assessment, for fear of unintended consequences. Nevertheless, current knowledge provides guiding principles for strategies to optimise liver regeneration potential.
Collapse
Affiliation(s)
- Christopher Hadjittofi
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Michael Feretis
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Jack Martin
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Simon Harper
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Emmanuel Huguet
- University Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Center, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
11
|
Xu Y, Wang X, Yu M, Ruan Y, Zhang J, Tian Y, Xiong J, Liu L, Cheng Y, Yang Y, Ren B, Chen G, Zhang Y, Zhao B, Wang J, Wang J, Jian R, Liu Y, Wang J. Identification, subcellular localization, and functional comparison of novel Yap splicing isoforms in mouse embryonic stem cells. IUBMB Life 2021; 73:1432-1445. [PMID: 34687583 DOI: 10.1002/iub.2571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/11/2021] [Indexed: 11/06/2022]
Abstract
Hippo signaling pathway is involved in many biological processes including the fate decision of embryonic stem cells (ESCs). Yes-associated protein (Yap) function as a key effector of Hippo pathway, but its role in ESCs is still controversial. So far, only two isoforms of Yap have been identified and they have both overlapping and distinct functions. Here, we identify six novel isoforms of mouse Yap, bringing the total number of isoforms to eight. According to the differences in the first exon, they are divided into two subtypes (a and b). Isoform-a and isoform-b exhibit different subcellular localizations. Moreover, isoform-a can fully reverse the impaired self-renewal phenotype induced by Yap knockout (KO). Upon overexpression, isoform-a moderately promotes mESCs self-renewal and markedly delays differentiation. On the contrary, no significant pro-self-renewal phenotype is observed when isoform-b overexpressed in wildtype (WT) mESCs or re-expressed in Yap KO cell lines. These finding not only help to clarify the role of Yap in mESCs, but also lay the foundation for advancing functional researches of Yap in other processes.
Collapse
Affiliation(s)
- Yixiao Xu
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing, China.,Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Xueyue Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China.,Department of Pediatrics, The General Hospital of PLA Tibet Military Area Command, Lhasa, China
| | - Meng Yu
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China.,Joint Surgery Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yan Ruan
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Junlei Zhang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Yanping Tian
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Jiaxiang Xiong
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Lianlian Liu
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Yuda Cheng
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Yi Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Bangqi Ren
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Guangxing Chen
- Joint Surgery Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yue Zhang
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing, China.,Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Binyu Zhao
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China.,Department of Physiology, Army Medical University, Chongqing, China
| | - Jiaqi Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China.,Institute of Immunology PLA and Department of Immunology, Army Medical University, Chongqing, China
| | - Jiangjun Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China.,Department of Cell Biology, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Rui Jian
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Army Medical University, Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Jiali Wang
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Sun T, Peng H, Mao W, Ma L, Liu H, Mai J, Jiao L. Autophagy-mediated negative feedback attenuates the oncogenic activity of YAP in pancreatic cancer. Int J Biol Sci 2021; 17:3634-3645. [PMID: 34512171 PMCID: PMC8416727 DOI: 10.7150/ijbs.61795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal malignancy in humans, and new therapeutic targets are urgently needed. Yes-associated protein (YAP) plays a significant role in cancer progression. Autophagy is also closely associated with various human cancers. However, the interplay between YAP and autophagy in PDAC remains poorly understood. In this study, we found that YAP was upregulated and activated in PDAC. Further analysis revealed that there is a YAP-autophagy feedback loop in pancreatic cancer. Mechanistically, YAP activates autophagy by promoting Atg5 transcription via TEAD1-mediated binding, while autophagy negatively regulates YAP through autophagic degradation. The hyperactivation of YAP in PDAC unbalances the YAP-autophagy circuit and promotes cancer progression. Inhibition of autophagy enhances the oncogenic activity of YAP in PDAC. The autophagy activator rapamycin promotes the antitumor effect of verteporfin, a YAP inhibitor. Therefore, our study elucidated the interaction between YAP and autophagy in PDAC and our results suggest that targeting the YAP-autophagy circuit may be a new therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hui Peng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wenhao Mao
- Department of Clinical Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jia Mai
- Department of Laboratory Medicine, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
13
|
Luo J, Zou H, Deng L, Sun X, Yuan P, Li P. Lin28 Inhibits the Differentiation from Mouse Embryonic Stem Cells to Glial Lineage Cells through Upregulation of Yap1. Stem Cells Int 2021; 2021:6674283. [PMID: 33688355 PMCID: PMC7920735 DOI: 10.1155/2021/6674283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
The RNA-binding protein Lin28 regulates neurogliogenesis in mammals, independently of the let-7 microRNA. However, the detailed regulatory mechanism remains obscured. Here, we established Lin28a or Lin28b overexpression mouse embryonic stem cells (ESCs) and found that these cells expressed similar levels of the core pluripotent factors, such as Oct4 and Sox2, and increased Yap1 but decreased lineage-specific markers compared to the control ESCs. Further differentiation of these ESCs to neuronal and glial lineage cells revealed that Lin28a/b overexpression did not affect the expression of neuronal marker βIII-tubulin, but dramatically inhibited the glial lineage markers, such as Gfap and Mbp. Interestingly, overexpression of Yap1 in mouse ESCs phenocopied Lin28a/b overexpression ESCs by showing defect in glial cell differentiation. Inhibition of Yap1/Tead-mediated transcription with verteporfin partially rescued the differentiation defect of Lin28a/b overexpression ESCs. Mechanistically, we demonstrated that Lin28 can directly bind to Yap1 mRNA, and the induction of Yap1 by Lin28a in mESCs is independent of Let7. Taken together, our results unravel a novel Lin28-Yap1 regulatory axis during mESC to glial lineage cell differentiation, which may shed light on glial cell generation in vitro.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Liang Deng
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiang Sun
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Ping Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
14
|
LeBlanc L, Ramirez N, Kim J. Context-dependent roles of YAP/TAZ in stem cell fates and cancer. Cell Mol Life Sci 2021; 78:4201-4219. [PMID: 33582842 PMCID: PMC8164607 DOI: 10.1007/s00018-021-03781-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/30/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
Hippo effectors YAP and TAZ control cell fate and survival through various mechanisms, including transcriptional regulation of key genes. However, much of this research has been marked by conflicting results, as well as controversy over whether YAP and TAZ are redundant. A substantial portion of the discordance stems from their contradictory roles in stem cell self-renewal vs. differentiation and cancer cell survival vs. apoptosis. In this review, we present an overview of the multiple context-dependent functions of YAP and TAZ in regulating cell fate decisions in stem cells and organoids, as well as their mechanisms of controlling programmed cell death pathways in cancer.
Collapse
Affiliation(s)
- Lucy LeBlanc
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Nereida Ramirez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.,Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA. .,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
15
|
Gu Y, Wang Y, Wang Y, Luo J, Wang X, Ma M, Hua W, Liu Y, Yu FX. Hypermethylation of LATS2 Promoter and Its Prognostic Value in IDH-Mutated Low-Grade Gliomas. Front Cell Dev Biol 2020; 8:586581. [PMID: 33195240 PMCID: PMC7642219 DOI: 10.3389/fcell.2020.586581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in the enzyme isocitrate dehydrogenase 1/2 (IDH1/2) are the most common somatic mutations in low-grade glioma (LGG). The Hippo signaling pathway is known to play a key role in organ size control, and its dysregulation is involved in the development of diverse cancers. Large tumor suppressor 1/2 (LATS1/2) are core Hippo pathway components that phosphorylate and inactivate Yes-associated protein (YAP), a transcriptional co-activator that regulates expression of genes involved in tumorigenesis. A recent report from The Cancer Genome Atlas (TCGA) has highlighted a frequent hypermethylation of LATS2 in IDH-mutant LGG. However, it is unclear if LATS2 hypermethylation is associated with YAP activation and prognosis of LGG patients. Here, we performed a network analysis of the status of the Hippo pathway in IDH-mutant LGG samples and determined its association with cancer prognosis. Combining TCGA data with our biochemical assays, we found hypermethylation of LATS2 promoter in IDH-mutant LGG. LATS2 hypermethylation, however, did not translate into YAP activation but highly correlated with IDH mutation. LATS2 hypermethylation may thus serve as an alternative for IDH mutation in diagnosis and a favorable prognostic factor for LGG patients.
