151
|
Hsu PC, Yang CT, Jablons DM, You L. The Crosstalk between Src and Hippo/YAP Signaling Pathways in Non-Small Cell Lung Cancer (NSCLC). Cancers (Basel) 2020; 12:1361. [PMID: 32466572 PMCID: PMC7352956 DOI: 10.3390/cancers12061361] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/19/2020] [Accepted: 05/23/2020] [Indexed: 12/22/2022] Open
Abstract
The advancement of new therapies, including targeted therapies and immunotherapies, has improved the survival of non-small-cell lung cancer (NSCLC) patients in the last decade. Some NSCLC patients still do not benefit from therapies or encounter progressive disease during the course of treatment because they have intrinsic resistance, acquired resistance, or lack a targetable driver mutation. More investigations on the molecular biology of NSCLC are needed to find useful biomarkers for current therapies and to develop novel therapeutic strategies. Src is a non-receptor tyrosine kinase protein that interacts with cell surface growth factor receptors and the intracellular signaling pathway to maintain cell survival tumorigenesis in NSCLC. The Yes-associated protein (YAP) is one of the main effectors of the Hippo pathway and has been identified as a promoter of drug resistance, cancer progression, and metastasis in NSCLC. Here, we review studies that have investigated the activation of YAP as mediated by Src kinases and demonstrate that Src regulates YAP through three main mechanisms: (1) direct phosphorylation; (2) the activation of pathways repressing Hippo kinases; and (3) Hippo-independent mechanisms. Further work should focus on the efficacy of Src inhibitors in inhibiting YAP activity in NSCLC. In addition, future efforts toward developing potentially reasonable combinations of therapy targeting the Src-YAP axis using other therapies, including targeted therapies and/or immunotherapies, are warranted.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (P.-C.H.); (D.M.J.)
- Division of Thoracic Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan;
| | - Cheng-Ta Yang
- Division of Thoracic Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan;
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - David M. Jablons
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (P.-C.H.); (D.M.J.)
| | - Liang You
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (P.-C.H.); (D.M.J.)
| |
Collapse
|
152
|
Zhao J, Li L, Yang T. MiR-216a-3p suppresses the proliferation and invasion of cervical cancer through downregulation of ACTL6A-mediated YAP signaling. J Cell Physiol 2020; 235:9718-9728. [PMID: 32401366 DOI: 10.1002/jcp.29783] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
The tumor-suppressive role of microRNA-216a-3p (miR-216a-3p) has been evidenced in multiple tumors. Yet, the relevance of miR-216a-3p in cervical cancer remains undermined. The current study was designed to determine the expression and potential function of miR-216a-3p in cervical cancer. Expression of miR-216a-3p was markedly decreased in cervical cancer and functional assays revealed an inhibitory effect of miR-216a-3p on the proliferation, colony formation, and invasion of cervical cancer. Actin-like 6A (ACTL6A) was identified as a target gene of miR-216a-3p. Elevated ACTL6A expression was detected in cervical cancer, and ACTL6A inhibition exhibited a tumor-suppressive effect. ACTL6A inhibition increased yes-associated protein (YAP) phosphorylation and downregulated YAP-mediated transcriptional activity. ACTL6A restoration or YAP reactivation partially abrogated the miR-216a-3p-mediated antitumor effect in cervical cancer cells. Taken together, these data demonstrate that miR-216a-3p acts as a potential tumor-suppressive miRNA in cervical cancer, which exerts its function through inhibition of YAP signaling via targeting ACTL6A.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Long Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ting Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
153
|