Collapse
Affiliation(s)
- Yuan Gu
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yebin Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiaqian Luo
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingyue Ma
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Eyeing the Extracellular Matrix in Vascular Development and Microvascular Diseases and Bridging the Divide between Vascular Mechanics and Function. Int J Mol Sci 2020; 21:ijms21103487. [PMID: 32429045 PMCID: PMC7278940 DOI: 10.3390/ijms21103487] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
The extracellular matrix (ECM) is critical in all aspects of vascular development and health: supporting cell anchorage, providing structure, organization and mechanical stability, and serving as a sink for growth factors and sustained survival signals. Abnormal changes in ECM protein expression, organization, and/or properties, and the ensuing changes in vascular compliance affect vasodilator responses, microvascular pressure transmission, and collateral perfusion. The changes in microvascular compliance are independent factors initiating, driving, and/or exacerbating a plethora of microvascular diseases of the eye including diabetic retinopathy (DR) and vitreoretinopathy, retinopathy of prematurity (ROP), wet age-related macular degeneration (AMD), and neovascular glaucoma. Congruently, one of the major challenges with most vascular regenerative therapies utilizing localized growth factor, endothelial progenitor, or genetically engineered cell delivery, is the regeneration of blood vessels with physiological compliance properties. Interestingly, vascular cells sense physical forces, including the stiffness of their ECM, through mechanosensitive integrins, their associated proteins and the actomyosin cytoskeleton, which generates biochemical signals that culminate in a rapid expression of matricellular proteins such as cellular communication network 1 (CCN1) and CCN2 (aka connective tissue growth factor or CTGF). Loss or gain of function of these proteins alters genetic programs of cell growth, ECM biosynthesis, and intercellular signaling, that culminate in changes in cell behavior, polarization, and barrier function. In particular, the function of the matricellular protein CCN2/CTGF is critical during retinal vessel development and regeneration wherein new blood vessels form and invest a preformed avascular neural retina following putative gradients of matrix stiffness. These observations underscore the need for further in-depth characterization of the ECM-derived cues that dictate structural and functional properties of the microvasculature, along with the development of new therapeutic strategies addressing the ECM-dependent regulation of pathophysiological stiffening of blood vessels in ischemic retinopathies.
Collapse
|
17
|
Bokhovchuk F, Mesrouze Y, Delaunay C, Martin T, Villard F, Meyerhofer M, Fontana P, Zimmermann C, Erdmann D, Furet P, Scheufler C, Schmelzle T, Chène P. Identification of FAM181A and FAM181B as new interactors with the TEAD transcription factors. Protein Sci 2019; 29:509-520. [PMID: 31697419 DOI: 10.1002/pro.3775] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022]
Abstract
The Hippo pathway is a key signaling pathway in the control of organ size and development. The most distal elements of this pathway, the TEAD transcription factors, are regulated by several proteins, such as YAP (Yes-associated protein), TAZ (transcriptional co-activator with PDZ-binding motif) and VGLL1-4 (Vestigial-like members 1-4). In this article, combining structural data and motif searches in protein databases, we identify two new TEAD interactors: FAM181A and FAM181B. Our structural data show that they bind to TEAD via an Ω-loop as YAP/TAZ do, but only FAM181B possesses the LxxLF motif (x any amino acid) found in YAP/TAZ. The affinity of different FAM181A/B fragments for TEAD is in the low micromolar range and full-length FAM181A/B proteins interact with TEAD in cells. These findings, together with a recent report showing that FAM181A/B proteins have a role in nervous system development, suggest a potential new involvement of the TEAD transcription factors in the development of this tissue.
Collapse
Affiliation(s)
- Fedir Bokhovchuk
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Yannick Mesrouze
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Clara Delaunay
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Typhaine Martin
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Frédéric Villard
- Chemical Biology & Therapeutics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marco Meyerhofer
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Patrizia Fontana
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Catherine Zimmermann
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Dirk Erdmann
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Pascal Furet
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Clemens Scheufler
- Chemical Biology & Therapeutics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Tobias Schmelzle
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Patrick Chène
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
18
|
Cui P, Liu X, Zhao K, Hou S, Chen C, Zhao D, Zeng H. The novel axis of YAP1, transcription enhancer factor 3 and Down Syndrome Candidate Region 1 isoform 1L is a common signaling pathway downstream of several angiogenic factors. Microvasc Res 2019; 129:103955. [PMID: 31733305 DOI: 10.1016/j.mvr.2019.103955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/06/2019] [Accepted: 11/11/2019] [Indexed: 01/06/2023]
Abstract
Angiogenesis is a hallmark of many diseases. Previously, we found that Down Syndrome Candidate Region 1 Isoform 1L (DSCR1-1L) was expressed in human tumor vessels, but was not detectable in normal tissues, and played important roles in angiogenesis induced by vascular endothelial growth factor (VEGF-A165). The expressions of DSCR1-1L mRNA and protein induced by VEGF-A165 were regulated via the direct interaction of transcription enhancer factor 3 (TEF3) with DSCR1-1L promoter. However, the function and the regulation of DSCR1-1L in angiogenesis had not been completely understood. In this study, we found that the expressions of DSCR1-1L mRNA and proteins were upregulated by other angiogenic factors, including VEGF-A121, VEGF-E, histamine, PAF, the endothelial cell (EC) growth medium, and the conditional medium obtained from cancer cells, but not by PlGF, bFGF, PDGF, and serotonin. The EC proliferation, migration and elongation induced by histamine and EC growth medium were inhibited by knocking down the mRNA and protein expressions of DSCR1-1L and TEF3. The TEF3 activation was regulated by its interaction with YAP1, and translocation from cytosol to nuclei, but not by increase of protein expression, after the stimulation of VEGF, histamine and EC growth medium. YAP1 regulated the protein expression of DSCR1-1L, the proliferation, migration and elongation of ECs induced by VEGF, histamine and EC growth medium. Taken together, this study identified a novel axis of YAP1, TEF3 and DSCR1-1L that was a common signaling pathway downstream of several angiogenic factors to regulate angiogenesis, suggesting that this pathway is an excellent therapeutic target for angiogenic diseases and cancers. Our results contribute significantly to the field of mechanistic studies.
Collapse
Affiliation(s)
- Pengfei Cui
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Pancreatic Disease Institute, Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xin Liu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Kevin Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Shiqiang Hou
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Surgery of Breast and Thyroid, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
19
|
Pantera H, Shy ME, Svaren J. Regulating PMP22 expression as a dosage sensitive neuropathy gene. Brain Res 2019; 1726:146491. [PMID: 31586623 DOI: 10.1016/j.brainres.2019.146491] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022]
Abstract
Structural variation in the human genome has emerged as a major cause of disease as genomic data have accumulated. One of the most common structural variants associated with human disease causes the heritable neuropathy known as Charcot-Marie-Tooth (CMT) disease type 1A. This 1.4 Mb duplication causes nearly half of the CMT cases that are genetically diagnosed. The PMP22 gene is highly induced in Schwann cells during development, although its precise role in myelin formation and homeostasis is still under active investigation. The PMP22 gene can be considered as a nucleoprotein complex with enzymatic activity to produce the PMP22 transcript, and the complex is allosterically regulated by transcription factors that respond to intracellular signals and epigenomic modifications. The control of PMP22 transcript levels has been one of the major therapeutic targets of therapy development, and this review summarizes those approaches as well as efforts to characterize the regulation of the PMP22 gene.
Collapse
Affiliation(s)
- Harrison Pantera
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, WI, USA
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
20
|
Zhou W, Li Y, Song J, Li C. Fluorescence polarization assay for the identification and evaluation of inhibitors at YAP-TEAD protein-protein interface 3. Anal Biochem 2019; 586:113413. [PMID: 31479631 DOI: 10.1016/j.ab.2019.113413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/31/2019] [Accepted: 08/29/2019] [Indexed: 01/06/2023]
Abstract
The Hippo signaling pathway controls cell-cell contact, cell proliferation, as well as organ size by integrating changes in the cellular microenvironment. In recent years, the pivotal role of Hippo signaling in cancers has been well recognized. Inhibition of the pathway promotes the translocation of the major Hippo pathway effectors, the yes-associated protein (YAP) and its paralog TAZ, to the nucleus, where they interact with the transcription factor family transcriptional enhancer associate domain (TEAD), thus coactivating the expression of downstream genes, leading to cell transformation, tissue overgrowth, and tumor development. Therefore, the interruption of the YAP-TEAD transcriptional complex represents a novel opportunity for the treatment of cancer. Here, we established a fluorescence polarization (FP)-based assay for the identification and evaluation of YAP-TEAD protein-protein interface (PPI) inhibitors at the YAP Ω-loop binding region of TEAD, which is also called interface 3 at the YAP-TEAD binding surface. Furthermore, a patented small molecule (Patent-22) was evaluated by the FP assay, which confirmed that it was a YAP-TEAD PPI inhibitor at interface 3. Possessing great application value, this FP method is reliable, robust, and economical for inhibitor assessment and drug discovery.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, United States
| | - Yiping Li
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Jinhua Song
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Chenglong Li
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, United States; Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States; Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL, 32610, United States.
| |
Collapse
|
21
|
Cruzeiro GAV, Lira RCP, de Almeida Magalhães T, Scrideli CA, Valera ET, Baumgartner M, Tone LG. CTGF expression is indicative of better survival rates in patients with medulloblastoma. Cancer Gene Ther 2019; 27:378-382. [PMID: 31073205 DOI: 10.1038/s41417-019-0100-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/27/2019] [Indexed: 11/09/2022]
Abstract
Medulloblastoma (MB) is the most frequent malignant brain tumor in children and it is subgrouped into 4 entities (SHH, WNT, Group 3, and Group 4). Molecular pathways involved in these different subgroups still are evolving and can be of clinical relevance to therapy. The YAP1-CTGF axis is known to regulate cell proliferation, differentiation, and cell death; however, its role in MB is poorly explored. We aimed to investigate the role of YAP1 gene in the MB SHH cell line DAOY and evaluate cell proliferation, doubling time and 3D spheroids invasion and its consequence on CTGF regulation. We assessed CTGF expression from 22 children with MB. Lastly, we validated our findings through in silico analysis in large cohorts dataset of patients. We observed an increased invasion rate of DAOY cells and CTGF downregulation under YAP1 knockdown (p < 0.0001). Additionally CTGF is overexpressed in MB with extensive nodularity subtype and an indicative of higher survival rates in pediatric MB (p < 0.05). Interestingly, no difference of CTGF expression was observed between molecular subgroups. These results provide new evidence ofCTGF as a potential prognostic marker for MB, corroborating to the role of YAP1 in restricting MB cell.
Collapse
Affiliation(s)
- Gustavo Alencastro Veiga Cruzeiro
- Department of Oncology, Children's Research Center, Neuro-Oncology group, University Children's Hospital Zürich, August-Forel Strasse 1, CH-8008, Zürich, Switzerland. .,Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas, University of São Paulo, Av.Bandeirantes 3900, Hospital das Clínicas, Ribeirão Preto, São Paulo, Brazil. .,Edwin L. Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, 100 Blossom Street, Cox 7, Boston, Massachusetts, USA.
| | - Regia Caroline Peixoto Lira
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas, University of São Paulo, Av.Bandeirantes 3900, Hospital das Clínicas, Ribeirão Preto, São Paulo, Brazil
| | - Taciani de Almeida Magalhães
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Carlos Alberto Scrideli
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas, University of São Paulo, Av.Bandeirantes 3900, Hospital das Clínicas, Ribeirão Preto, São Paulo, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas, University of São Paulo, Av.Bandeirantes 3900, Hospital das Clínicas, Ribeirão Preto, São Paulo, Brazil
| | - Martin Baumgartner
- Department of Oncology, Children's Research Center, Neuro-Oncology group, University Children's Hospital Zürich, August-Forel Strasse 1, CH-8008, Zürich, Switzerland
| | - Luiz Gonzaga Tone
- Department of Pediatrics, Ribeirão Preto Medical School, Hospital das Clínicas, University of São Paulo, Av.Bandeirantes 3900, Hospital das Clínicas, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
22
|
Deng LJ, Qi M, Peng QL, Chen MF, Qi Q, Zhang JY, Yao N, Huang MH, Li XB, Peng YH, Liu JS, Fu DR, Chen JX, Ye WC, Zhang DM. Arenobufagin induces MCF-7 cell apoptosis by promoting JNK-mediated multisite phosphorylation of Yes-associated protein. Cancer Cell Int 2018; 18:209. [PMID: 30574018 PMCID: PMC6299615 DOI: 10.1186/s12935-018-0706-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
Background It has been demonstrated that bufadienolides exert potent anti-cancer activity in various tumor types. However, the mechanisms that underlie their anti-cancer properties remain unclear. Yes-associated protein, a key effector of Hippo signaling, functions as a transcription coactivator, plays oncogenic and tumor suppressor roles under different conditions. Here, we report that arenobufagin (ABF), a representative bufadienolide, induced breast cancer MCF-7 cells to undergo apoptosis, which occurred through the JNK-mediated multisite phosphorylation of YAP. Methods Cytotoxicity was examined using an MTT assay. ABF-induced apoptosis was measured with a TUNEL assay and Annexin V-FITC/PI double staining assay. Western blotting, immunofluorescence, qRT-PCR and coimmunoprecipitation were employed to assess the expression levels of the indicated molecules. Lose-of-function experiments were carried out with siRNA transfection and pharmacological inhibitors. ABF-induced phosphopeptides were enriched with Ti4+-IMAC chromatography and further subjected to reverse-phase nano-LC–MS/MS analysis. Results ABF significantly reduced the viability of MCF-7 cells and increased the percentage of early and late apoptotic cells in a concentration- and time-dependent manner. Following ABF treatment, YAP accumulated in the nucleus and bound to p73, which enhanced the transcription of the pro-apoptotic genes Bax and p53AIP1. YAP knock-down significantly attenuated ABF-induced apoptotic cell death. Importantly, we found that the mobility shift of YAP was derived from its phosphorylation at multiple sites, including Tyr357. Moreover, mass spectrometry analysis identified 19 potential phosphorylation sites in YAP, with a distribution of 14 phosphoserine and 5 phosphothreonine residues. Furthermore, we found that the JNK inhibitor SP600125 completely diminished the mobility shift of YAP and its phosphorylation at Tyr357, the binding of YAP and p73, the transcription of Bax and p53AIP1 as well as the apoptosis induced by ABF. These data indicate that ABF induced YAP multisite phosphorylation, which was associated with p73 binding, and that apoptosis was mediated by the JNK signaling pathway. Conclusions Our data demonstrate that ABF suppresses MCF-7 breast cancer proliferation by triggering the pro-apoptotic activity of YAP, which is mediated by JNK signaling-induced YAP multisite phosphorylation as well as its association with p73. The present work not only provides additional information on the use of ABF as an anti-breast cancer drug, but also offers evidence that the induction of the tumor suppressor role of YAP may be a therapeutic strategy. Electronic supplementary material The online version of this article (10.1186/s12935-018-0706-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Juan Deng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,2Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Ming Qi
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Qun-Long Peng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Min-Feng Chen
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Qi Qi
- 4Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Jia-Yan Zhang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Nan Yao
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Mao-Hua Huang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Xiao-Bo Li
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Yin-Hui Peng
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Jun-Shan Liu
- 5School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515 People's Republic of China
| | - Deng-Rui Fu
- Guangzhou Yucai Middle School, Fujin Road 2#, Dongshan District, Guangzhou, China
| | - Jia-Xu Chen
- 2Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Wen-Cai Ye
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| | - Dong-Mei Zhang
- 1Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632 China.,3College of Pharmacy, Jinan University, Guangzhou, 510632 People's Republic of China
| |
Collapse
|
23
|
Abstract
Hippo signaling plays critical roles in regulation of tissue homeostasis, organ size, and tumorigenesis by inhibiting YES-associated protein (YAP) and PDZ-binding protein TAZ through MST1/2 and LATS1/2 pathway. It is also engaged in cross-talk with various other signaling pathways, including WNT, BMPs, Notch, GPCRs, and Hedgehog to further modulate activities of YAP/TAZ. Because YAP and TAZ are transcriptional coactivators that lack DNA-binding activity, both proteins must interact with DNA-binding transcription factors to regulate target gene’s expression. To activate target genes involved in cell proliferation, TEAD family members are major DNA-binding partners of YAP/TAZ. Accordingly, YAP/TAZ were originally classified as oncogenes. However, YAP might also play tumor-suppressing role. For example, YAP can bind to DNA-binding tumor suppressors including RUNXs and p73. Thus, YAP might act either as an oncogene or tumor suppressor depending on its binding partners. Here, we summarize roles of YAP depending on its DNA-binding partners and discuss context-dependent functions of YAP/TAZ.