Li X, Li Y, Han Y, Dong B, Liu D, Che L, Liu Y, Wang Y. miR-205 Promotes Apoptosis of Cervical Cancer Cells and Enhances Drug Sensitivity of Cisplatin by Inhibiting YAP1. Cancer Biother Radiopharm 2020; 35:338-344. [PMID: 32379984 DOI: 10.1089/cbr.2019.2983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Elevated expression of Yes-associated protein (YAP1) involves in the pathogenesis of cervical cancer. Bioinformatics analysis showed a targeting relationship between miR-205 and the 3'-UTR of YAP1. In this study, we aim to explore the role of miR-205 in the proliferation, apoptosis, or cisplatin (CDDP) resistance of cervical cancer cells. Patients and Methods: The dual luciferase reporter gene assay verified the relationship between miR-205 and YAP1. The CDDP-resistant cell line Hela/CDDP cells were cultured in vitro and divided into miR-NC group, miR-205 mimic group, and miR-205 inhibitor group followed by analysis of the expression of miR-205 and YAP1 mRNA by quantitative real-time polymerase chain reaction (qRT-PCR), and YAP1 protein level by western blot. Results: There was a targeted relationship between miR-205 and YAP1 mRNA. Compared with cervical cell line HCerEpiC cells, miR-205 expression was significantly decreased and YAP1 mRNA and protein expression was significantly increased in Hela cells (p < 0.01). Compared with miR-NC group, YAP1 protein expression in HeLa/CDDP cells was significantly decreased, cell apoptosis was increased, and proliferation was inhibited in miR-205 mimic-transfected Hela/CDDP cells (p < 0.01). Opposite results were obtained in miR-205 inhibitor-transfected Hela/CDDP cells. Conclusions: The expression of miR-205 is related to the CDDP resistance of cervical cancer cells. Increasing the expression of miR-205 can downregulate the expression of YAP1, inhibit the proliferation and promote apoptosis of cervical cancer cells, and enhance the sensitivity to CDDP.
Collapse
Affiliation(s)
- Xingmei Li
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yuewen Li
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yuning Han
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Bing Dong
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Dan Liu
- Department of Genetics, Qiqihar Medical University, Qiqihar, China
| | - Liqun Che
- Department of Endocrinology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yu Liu
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yuchun Wang
- Department of Pharmacology, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
154
|
Lingling Z, Jiewei L, Li W, Danli Y, Jie Z, Wen L, Dan P, Lei P, Qinghua Z. Molecular regulatory network of PD-1/PD-L1 in non-small cell lung cancer. Pathol Res Pract 2020; 216:152852. [DOI: 10.1016/j.prp.2020.152852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/03/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022]
|
155
|
Pobbati AV, Hong W. A combat with the YAP/TAZ-TEAD oncoproteins for cancer therapy. Theranostics 2020; 10:3622-3635. [PMID: 32206112 PMCID: PMC7069086 DOI: 10.7150/thno.40889] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022] Open
Abstract
The transcriptional co-regulators YAP and TAZ pair primarily with the TEAD family of transcription factors to elicit a gene expression signature that plays a prominent role in cancer development, progression and metastasis. YAP and TAZ endow cells with various oncogenic traits such that they sustain proliferation, inhibit apoptosis, maintain stemness, respond to mechanical stimuli, engineer metabolism, promote angiogenesis, suppress immune response and develop resistance to therapies. Therefore, inhibiting YAP/TAZ- TEAD is an attractive and viable option for novel cancer therapy. It is exciting to know that many drugs already in the clinic restrict YAP/TAZ activities and several novel YAP/TAZ inhibitors are currently under development. We have classified YAP/TAZ-inhibiting drugs into three groups. Group I drugs act on the upstream regulators that are stimulators of YAP/TAZ activities. Many of the Group I drugs have the potential to be repurposed as YAP/TAZ indirect inhibitors to treat various solid cancers. Group II modalities act directly on YAP/TAZ or TEADs and disrupt their interaction; targeting TEADs has emerged as a novel option to inhibit YAP/TAZ, as TEADs are major mediators of their oncogenic programs. TEADs can also be leveraged on using small molecules to activate YAP/TAZ-dependent gene expression for use in regenerative medicine. Group III drugs focus on targeting one of the oncogenic downstream YAP/TAZ transcriptional target genes. With the right strategy and impetus, it is not far-fetched to expect a repurposed group I drug or a novel group II drug to combat YAP and TAZ in cancers in the near future.