Collapse
Affiliation(s)
- Min-Kyu Kim
- Department of Biochemistry, College of Medicine, and Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Ju-Won Jang
- Department of Biochemistry, College of Medicine, and Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| | - Suk-Chul Bae
- Department of Biochemistry, College of Medicine, and Institute for Tumor Research, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
24
|
Zhu JY, Lin S, Ye J. YAP and TAZ, the conductors that orchestrate eye development, homeostasis, and disease. J Cell Physiol 2018; 234:246-258. [PMID: 30094836 DOI: 10.1002/jcp.26870] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 12/25/2022]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators established as a nexus in numerous signaling pathways, notably in Hippo signaling. Previous research revealed multifarious function of YAP and TAZ in oncology and cardiovasology. Recently, the focus has been laid on their pivotal role in eye morphogenesis and homeostasis. In this review, we synthesize advances of YAP and TAZ function during eye development in different model organisms, introduce their function in different ocular tissues and eye diseases, and highlight the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Jing-Yi Zhu
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
25
|
Gong P, Zhang Z, Zou C, Tian Q, Chen X, Hong M, Liu X, Chen Q, Xu Z, Li M, Wang J. Hippo/YAP signaling pathway mitigates blood-brain barrier disruption after cerebral ischemia/reperfusion injury. Behav Brain Res 2018; 356:8-17. [PMID: 30092249 DOI: 10.1016/j.bbr.2018.08.003] [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: 06/06/2018] [Revised: 07/23/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022]
Abstract
Ischemia/reperfusion (I/R) injuries commonly lead to breakdown of the blood-brain barrier (BBB). Restoration of the BBB can relieve neurologic damage caused by I/R injuries. The Hippo/YAP signaling pathway mediates cell proliferation, regulated cell death, and differentiation in various organisms and has been shown to participate in the restoration of the heart after I/R. In this study, we investigated whether the Hippo/YAP pathway plays a role in I/R injury in brain, especially in regard to I/R-induced BBB breakdown. The results of our study indicate that I/R injury led to an overall decrease in activity of the core proteins, YAP and TAZ, over a 24-h period. The most dramatic change was observed 1.5 h after reperfusion. In rats that underwent 1.5 h of reperfusion, intraperitoneal injection of YAP agonist dexamethasone activated YAP and TAZ and led to improved neurologic function, smaller brain infarct sizes, increased levels of tight junction proteins, decreased BBB permeability, decreased cerebral edema, and less apoptosis. Our results suggest that YAP exerts neuroprotective effects on the damaged brain that are likely related to restoration of the BBB.
Collapse
Affiliation(s)
- Pian Gong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhan Zhang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Changlin Zou
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, PR China
| | - Michael Hong
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xi Liu
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Type I collagen-induced YAP nuclear expression promotes primary cilia growth and contributes to cell migration in confluent mouse embryo fibroblast 3T3-L1 cells. Mol Cell Biochem 2018; 450:87-96. [DOI: 10.1007/s11010-018-3375-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 05/23/2018] [Indexed: 12/18/2022]
|
27
|
Huang T, Zhou Y, Zhang J, Cheng ASL, Yu J, To KF, Kang W. The physiological role of Motin family and its dysregulation in tumorigenesis. J Transl Med 2018; 16:98. [PMID: 29650031 PMCID: PMC5898069 DOI: 10.1186/s12967-018-1466-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/28/2018] [Indexed: 11/30/2022] Open
Abstract
Members in Motin family, or Angiomotins (AMOTs), are adaptor proteins that localize in the membranous, cytoplasmic or nuclear fraction in a cell context-dependent manner. They control the bioprocesses such as migration, tight junction formation, cell polarity, and angiogenesis. Emerging evidences have demonstrated that AMOTs participate in cancer initiation and progression. Many of the previous studies have focused on the involvement of AMOTs in Hippo-YAP1 pathway. However, it has been controversial for years that AMOTs serve as either positive or negative growth regulators in different cancer types because of the various cellular origins. The molecular mechanisms of these opposite roles of AMOTs remain elusive. This review comprehensively summarized how AMOTs function physiologically and how their dysregulation promotes or inhibits tumorigenesis. Better understanding the functional roles of AMOTs in cancers may lead to an improvement of clinical interventions as well as development of novel therapeutic strategies for cancer patients.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China
| | - Jinglin Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China
| | - Alfred S L Cheng
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, People's Republic of China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
| |
Collapse
|
28
|
Mesrouze Y, Meyerhofer M, Bokhovchuk F, Fontana P, Zimmermann C, Martin T, Delaunay C, Izaac A, Kallen J, Schmelzle T, Erdmann D, Chène P. Effect of the acylation of TEAD4 on its interaction with co-activators YAP and TAZ. Protein Sci 2017; 26:2399-2409. [PMID: 28960584 DOI: 10.1002/pro.3312] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/30/2017] [Accepted: 09/25/2017] [Indexed: 01/02/2023]
Abstract
The Hippo pathway is deregulated in various cancers, and the discovery of molecules that modulate this pathway may open new therapeutic avenues in oncology. TEA/ATTS domain (TEAD) transcription factors are the most distal elements of the Hippo pathway and their transcriptional activity is regulated by the Yes-associated protein (YAP). Amongst the various possibilities for targeting this pathway, inhibition of the YAP:TEAD interaction is an attractive strategy. It has been shown recently that TEAD proteins are covalently linked via a conserved cysteine to a fatty acid molecule (palmitate) that binds to a deep hydrophobic cavity present in these proteins. This acylation of TEAD seems to be required for efficient binding to YAP, and understanding how it modulates the YAP:TEAD interaction may provide useful information on the regulation of TEAD function. In this report we have studied the effect of TEAD4 acylation on its interaction with YAP and the other co-activator transcriptional co-activator with PDZ-binding motif (TAZ). We show in our biochemical and cellular assays that YAP and TAZ bind in a similar manner to acylated and non-acylated TEAD4. This indicates that TEAD4 acylation is not a prerequisite for its interaction with YAP or TAZ. However, we observed that TEAD4 acylation significantly enhances its stability, suggesting that it may help this transcription factor to acquire and/or maintain its active conformation.