Collapse
|
156
|
PABPC1-induced stabilization of BDNF-AS inhibits malignant progression of glioblastoma cells through STAU1-mediated decay. Cell Death Dis 2020; 11:81. [PMID: 32015336 PMCID: PMC6997171 DOI: 10.1038/s41419-020-2267-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/29/2022]
Abstract
Glioblastoma is the most common and malignant form of primary central nervous tumor in adults. Long noncoding RNAs (lncRNAs) have been reported to play a pivotal role in modulating gene expression and regulating human tumor’s malignant behaviors. In this study, we confirmed that lncRNA brain-derived neurotrophic factor antisense (BDNF-AS) was downregulated in glioblastoma tissues and cells, interacted and stabilized by polyadenylate-binding protein cytoplasmic 1 (PABPC1). Overexpression of BDNF-AS inhibited the proliferation, migration, and invasion, as well as induced the apoptosis of glioblastoma cells. In the in vivo study, PABPC1 overexpression combined with BDNF-AS overexpression produced the smallest tumor and the longest survival. Moreover, BDNF-AS could elicit retina and anterior neural fold homeobox 2 (RAX2) mRNA decay through STAU1-mediated decay (SMD), and thereby regulated the malignant behaviors glioblastoma cells. Knockdown of RAX2 produced tumor-suppressive function in glioblastoma cells and increased the expression of discs large homolog 5 (DLG5), leading to the activation of the Hippo pathway. In general, this study elucidated that the PABPC1-BDNF-AS-RAX2-DLG5 mechanism may contribute to the anticancer potential of glioma cells and may provide potential therapeutic targets for human glioma.
Collapse
|
157
|
Wang D, He J, Dong J, Meyer TF, Xu T. The HIPPO pathway in gynecological malignancies. Am J Cancer Res 2020; 10:610-629. [PMID: 32195031 PMCID: PMC7061741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 01/27/2020] [Indexed: 06/10/2023] Open
Abstract
The Hippo pathway has been initially discovered by screening genes that regulate organ size in Drosophila. Recent studies have highlighted the role of the Hippo pathway in controlling organ size, tissue homeostasis and regeneration, and signaling dysregulation, especially the overactivation of the transcriptional coactivator YAP/TAZ, which leads to uncontrolled cell growth and malignant transformation. The core components of the Hippo pathway may initiate tumorigenesis by inducing tumor stem cells and proliferation, ultimately leading to metastasis and drug resistance, which occurs extensively in gynecological malignancies, including cervical cancer, ovarian cancer, and endometrial cancer. In this review, we attempt to systematically summarize recent progress in our understanding of the mechanism of Hippo pathway regulation in tumorigenesis and the mechanisms that underlie alterations during gynecological malignancies, as well as new therapeutic strategies.
Collapse
Affiliation(s)
- Dongying Wang
- Department of Obstetrics and Gynecology, Second Hospital of Jilin UniversityChangchun, Jilin, P. R. China
| | - Jiaxing He
- Department of Obstetrics and Gynecology, Second Hospital of Jilin UniversityChangchun, Jilin, P. R. China
| | - Junxue Dong
- Department of Obstetrics and Gynecology, Second Hospital of Jilin UniversityChangchun, Jilin, P. R. China
- Department of Molecular Biology, Max Planck Institute for Infection BiologyBerlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection BiologyBerlin, Germany
| | - Tianmin Xu
- Department of Obstetrics and Gynecology, Second Hospital of Jilin UniversityChangchun, Jilin, P. R. China
| |
Collapse
|
158
|
Impact of HPV E5 on viral life cycle via EGFR signaling. Microb Pathog 2020; 139:103923. [DOI: 10.1016/j.micpath.2019.103923] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/08/2019] [Accepted: 12/10/2019] [Indexed: 12/28/2022]
|
159
|
Liu X, Zhou Y, Ning YE, Gu H, Tong Y, Wang N. MiR-195-5p Inhibits Malignant Progression of Cervical Cancer by Targeting YAP1. Onco Targets Ther 2020; 13:931-944. [PMID: 32099397 PMCID: PMC6996614 DOI: 10.2147/ott.s227826] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/20/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose Our previous studies have shown that miR-195 is reduced in cervical cancer tissues, and that upregulation of miR-195 suppressed cervical cancer cell growth and induced a cell cycle block. In this study, we aimed to further elucidate the mechanism of action between miR-195-5p and Yes-associated protein 1 (YAP1) in the malignant progression of cervical cancer. Methods MiR-195-5p and YAP1 were detected using qRT-PCR in cervical cancer cells transfected with miR-195-5p mimics or inhibitor. Cell proliferation, migration, and invasion ability were detected using MTT, wound healing, and transwell invasion assays. Dual luciferase reporter assay, qRT-PCR, and Western blot analysis were used to demonstrate that YAP1 was a target of miR-195-5p. Results Our results showed that miR-195-5p is negatively correlated with YAP1 protein levels but not with mRNA expression. Moreover, upregulation of miR-195-5p by transient transfection with miR-195-5p mimics in HeLa and SiHa cells inhibited cell proliferation, migration ability, invasiveness, and the EMT. Conversely, miR-195-5p downregulation produced opposite results. In addition, multiple miRNA target prediction sites showed that YAP1 was a potential target gene; this was confirmed by dual luciferase assay. Rescue experiments further confirmed that YAP1 is involved in miR-195-5p-mediated inhibition of proliferation, migration ability, invasiveness, and the EMT of cervical cancer cells. Conclusion Taken together, our data suggest that miR-195-5p may act as a tumor suppressor which could provide a theoretical basis for cervical cancer patient targeted therapy.
Collapse
Affiliation(s)
- Xiaomin Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110000, People's Republic of China
| | - Yi Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110000, People's Republic of China
| | - Yu-E Ning
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110000, People's Republic of China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Yuxin Tong
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People's Republic of China
| | - Ning Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110000, People's Republic of China
| |
Collapse
|
160
|
Wang Z, Lu W, Zhang Y, Zou F, Jin Z, Zhao T. The Hippo Pathway and Viral Infections. Front Microbiol 2020; 10:3033. [PMID: 32038526 PMCID: PMC6990114 DOI: 10.3389/fmicb.2019.03033] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
The Hippo signaling pathway is a novel tumor suppressor pathway, initially found in Drosophila. Recent studies have discovered that the Hippo signaling pathway plays a critical role in a wide range of biological processes, including organ size control, cell proliferation, cancer development, and virus-induced diseases. In this review, we summarize the current understanding of the biological feature and pathological role of the Hippo pathway, focusing particularly on current findings in the function of the Hippo pathway in virus infection and pathogenesis.
Collapse
Affiliation(s)
- Zhilong Wang
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Wanhang Lu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Yiling Zhang
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Feng Zou
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Zhigang Jin
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Tiejun Zhao
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
161
|
Cheng Y, Lan K, Yang X, Liang D, Xia L, Cui J. Role of Cervical Cancer Radiotherapy in the Expression of EGFR and p53 Gene. CURR PROTEOMICS 2020. [DOI: 10.2174/1570164616666190204155403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Cervical cancer arises from the cervix and it is the 3rd most diagnosed malignancy and a foremost cause of cancer-related death in females. On the other hand, the expressions of EGFR and p53 are two important proteins observed in various studies on cervical cancer.Objective:The study aims to evaluate the beneficial effect of radiotherapy based on the regulation of p53 and EGFR gene in patients with cervical cancer.Methods:In this investigation, the regulation of important molecules responsible for cancer cell proliferation and DNA repair in the cervical cancer cell line was evaluated. The study comprises of an evaluation based on clinical study design from the malignant biopsies of 15 cervical cancer patients. The patterns of expression for the p53 gene and Epidermal Growth Factor Receptor (EGFR) were evaluated in DoTc2 and SiHa cervical cancer cell lines using clonogenic assay, western blotting and immunohistochemistry techniques from the malignant biopsies of the 15 patients.Results:The study observed that the regulation of p53 and EGFR was very weak after the exposure of the radiation. In addition, the expression of p53 and EGFR was observed in malevolent biopsy samples after radiation with a dosage of 1.8 Gy radiations. Additionally, the expression of p53 and EGFR was able to induce by a single dose of radiotherapy in the malignant biopsies whereas it was unable to induce in DoTc2 and SiHa cervical cancer cells.Conclusion:The study observed that radiation exposed cancer cell lines modulates the expression of p53 and EGFR gene. The study also highlights the gap between in vitro experimental models and clinical study design.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Kuntian Lan
- Department of Gynecology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Xiaoxia Yang
- Department of Gynecology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Dongxia Liang
- Department of Gynecology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Li Xia
- Department of Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Jinquan Cui
- Department of Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| |
Collapse
|
162
|