Collapse
Affiliation(s)
- Yannick Mesrouze
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marco Meyerhofer
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Fedir Bokhovchuk
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Patrizia Fontana
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Catherine Zimmermann
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Typhaine Martin
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Clara Delaunay
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Aude Izaac
- Chemical Biology & Therapeutics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Joerg Kallen
- Chemical Biology & Therapeutics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Tobias Schmelzle
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Dirk Erdmann
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Patrick Chène
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
29
|
Michel-Ramirez G, Recio-Vega R, Ocampo-Gomez G, Palacios-Sanchez E, Delgado-Macias M, Delgado-Gaona M, Lantz RC, Gandolfi J, Gonzalez-Cortes T. Association between YAP expression in neoplastic and non-neoplastic breast tissue with arsenic urinary levels. J Appl Toxicol 2017; 37:1195-1202. [PMID: 28524356 DOI: 10.1002/jat.3481] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/07/2017] [Accepted: 03/30/2017] [Indexed: 11/11/2022]
Abstract
The Hippo pathway regulates cell proliferation and apoptosis and it has been noted that loss of critical components of this pathway can lead to uncontrolled cell growth. Yes-associated protein (YAP) is an important component of this Hippo pathway because YAP is the nuclear effector of the Hippo tumor suppressor pathway and it is crucial for the response to oxidative stress induced by cellular process and by different xenobiotics, including arsenic. It has been proposed that YAP dysregulation can contribute to a malignant cellular phenotype acting as both a tumor suppressor and an oncogene. The aim of the study was to assess and compare the expression of YAP in neoplastic and non-neoplastic breast tissue of women chronically exposed to arsenic through drinking water. YAP expression was assessed by immunohistochemistry in 120 breast biopsies from women with breast cancer and from women with other non-neoplastic breast pathologies. Arsenic concentration was quantified in urine. The results disclosed a significant lower percentage of cytoplasm YAP expression in cases and that YAP high-intensity staining in the cytoplasm but not in the nucleus decreases the risk for breast cancer. In conclusion, our overall data suggest that YAP may act as a tumor suppressor protein because their reduced expression in cases, which can induce an environment favorable for inhibition of apoptosis and promoting cellular proliferation by increasing genetic instability of cells, which might contribute to the pathogenesis of cancer. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gladis Michel-Ramirez
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico
| | - Rogelio Recio-Vega
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico
| | - Guadalupe Ocampo-Gomez
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico
| | - Eduardo Palacios-Sanchez
- Department of Gynecologic Oncology, Mexican Institute of Social Security, Torreon, Coahuila, México
| | - Manuel Delgado-Macias
- Department of Medical Education, School of Medicine, University of Coahuila, Torreon, Coahuila, México
| | | | - Robert Clark Lantz
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, USA.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Jay Gandolfi
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Tania Gonzalez-Cortes
- Department of Environmental Health, Biomedical Research Center, School of Medicine, University of Coahuila, Torreon, Coahuila, Mexico
| |
Collapse
|
30
|
Mesrouze Y, Bokhovchuk F, Meyerhofer M, Fontana P, Zimmermann C, Martin T, Delaunay C, Erdmann D, Schmelzle T, Chène P. Dissection of the interaction between the intrinsically disordered YAP protein and the transcription factor TEAD. eLife 2017; 6. [PMID: 28430104 PMCID: PMC5400505 DOI: 10.7554/elife.25068] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/06/2017] [Indexed: 01/23/2023] Open
Abstract
TEAD (TEA/ATTS domain) transcription factors are the most distal effectors of the Hippo pathway. YAP (Yes-associated protein) is a coactivator protein which, upon binding to TEAD proteins, stimulates their transcriptional activity. Since the Hippo pathway is deregulated in various cancers, designing inhibitors of the YAP:TEAD interaction is an attractive therapeutic strategy for oncology. Understanding the molecular events that take place at the YAP:TEAD interface is therefore important not only to devise drug discovery approaches, but also to gain knowledge on TEAD regulation. In this report, combining single site-directed mutagenesis and double mutant analyses, we conduct a detailed analysis on the role of several residues located at the YAP:TEAD interface. Our results provide quantitative understanding of the interactions taking place at the YAP:TEAD interface and give insights into the formation of the YAP:TEAD complex and more particularly on the interaction between TEAD and the Ω-loop found in YAP. DOI:http://dx.doi.org/10.7554/eLife.25068.001
Collapse
Affiliation(s)
- Yannick Mesrouze
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Fedir Bokhovchuk
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marco Meyerhofer
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Patrizia Fontana
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Catherine Zimmermann
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Typhaine Martin
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Clara Delaunay
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Dirk Erdmann
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Tobias Schmelzle
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Patrick Chène
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
31
|
Clinicopathological and prognostic significance of Yes-associated protein expression in hepatocellular carcinoma and hepatic cholangiocarcinoma. Tumour Biol 2016; 37:13499-13508. [DOI: 10.1007/s13277-016-5211-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
|
32
|
Landin-Malt A, Benhaddou A, Zider A, Flagiello D. An evolutionary, structural and functional overview of the mammalian TEAD1 and TEAD2 transcription factors. Gene 2016; 591:292-303. [PMID: 27421669 DOI: 10.1016/j.gene.2016.07.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 01/22/2023]
Abstract
TEAD proteins constitute a family of highly conserved transcription factors, characterized by a DNA-binding domain called the TEA domain and a protein-binding domain that permits association with transcriptional co-activators. TEAD proteins are unable to induce transcription on their own. They have to interact with transcriptional cofactors to do so. Once TEADs bind their co-activators, the different complexes formed are known to regulate the expression of genes that are crucial for embryonic development, important for organ formation (heart, muscles), and involved in cell death and proliferation. In the first part of this review we describe what is known of the structure of TEAD proteins. We then focus on two members of the family: TEAD1 and TEAD2. First the different transcriptional cofactors are described. These proteins can be classified in three categories: i), cofactors regulating chromatin conformation, ii), cofactors able to bind DNA, and iii), transcriptional cofactors without DNA binding domain. Finally we discuss the recent findings that identified TEAD1 and 2 and its coactivators involved in cancer progression.
Collapse
Affiliation(s)
- André Landin-Malt
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22908, USA.
| | - Ataaillah Benhaddou
- Univ Paris Diderot, Sorbonne Paris Cité, Team Regulation of Cell-Fate Specification in the Mouse, IJM, UMR 7592 CNRS, Paris, France.
| | - Alain Zider
- Univ Paris Diderot, Sorbonne Paris Cité, Team Molecular Oncology and Ovarian Pathologies, IJM, UMR 7592 CNRS, Paris, France.
| | - Domenico Flagiello
- Univ Paris Diderot, Sorbonne Paris Cité, Team Regulation of Cell-Fate Specification in the Mouse, IJM, UMR 7592 CNRS, Paris, France.
| |
Collapse
|
33
|
Simon E, Faucheux C, Zider A, Thézé N, Thiébaud P. From vestigial to vestigial-like: the Drosophila gene that has taken wing. Dev Genes Evol 2016; 226:297-315. [PMID: 27116603 DOI: 10.1007/s00427-016-0546-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/10/2016] [Indexed: 12/16/2022]
Abstract
The members of the vestigial-like gene family have been identified as homologs of the Drosophila vestigial, which is essential to wing formation. All members of the family are characterized by the presence of the TONDU domain, a highly conserved sequence that mediates their interaction with the transcription factors of the TEAD family. Mammals possess four vestigial-like genes that can be subdivided into two classes, depending on the number of Tondu domains present. While vestigial proteins have been studied in great depth in Drosophila, we still have sketchy knowledge of the functions of vestigial-like proteins in vertebrates. Recent studies have unveiled unexpected functions for some of these members and reveal the role they play in the Hippo pathway. Here, we present the current knowledge about vestigial-like family gene members and their functions, together with their identification in different taxa.
Collapse
Affiliation(s)
- Emilie Simon
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France
| | - Corinne Faucheux
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France
| | - Alain Zider
- Univ Paris Diderot, Sorbonne Paris Cité, IJM, UMR 7592 CNRS, F-75205, Paris, France
| | - Nadine Thézé
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France
| | - Pierre Thiébaud
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France.
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France.
- Univ. Bordeaux, INSERM U1035, 146 rue Léo Saignat, 33076, Bordeaux CEDEX, France.
| |
Collapse
|
34
|
Lopez-Anido C, Poitelon Y, Gopinath C, Moran JJ, Ma KH, Law WD, Antonellis A, Feltri ML, Svaren J. Tead1 regulates the expression of Peripheral Myelin Protein 22 during Schwann cell development. Hum Mol Genet 2016; 25:3055-3069. [PMID: 27288457 DOI: 10.1093/hmg/ddw158] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/14/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022] Open
Abstract
Schwann cells are myelinating glia in the peripheral nervous system that form the myelin sheath. A major cause of peripheral neuropathy is a copy number variant involving the Peripheral Myelin Protein 22 (PMP22) gene, which is located within a 1.4-Mb duplication on chromosome 17 associated with the most common form of Charcot-Marie-Tooth Disease (CMT1A). Rodent models of CMT1A have been used to show that reducing Pmp22 overexpression mitigates several aspects of a CMT1A-related phenotype. Mechanistic studies of Pmp22 regulation identified enhancers regulated by the Sox10 (SRY sex determining region Y-box 10) and Egr2/Krox20 (Early growth response protein 2) transcription factors in myelinated nerves. However, relatively little is known regarding how other transcription factors induce Pmp22 expression during Schwann cell development and myelination. Here, we examined Pmp22 enhancers as a function of cell type-specificity, nerve injury and development. While Pmp22 enhancers marked by active histone modifications were lost or remodeled after injury, we found that these enhancers were permissive in early development prior to Pmp22 upregulation. Pmp22 enhancers contain binding motifs for TEA domain (Tead) transcription factors of the Hippo signaling pathway. We discovered that Tead1 and co-activators Yap and Taz are required for Pmp22 expression, as well as for the expression of Egr2 Tead1 directly binds Pmp22 and Egr2 enhancers early in development and Tead1 binding is induced during myelination, correlating with Pmp22 expression. The data identify Tead1 as a novel regulator of Pmp22 expression during development in concert with Sox10 and Egr2.