Yi X, Deng X, Zhao Y, Deng B, Deng J, Fan H, Du Y, Hao L. Ubiquitin-like protein FAT10 promotes osteosarcoma growth by modifying the ubiquitination and degradation of YAP1. Exp Cell Res 2019; 387:111804. [PMID: 31877302 DOI: 10.1016/j.yexcr.2019.111804] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/19/2019] [Accepted: 12/22/2019] [Indexed: 02/02/2023]
Abstract
Osteosarcoma is a common malignancy of the bone tissue. The rapid growth exhibited by this cancer is a primary challenge in its treatment. In many types of cancers, FAT10, a ubiquitin-like protein, is involved in several biological activities, especially cell proliferation. Herein, we demonstrate that FAT10 plays a vital role in tumorigenesis and is overexpressed in tumor tissues compared to its expression in adjacent normal tissues. Functional assays revealed that knockdown of FAT10 expression significantly repressed the proliferation of osteosarcoma in vitro and in vivo. Furthermore, our results indicate that FAT10 exhibits oncogenic functions by regulating the level of YAP1, a key protein of the Hippo/YAP signaling pathway, and a significant positive correlation exists between the levels of FAT10 and YAP1. Further analysis showed that FAT10-induced growth of osteosarcoma cells is dependent on YAP1. Mechanistically, FAT10 stabilizes YAP1 expression by regulating its ubiquitination and degradation. Taken together, our results link the two drivers of cell growth in osteosarcoma and reveal a novel pathway for FAT10 regulation. We provide new evidence for the biological and clinical significance of FAT10 as a potential biomarker for osteosarcoma.
Collapse
Affiliation(s)
- Xuan Yi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xueqiang Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanzhi Zhao
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Binbin Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianyong Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huimin Fan
- Department of Ophthalmology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunyan Du
- Department of Medical, Jiangxi Provincial People's Hospital, Nanchang, China; Department of Otorhinolaryngology, Jiangxi Provincial People's Hospital, Nanchang, China.
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
163
|
Zinatizadeh MR, Miri SR, Zarandi PK, Chalbatani GM, Rapôso C, Mirzaei HR, Akbari ME, Mahmoodzadeh H. The Hippo Tumor Suppressor Pathway (YAP/TAZ/TEAD/MST/LATS) and EGFR-RAS-RAF-MEK in cancer metastasis. Genes Dis 2019; 8:48-60. [PMID: 33569513 PMCID: PMC7859453 DOI: 10.1016/j.gendis.2019.11.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/24/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023] Open
Abstract
Hippo Tumor Suppressor Pathway is the main pathway for cell growth that regulates tissue enlargement and organ size by limiting cell growth. This pathway is activated in response to cell cycle arrest signals (cell polarity, transduction, and DNA damage) and limited by growth factors or mitogens associated with EGF and LPA. The major pathway consists of the central kinase of Ste20 MAPK (Saccharomyces cerevisiae), Hpo (Drosophila melanogaster) or MST kinases (mammalian) that activates the mammalian AGC kinase dmWts or LATS effector (MST and LATS). YAP in the nucleus work as a cofactor for a wide range of transcription factors involved in proliferation (TEA domain family, TEAD1-4), stem cells (Oct4 mononuclear factor and SMAD-related TGFβ effector), differentiation (RUNX1), and Cell cycle/apoptosis control (p53, p63, and p73 family members). This is due to the diverse roles of YAP and may limit tumor progression and establishment. TEAD also coordinates various signal transduction pathways such as Hippo, WNT, TGFβ and EGFR, and effects on lack of regulation of TEAD cancerous genes, such as KRAS, BRAF, LKB1, NF2 and MYC, which play essential roles in tumor progression, metastasis, cancer metabolism, immunity, and drug resistance. However, RAS signaling is a pivotal factor in the inactivation of Hippo, which controls EGFR-RAS-RAF-MEK-ERK-mediated interaction of Hippo signaling. Thus, the loss of the Hippo pathway may have significant consequences on the targets of RAS-RAF mutations in cancer.
Collapse
Affiliation(s)
- Mohammad Reza Zinatizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
- Corresponding author. Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Rouhollah Miri
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Peyman Kheirandish Zarandi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Ghanbar Mahmoodi Chalbatani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Catarina Rapôso
- Faculty of Pharmaceutical Sciences State University of Campinas – UNICAMP Campinas, SP, Brazil
| | - Hamid Reza Mirzaei
- Cancer Research Center, Shohadae Tajrish Hospital, Department of Radiation Oncology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Habibollah Mahmoodzadeh
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
- Corresponding author. Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
164
|
Shuai Y, Ma Z, Lu J, Feng J. LncRNA SNHG15: A new budding star in human cancers. Cell Prolif 2019; 53:e12716. [PMID: 31774607 PMCID: PMC6985667 DOI: 10.1111/cpr.12716] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/16/2019] [Accepted: 10/07/2019] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Long non-coding RNAs (lncRNAs) represent an important group of non-coding RNAs (ncRNAs) with more than 200 nucleotides in length that are transcribed from the so-called genomic "dark matter." Mounting evidence has shown that lncRNAs have manifested a paramount function in the pathophysiology of human diseases, especially in the pathogenesis and progression of cancers. Despite the exponential growth in lncRNA publications, our understanding of regulatory mechanism of lncRNAs is still limited, and a lot of controversies remain in the current lncRNA knowledge.The purpose of this article is to explore the clinical significance and molecular mechanism of SNHG15 in tumors. MATERIALS & METHODS We have systematically searched the Pubmed, Web of Science, Embase and Cochrane databases. We provide an overview of current evidence concerning the functional role, mechanistic models and clinical utilities of SNHG15 in human cancers in this review. RESULTS Small nucleolar RNA host gene 15 (SNHG15), a novel lncRNA, is identified as a key regulator in tumorigenesis and progression of various human cancers, including colorectal cancer (CRC), gastric cancer (GC), pancreatic cancer (PC) and hepatocellular carcinoma (HCC). Dysregulation of SNHG15 has been revealed to be dramatically correlated with advanced clinicopathological factors and predicts poor prognosis, suggesting its potential clinical value as a promising biomarker and therapeutic target for cancer patients. CONCLUSIONS LncRNA SNHG15 may serve as a prospective and novel biomarker for molecular diagnosis and therapeutics in patients with cancer.
Collapse
Affiliation(s)
- You Shuai
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhonghua Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Division of Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing, China.,Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianwei Lu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
165
|
Qu Y, Zhang L, Wang J, Chen P, Jia Y, Wang C, Yang W, Wen Z, Song Q, Tan B, Cheng Y. Yes-associated protein (YAP) predicts poor prognosis and regulates progression of esophageal squamous cell cancer through epithelial-mesenchymal transition. Exp Ther Med 2019; 18:2993-3001. [PMID: 31572541 PMCID: PMC6755466 DOI: 10.3892/etm.2019.7896] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/27/2019] [Indexed: 12/24/2022] Open
Abstract
The impact of yes-associated protein (YAP) on the prognosis of patients with esophageal squamous cell cancer (ESCC) and its mechanism of action has seldom been reported. In the present study, the role of YAP on the prognosis of patients with ESCC and the mechanism of action of YAP in promoting the progression of ESCC was investigated. Tumor tissue samples from patients with ESCC were collected and the level of YAP expression was detected using immunohistochemical staining. In addition, YAP was knocked-down in ESCC cell lines and the effects on cell migration and invasion were examined. The expression levels of vimentin, N-cadherin, and E-cadherin were further investigated to examine the association between YAP and epithelial-mesenchymal transition (EMT). Results showed that overexpression of YAP was associated with larger lymph node metastasis and poor disease-free survival and overall survival. Compared with patients in early stage ESCC, the association was more significant in patients with late stage ESCC. Univariate and multivariate analyses further indicated that YAP expression could be an independent prognostic factor for ESCC. Downregulation of YAP inhibited cell migration and invasion. Western blot analysis showed that when YAP was knocked down, expression levels of vimentin and N-cadherin were reduced, whereas that of E-cadherin was increased. In conclusion, the results indicates that YAP expression level could be a novel marker for predicting the prognosis of patients with ESCC, and YAP-promoted tumor migration and invasion might be through EMT in ESCC.