Collapse
Affiliation(s)
- Camila Lopez-Anido
- Waisman Center, Madison, WI, USA.,Comparative Biomedical Sciences Graduate Program, Madison, WI, USA
| | | | - Chetna Gopinath
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Ki Hwan Ma
- Waisman Center, Madison, WI, USA.,Cellular and Molecular Pathology Graduate Program, Madison, WI, USA
| | - William D Law
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anthony Antonellis
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - M Laura Feltri
- Hunter James Kelly Research Institute, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - John Svaren
- Waisman Center, Madison, WI, USA .,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
35
|
YAP/TAZ as therapeutic targets in cancer. Curr Opin Pharmacol 2016; 29:26-33. [PMID: 27262779 DOI: 10.1016/j.coph.2016.05.002] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/14/2022]
Abstract
The biology and regulation of YAP and TAZ, two closely related transcriptional regulators, are receiving increasing attention owing to their fundamental roles in organ growth, tissue repair and cancer. In particular, the widespread activation of YAP/TAZ in carcinomas, and the crucial role of YAP/TAZ activation for many 'hallmarks' of cancer are indicating YAP/TAZ as prime targets for designing anti-cancer drugs. Here, we start from the known modalities to regulate YAP/TAZ to highlight possible routes of therapeutic intervention.
Collapse
|
36
|
Lopez-Anido C, Sun G, Koenning M, Srinivasan R, Hung HA, Emery B, Keles S, Svaren J. Differential Sox10 genomic occupancy in myelinating glia. Glia 2015; 63:1897-1914. [PMID: 25974668 PMCID: PMC4644515 DOI: 10.1002/glia.22855] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/22/2015] [Indexed: 11/11/2022]
Abstract
Myelin is formed by specialized myelinating glia: oligodendrocytes and Schwann cells in the central and peripheral nervous systems, respectively. While there are distinct developmental aspects and regulatory pathways in these two cell types, myelination in both systems requires the transcriptional activator Sox10. Sox10 interacts with cell type-specific transcription factors at some loci to induce myelin gene expression, but it is largely unknown how Sox10 transcriptional networks globally compare between oligodendrocytes and Schwann cells. We used in vivo ChIP-Seq analysis of spinal cord and peripheral nerve (sciatic nerve) to identify unique and shared Sox10 binding sites and assess their correlation with active enhancers and transcriptional profiles in oligodendrocytes and Schwann cells. Sox10 binding sites overlap with active enhancers and critical cell type-specific regulators of myelination, such as Olig2 and Myrf in oligodendrocytes, and Egr2/Krox20 in Schwann cells. Sox10 sites also associate with genes critical for myelination in both oligodendrocytes and Schwann cells and are found within super-enhancers previously defined in brain. In Schwann cells, Sox10 sites contain binding motifs of putative partners in the Sp/Klf, Tead, and nuclear receptor protein families. Specifically, siRNA analysis of nuclear receptors Nr2f1 and Nr2f2 revealed downregulation of myelin genes Mbp and Ndrg1 in primary Schwann cells. Our analysis highlights different mechanisms that establish cell type-specific genomic occupancy of Sox10, which reflects the unique characteristics of oligodendrocyte and Schwann cell differentiation. GLIA 2015;63:1897-1914.
Collapse
Affiliation(s)
- Camila Lopez-Anido
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Guannan Sun
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Matthias Koenning
- Department of Anatomy and Neuroscience and the Centre for Neuroscience Research, University of Melbourne, Melbourne, Australia
| | - Rajini Srinivasan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Holly A. Hung
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ben Emery
- Department of Anatomy and Neuroscience and the Centre for Neuroscience Research, University of Melbourne, Melbourne, Australia
| | - Sunduz Keles
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
37
|
Cravo AS, Carter E, Erkan M, Harvey E, Furutani-Seiki M, Mrsny R. Hippo pathway elements Co-localize with Occludin: A possible sensor system in pancreatic epithelial cells. Tissue Barriers 2015; 3:e1037948. [PMID: 26451343 PMCID: PMC4574897 DOI: 10.1080/21688370.2015.1037948] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/22/2015] [Accepted: 03/27/2015] [Indexed: 12/14/2022] Open
Abstract
External adherens junction-based cell-cell contacts involving E-cadherin interactions function to sense planar cell status and modulate epithelial cell proliferation through Hippo (Hpo) and non-canonical Wnt pathways signaling. We hypothesized these regulatory processes should also be sensitive to a similar cell-cell contact sensor associated with apical-basal polarity events at epithelial surfaces. We used 2 human pancreatic cancer cell lines to explore this hypothesis: one with the capacity to form functional tight junction structures and polarize (HPAFII) and one lacking this capacity (AsPc1). Occludin (Ocln), a tetraspanning protein associated with TJ structures and capable of establishing external cell-cell contacts, was observed to partially co-localize with Hpo elements YAP (c-yes associated protein) and TEAD (TEA-dependent), which function to drive a proliferative transcription program. Treatment with dobutamine, known to affect YAP, was shown to suppress proliferation in an Ocln-dependent manner. Blockade of protein kinase C-zeta (PKC-ζ) diminished transepithelial electrical resistance (TER) of HPAFII monolayers that was not corrected by dobutamine treatment while the loss of TER resulting from inhibition of ROCK1 could be partially recovered. Examination of normal and cancerous human pancreatic biopsies showed that the cellular localization of Ocln, c-Yes, YAP, and TEAD were similar to HPAFII for normal cells and AsPc1 for cancerous cells. Together, these results suggest a link between Hpo and signals emanating from cell-cell contacts involving Ocln that may regulate pancreatic cell proliferation through the coordination of planar cell polarity with apical-basal polarity events.
Collapse
Affiliation(s)
- Ana Santos Cravo
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| | - Edward Carter
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| | - Mert Erkan
- Department of Surgery; Koc University School of Medicine ; Istanbul, Turkey
| | - Emma Harvey
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| | | | - Randall Mrsny
- Department of Pharmacy and Pharmacology; University of Bath ; Bath, UK
| |
Collapse
|
38
|
Hung HA, Sun G, Keles S, Svaren J. Dynamic regulation of Schwann cell enhancers after peripheral nerve injury. J Biol Chem 2015; 290:6937-50. [PMID: 25614629 PMCID: PMC4358118 DOI: 10.1074/jbc.m114.622878] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/31/2014] [Indexed: 12/20/2022] Open
Abstract
Myelination of the peripheral nervous system is required for axonal function and long term stability. After peripheral nerve injury, Schwann cells transition from axon myelination to a demyelinated state that supports neuronal survival and ultimately remyelination of axons. Reprogramming of gene expression patterns during development and injury responses is shaped by the actions of distal regulatory elements that integrate the actions of multiple transcription factors. We used ChIP-seq to measure changes in histone H3K27 acetylation, a mark of active enhancers, to identify enhancers in myelinating rat peripheral nerve and their dynamics after demyelinating nerve injury. Analysis of injury-induced enhancers identified enriched motifs for c-Jun, a transcription factor required for Schwann cells to support nerve regeneration. We identify a c-Jun-bound enhancer in the gene for Runx2, a transcription factor induced after nerve injury, and we show that Runx2 is required for activation of other induced genes. In contrast, enhancers that lose H3K27ac after nerve injury are enriched for binding sites of the Sox10 and early growth response 2 (Egr2/Krox20) transcription factors, which are critical determinants of Schwann cell differentiation. Egr2 expression is lost after nerve injury, and many Egr2-binding sites lose H3K27ac after nerve injury. However, the majority of Egr2-bound enhancers retain H3K27ac, indicating that other transcription factors maintain active enhancer status after nerve injury. The global epigenomic changes in H3K27ac deposition pinpoint dynamic changes in enhancers that mediate the effects of transcription factors that control Schwann cell myelination and peripheral nervous system responses to nerve injury.
Collapse
Affiliation(s)
- Holly A Hung
- From the Waisman Center, Cellular and Molecular Pathology Graduate Program, and
| | - Guannan Sun
- Departments of Biostatistics and Medical Informatics and
| | - Sunduz Keles
- Departments of Biostatistics and Medical Informatics and
| | - John Svaren
- From the Waisman Center, Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
39
|
Abstract
The arrival of multicellularity in evolution facilitated cell-cell signaling in conjunction with adhesion. As the ectodomains of cadherins interact with each other directly in trans (as well as in cis), spanning the plasma membrane and associating with multiple other entities, cadherins enable the transduction of "outside-in" or "inside-out" signals. We focus this review on signals that originate from the larger family of cadherins that are inwardly directed to the nucleus, and thus have roles in gene control or nuclear structure-function. The nature of cadherin complexes varies considerably depending on the type of cadherin and its context, and we will address some of these variables for classical cadherins versus other family members. Substantial but still fragmentary progress has been made in understanding the signaling mediators used by varied cadherin complexes to coordinate the state of cell-cell adhesion with gene expression. Evidence that cadherin intracellular binding partners also localize to the nucleus is a major point of interest. In some models, catenins show reduced binding to cadherin cytoplasmic tails favoring their engagement in gene control. When bound, cadherins may serve as stoichiometric competitors of nuclear signals. Cadherins also directly or indirectly affect numerous signaling pathways (e.g., Wnt, receptor tyrosine kinase, Hippo, NFκB, and JAK/STAT), enabling cell-cell contacts to touch upon multiple biological outcomes in embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center; Program in Genes & Development, Graduate School in Biomedical Sciences, Houston, Texas, USA.
| | - Meghan T Maher
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cara J Gottardi
- Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
40
|
Guo C, Wang X, Liang L. LATS2-mediated YAP1 phosphorylation is involved in HCC tumorigenesis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1690-1697. [PMID: 25973055 PMCID: PMC4396227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/28/2015] [Indexed: 06/04/2023]
Abstract
YAP (yes-associated protein) is a transcriptional co-activator that acts downstream of the Hippo signaling pathway and regulates multiple cellular processes, including proliferation and apoptosis. Although YAP plays an important role in various tumors, the underlying mechanism in hepatocellular carcinoma (HCC) tumorigenesis remains unclear. In this study, we observed that the LATS2 was highly expressed in Bel-7402 and HepG2 cell lines, and LATS2 protein level was negatively correlated with YAP1 in HCC cells. And then, we inhibited LATS2 expression by transfecting with siRNA. Western blot and Immunofluorescent staining analysis demonstrated that LATS2 inhibition decreased the dephosphorylation of YAP1 protein and promoted YAP1 nuclear accumulation in HCC cells. Moreover, Immunoprecipitation assay results also indicated that Yap binds directly to TEAD2 and LATS2 inhibition-mediated dephosphorylation increased the YAP1/TEAD2 association, leading to YAP1/TEAD2 transcriptional activation, which in turn upregulated cell invasion in HCC cells. Taken together, our current data indicated a new regulatory mechanism of YAP1 by the LATS2-mediated phosphorylation that was involved in HCC tumorigenesis.
Collapse
Affiliation(s)
- Chao Guo
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital Changsha 410005, P.R. China
| | - Xintian Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital Changsha 410005, P.R. China
| | - Lufeng Liang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital Changsha 410005, P.R. China
| |
Collapse
|
41
|
Li H, Wang S, Wang G, Zhang Z, Wu X, Zhang T, Fu B, Chen G. Yes-associated protein expression is a predictive marker for recurrence of hepatocellular carcinoma after liver transplantation. Dig Surg 2015; 31:468-478. [PMID: 25632982 DOI: 10.1159/000370252] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/30/2014] [Indexed: 12/10/2022]
Abstract
PURPOSE To explore the expression of Yes-associated protein (YAP) in hepatocellular carcinoma (HCC) patients, and assess its prognostic value to recurrence of HCC after liver transplantation (LT). METHODS Collected data of 105 consecutive patients undergoing LT for HCC were analyzed retrospectively. The immunohistochemistry was used to detect the expression of YAP, Mst1, Lats1/2, pYAP, pLats1/2 and pMst1/2 in tumor tissues. Contingency table and χ(2)-test were used to investigate the correlation between expression of YAP, Mst1, Lats1/2 and clinical characteristics. Univariate survival analysis and Multivariate Cox regression analysis were also performed to analyze the correlation of clinical and pathological factors with tumor recurrence after LT. The Kaplan-Meier method and log-rank test were used to analyze HCC-specific disease-free survival (DFS) rate. RESULTS Forty patients fulfilled Milan criteria with 1-, 2-, 3- and 5-years DFS of 86.7, 84.6, 84, 84%, respectively. The positive rates of YAP, Lats1/2, Mst1 in HCC were 51.4, 45.7, 64.8%, respectively. YAP expression in HCC tumors was significantly associated with tumor size (p = 0.041), venous infiltration (p = 0.002), AJCC tumor stage (p = 0.027). Lats1/2 expression was significantly associated with tumor size (p = 0.001) and AJCC tumor stage (p = 0.019). Mst1 expression was also significantly associated with tumor size (p = 0.042). HCC-specific DFS was significantly longer for patients with YAP negative expression compared with patients with YAP positive expression (1-, 2-, 3- and 5-years DFS of 71.7, 65.3, 65.3, 65.3 vs. 42.5, 36.6, 32.5, 30.4%, respectively, log-rank = 12.89, p < 0.001). Multivariate Cox regression analysis indicated that YAP expression (HR = 2.011, p = 0.020) in HCC was an independent prognostic factor for HCC-specific DFS after liver transplantation. CONCLUSIONS YAP is an independent prognostic marker for tumor recurrence for HCC patients after liver transplantation.
Collapse
Affiliation(s)
- Hua Li
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou,Guangdong, PR China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
YAP expression in normal and neoplastic breast tissue: an immunohistochemical study. Arch Med Res 2014; 45:223-8. [PMID: 24606817 DOI: 10.1016/j.arcmed.2014.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 01/29/2014] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Yes-associated protein (YAP) is a transcriptional factor involved in normal cell proliferation, apoptosis and carcinogenesis; however, its contribution to breast cancer (BC) is still controversial. We undertook this study to compare the expression of YAP by immunohistochemistry (IHC) in normal breast tissue of women without breast cancer (BC) (controls), non-neoplastic breast tissue in women with cancer (internal controls) and in four different subtypes of invasive ductal carcinoma. METHODS There were 17 controls and 105 tumor cases (53 luminal A, 15 luminal B, 20 overexpression of HER2 and 17 triple negative cases) studied by IHC. Statistical analysis included χ(2) for linear trend (Extended Mantel-Haenszel). RESULTS There were 40% of internal controls that showed expression of YAP in myoepithelial cells, whereas in controls expression was 100%. In controls, 3/17 (17.6%) showed cytoplasmic staining in luminal cells. There was a significant difference in nuclear expression between the ductal BC subtypes. Luminal A had 4% of positive cases with <10% of cells affected in each case; in contrast, there were 17-20% of positive cases in the other groups with 50% or more of stained cells. YAP expression in stromal cells was not observed in controls or in triple-negative cases, and luminal B pattern had the highest YAP nuclear expression (20%). CONCLUSIONS YAP showed decreased expression in tumor cells compared with normal breast tissue. These findings are consistent with a role of YAP as a suppressor gene in BC and show differences in YAP expression in different patterns of ductal BC.