Collapse
Affiliation(s)
- Yan Qu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lin Zhang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jianbo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yibin Jia
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wenjing Yang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhihua Wen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qingxu Song
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bingxu Tan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
166
|
Wang C, Davis JS. At the center of cervical carcinogenesis: synergism between high-risk HPV and the hyperactivated YAP1. Mol Cell Oncol 2019; 6:e1612677. [PMID: 31528691 PMCID: PMC6736164 DOI: 10.1080/23723556.2019.1612677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human papillomavirus (HPV) is recognized as a necessary, but insufficient cause of cervical cancer. Recent in vitro and in vivo evidence from our laboratory demonstrate that hyperactivation of yes-associated protein 1 (YAP1) is sufficient to induce cervical carcinogenesis. We found that synergism between hyperactivated YAP1 and high-risk HPV is a key driver of cervical cancer initiation and progression.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, and VA Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
167
|
Li M, Lu B, Dong X, Zhou Y, He Y, Zhang T, Bao L. Enhancement of cisplatin-induced cytotoxicity against cervical cancer spheroid cells by targeting long non-coding RNAs. Pathol Res Pract 2019; 215:152653. [PMID: 31570280 DOI: 10.1016/j.prp.2019.152653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 01/15/2023]
Abstract
Cervical cancer (CVC) is one of the most common types of gynecologic malignant tumor in the world. Unfortunately, current treatments including chemo-/radiotherapy still show the limitation on CVC progress. It is well known that cancer stem cells (CSCs) plays a critical role in drug resistance and metastasis. Furthermore, dysregulation of long non-coding RNA (LncRNA) shows close association to tumorigenesis and development of multiple cancers. In this study, we investigated the cytotoxic effect of cisplatin, a clinical chemotherapeutic for cervical cancer treatment, on parental and spheroid CVC cells and surveyed the effect of LncRNA on drug-resistance. We found that spheroid CVC cells showed much more resistant to cisplatin-induced cytotoxicity compared with parental CVC cells. Furthermore, cisplatin significantly induced apoptotic cell death, while it induced cell cycle arrest in G0/G1 phase at the same dose (10 μg/ml). We also found the significant expression of EGFR in spheroid instead of parental CVC cells. Interestingly, we revealed that protruding target lncRNAs were up-regulated in cisplatin-treated spheroid CVC cells, and inhibition of these lncRNAs enhanced the cytotoxicity of cisplatin against spheroid CVC cells. These data suggest that LncRNA might act as a critical modulator on drug-resistant capability of cervical CSCs and would be a novel target for cervical cancer treatment.
Collapse
Affiliation(s)
- Meiping Li
- Pathology Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Bo Lu
- Pathology Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Xiaoqian Dong
- Laboratory Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Ying Zhou
- Laboratory Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Yao He
- Gynecology Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Tao Zhang
- Genetics Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China
| | - Lei Bao
- Pathology Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing 312000, Zhejiang Province, China.
| |
Collapse
|
168
|
Yang Z, Xiong H, Wei S, Liu Q, Gao Y, Liu L, Hu Z, Han K, Wang M, Chen P, Li Q, Zeng K. Yes-Associated Protein Promotes the Development of Condyloma Acuminatum through EGFR Pathway Activation. Dermatology 2019; 236:454-466. [PMID: 31522174 DOI: 10.1159/000500216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/02/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Investigate the role of Yes-associated protein (YAP1) in the development of condyloma acuminatum (CA). METHODS We enrolled 30 male patients with CA and 20 healthy individuals as a control group, to compare the YAP1 expression in their tissue samples. Following this, we overexpressed and downregulated YAP1 expression in HaCaT cells to examine the migratory, proliferative, and apoptotic potential of HaCaT cells expressing different levels of YAP1. RESULTS In the CA patient tissue samples, an increase in YAP1 expression can be observed. In vitro,the overexpression of YAP1 was shown to promote the growth and migration of HaCaT cells and to activate epidermal growth factor receptor (EGFR) pathway-associated proteins, while the downregulation of YAP1 inhibited cell growth and migration of these cells. CONCLUSIONS YAP1 promotes the growth of keratinocytes in CA through the activation of the EGFR pathway, and it may mediate the development of human papilloma virus-associated diseases.