Collapse
|
43
|
Ma J, Zhang L, Tipton AR, Wu J, Messmer-Blust AF, Philbrick MJ, Qi Y, Liu ST, Liu H, Li J, Guo S. Structural and functional analysis of the related transcriptional enhancer factor-1 and NF-κB interaction. Am J Physiol Heart Circ Physiol 2013; 306:H233-42. [PMID: 24213609 DOI: 10.1152/ajpheart.00069.2013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The related transcriptional enhancer factor-1 (RTEF-1) increases gene transcription of hypoxia-inducible factor 1α (HIF-1α) and enhances angiogenesis in endothelium. Both hypoxia and inflammatory factor TNF-α regulate gene expression of HIF-1α, but how RTEF-1 and TNF-α coordinately regulate HIF-1α gene transcription is unclear. Here, we found that RTEF-1 interacts with p65 subunit of NF-κB, a primary mediator of TNF-α. RTEF-1 increased HIF-1α promoter activity, whereas expression of p65 subunit inhibited the stimulatory effect. By contrast, knockdown of p65 markedly enhanced RTEF-1 stimulation on the HIF-1α promoter activity (7-fold). A physical interaction between RTEF-1 and p65 was confirmed by coimmunoprecipitation experiments in cells and glutathione S-transferase (GST)-pull-down assays. A computational analysis of RTEF-1 crystal structures revealed that a conserved surface of RTEF-1 potentially interacts with p65 via four amino acid residues located at T347, Y349, R351, and Y352. We performed site-directed mutagenesis and GST-pull-down assays and demonstrated that Tyr352 (Y352) in RTEF-1 is a key site for the formation of RTEF-1 and p65-NF-κB complex. An alanine mutation at Y352 of RTEF-1 disrupted the interaction of RTEF-1 with p65. Moreover, expression of RTEF-1 decreased TNF-α-induced HIF-1α promoter activity, IL-1β, and IL-6 mRNA levels in cells; however, the effect of RTEF-1 was largely lost when Y352 was mutated to alanine. These results indicate that RTEF-1 interacts with p65-NF-κB through Y352 and that they antagonize each other for HIF-1α transcriptional activation, suggesting a novel mechanism by which RTEF-1 regulates gene expression, linking hypoxia to inflammation.
Collapse
Affiliation(s)
- Jieliang Ma
- College of Life Science, Liaoning University, Shenyang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li N, Lim G, Chen L, McCabe MF, Kim H, Zhang S, Mao J. Spinal expression of Hippo signaling components YAP and TAZ following peripheral nerve injury in rats. Brain Res 2013; 1535:137-47. [PMID: 23998984 DOI: 10.1016/j.brainres.2013.08.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/21/2013] [Accepted: 08/25/2013] [Indexed: 01/23/2023]
Abstract
Previous studies have shown that the morphology and number of cells in the spinal cord dorsal horn could change following peripheral nerve injury and that the Hippo signaling pathway plays an important role in cell growth, proliferation, apoptosis, and dendritic remolding. In the present study, we examined whether the expression of YAP and TAZ, two critical components regulated by Hippo signaling, in the spinal cord dorsal horn would be altered by chronic constriction sciatic nerve injury (CCI). We found that (1) YAP was mainly expressed on CGRP- and IB4-immunoreactive primary afferent nerve terminals without noticeable expression on glial cells, whereas TAZ was mainly expressed on spinal cord second order neurons as well as microglia; (2) upregulation of YAP and TAZ expression followed two distinct temporal patterns after CCI, such that the highest expression of YAP and TAZ was on day 14 and day 1 after CCI, respectively; (3) there were also unique topographic patterns of YAP and TAZ distribution in the spinal cord dorsal horn consistent with their distinctive association with primary afferents and second order neurons; (4) changes in the YAP expression were selectively induced by CCI but not CFA-induced hindpaw inflammation; and (5) the number of nuclear profiles of TAZ expression was significantly increased after CCI, indicating translocation of TAZ from the cytoplasma to nucleus. These findings indicate that peripheral nerve injury induced time-dependent and region-specific changes in the spinal YAP and TAZ expression. A role for Hippo signaling in synaptic and structural plasticity is discussed in relation to the cellular mechanism of neuropathic pain.
Collapse
Affiliation(s)
- Na Li
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Anesthesiology, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan, China.
| | | | | | | | | | | | | |
Collapse
|
45
|
Jie L, Fan W, Weiqi D, Yingqun Z, Ling X, Miao S, Ping C, Chuanyong G. The hippo-yes association protein pathway in liver cancer. Gastroenterol Res Pract 2013; 2013:187070. [PMID: 23986776 PMCID: PMC3748736 DOI: 10.1155/2013/187070] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 06/18/2013] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide and the third leading cause of cancer mortality. Despite continuing development of new therapies, prognosis for patients with HCC remains extremely poor. In recent years, control of organ size becomes a hot topic in HCC development. The Hippo signaling pathway has been delineated and shown to be critical in controlling organ size in both Drosophila and mammals. The Hippo kinase cascade, a singling pathway that antagonizes the transcriptional coactivator Yes-associated protein (YAP), plays an important role in animal organ size control by regulating cell proliferation and apoptosis rates. During HCC development, this pathway is likely inactivated in tumor initiated cells that escape suppressive constrain exerted by the surrounding normal tissue, thus allowing clonal expansion and tumor development. We have reviewed evolutionary changes in YAP as well as other components of the Hippo pathway and described the relationships between YAP genes and HCC. We also discuss regulation of transcription factors that are up- and downstream of YAP in liver cancer development.
Collapse
Affiliation(s)
- Lu Jie
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Wang Fan
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Dai Weiqi
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Zhou Yingqun
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Xu Ling
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Shen Miao
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Cheng Ping
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| | - Guo Chuanyong
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, School of Medicine, No. 301, Yanchang Road, Shanghai 200072, China
| |
Collapse
|
46
|
Hau JC, Erdmann D, Mesrouze Y, Furet P, Fontana P, Zimmermann C, Schmelzle T, Hofmann F, Chène P. The TEAD4-YAP/TAZ Protein-Protein Interaction: Expected Similarities and Unexpected Differences. Chembiochem 2013; 14:1218-25. [DOI: 10.1002/cbic.201300163] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Indexed: 12/20/2022]
|
47
|
The Hippo pathway: key interaction and catalytic domains in organ growth control, stem cell self-renewal and tissue regeneration. Essays Biochem 2012; 53:111-27. [PMID: 22928512 DOI: 10.1042/bse0530111] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The Hippo pathway is a conserved pathway that interconnects with several other pathways to regulate organ growth, tissue homoeostasis and regeneration, and stem cell self-renewal. This pathway is unique in its capacity to orchestrate multiple processes, from sensing to execution, necessary for organ expansion. Activation of the Hippo pathway core kinase cassette leads to cytoplasmic sequestration of the nuclear effectors YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif), consequently disabling their transcriptional co-activation function. Components upstream of the core kinase cassette have not been well understood, especially in vertebrates, but are gradually being elucidated and include cell polarity and cell adhesion proteins.
Collapse
|
48
|
Xie C, Guo Y, Zhu T, Zhang J, Ma PX, Chen YE. Yap1 protein regulates vascular smooth muscle cell phenotypic switch by interaction with myocardin. J Biol Chem 2012; 287:14598-605. [PMID: 22411986 DOI: 10.1074/jbc.m111.329268] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The Hippo-Yap (Yes-associated protein) signaling pathway has emerged as one of the critical pathways regulating cell proliferation, differentiation, and apoptosis in response to environmental and developmental cues. However, Yap1 roles in vascular smooth muscle cell (VSMC) biology have not been investigated. VSMCs undergo phenotypic switch, a process characterized by decreased gene expression of VSMC contractile markers and increased proliferation, migration, and matrix synthesis. The goals of the present studies were to investigate the relationship between Yap1 and VSMC phenotypic switch and to determine the molecular mechanisms by which Yap1 affects this essential process in VSMC biology. Results demonstrated that the expression of Yap1 was rapidly up-regulated by stimulation with PDGF-BB (a known inducer of phenotypic switch in VSMCs) and in the injured vessel wall. Knockdown of Yap1 impaired VSMC proliferation in vitro and enhanced the expression of VSMC contractile genes as well by increasing serum response factor binding to CArG-containing regions of VSMC-specific contractile genes within intact chromatin. Conversely, the interaction between serum response factor and its co-activator myocardin was reduced by overexpression of Yap1 in a dose-dependent manner. Taken together, these results indicate that down-regulation of Yap1 promotes VSMC contractile phenotype by both up-regulating myocardin expression and promoting the association of the serum response factor-myocardin complex with VSMC contractile gene promoters and suggest that the Yap1 signaling pathway is a central regulator of phenotypic switch of VSMCs.
Collapse
Affiliation(s)
- Changqing Xie
- Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | |
Collapse
|