Collapse
Affiliation(s)
- Zhenghui Yang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Xiong
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shanshan Wei
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingxiu Liu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Gao
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lishi Liu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhili Hu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Han
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Menglei Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pingjiao Chen
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Li
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China,
| |
Collapse
|
169
|
Young JM, Zine El Abidine A, Gómez-Martinez RA, Ozbun MA. The Known and Potential Intersections of Rab-GTPases in Human Papillomavirus Infections. Front Cell Dev Biol 2019; 7:139. [PMID: 31475144 PMCID: PMC6702953 DOI: 10.3389/fcell.2019.00139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 07/09/2019] [Indexed: 12/16/2022] Open
Abstract
Papillomaviruses (PVs) were the first viruses recognized to cause tumors and cancers in mammalian hosts by Shope, nearly a century ago (Shope and Hurst, 1933). Over 40 years ago, zur Hausen (1976) first proposed that human papillomaviruses (HPVs) played a role in cervical cancer; in 2008, he shared the Nobel Prize in Medicine for his abundant contributions demonstrating the etiology of HPVs in genital cancers. Despite effective vaccines and screening, HPV infection and morbidity remain a significant worldwide burden, with HPV infections and HPV-related cancers expected increase through 2040. Although HPVs have long-recognized roles in tumorigenesis and cancers, our understanding of the molecular mechanisms by which these viruses interact with cells and usurp cellular processes to initiate infections and produce progeny virions is limited. This is due to longstanding challenges in both obtaining well-characterized infectious virus stocks and modeling tissue-based infection and the replicative cycles in vitro. In the last 20 years, the development of methods to produce virus-like particles (VLPs) and pseudovirions (PsV) along with more physiologically relevant cell- and tissue-based models has facilitated progress in this area. However, many questions regarding HPV infection remain difficult to address experimentally and are, thus, unanswered. Although an obligatory cellular uptake receptor has yet to be identified for any PV species, Rab-GTPases contribute to HPV uptake and transport of viral genomes toward the nucleus. Here, we provide a general overview of the current HPV infection paradigm, the epithelial differentiation-dependent HPV replicative cycle, and review the specifics of how HPVs usurp Rab-related functions during infectious entry. We also suggest other potential interactions based on how HPVs alter cellular activities to complete their replicative-cycle in differentiating epithelium. Understanding how HPVs interface with Rab functions during their complex replicative cycle may provide insight for the development of therapeutic interventions, as current viral counter-measures are solely prophylactic and therapies for HPV-positive individuals remain archaic and limited.
Collapse
Affiliation(s)
- Jesse M. Young
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Amira Zine El Abidine
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Ricardo A. Gómez-Martinez
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
- Department of Obstetrics & Gynecology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Michelle A. Ozbun
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
- Department of Obstetrics & Gynecology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| |
Collapse
|
170
|
Xu X, Chen Y, Wang X, Mu X. Role of Hippo/YAP signaling in irradiation-induced glioma cell apoptosis. Cancer Manag Res 2019; 11:7577-7585. [PMID: 31496812 PMCID: PMC6693089 DOI: 10.2147/cmar.s210825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/06/2019] [Indexed: 12/22/2022] Open
Abstract
Background Although Hippo/Yes-associated protein (YAP) signaling plays crucial roles in radiation sensitivity and resistance of multiple kinds of cancers, its role in the radiation sensitivity of glioma cells remains unclear. The present study aimed to reveal Hippo/YAP role in the radiation sensitivity of glioma cells. Methods Glioma U251 cells were administrated with different doses of irradiation. Cell Counting Kit-8 (CCK-8) and flow cytometry assays were used to assess cell viability and apoptosis. Co-immunoprecipitation (co-IP) assay was used to assess the interactions between proteins. Results The results showed that irradiation exposure significantly inhibited cell viability and induced cell apoptosis in a dose-dependent manner, as well as decreased YAP1 expression via enhancing RCHY1-mediated YAP1 protein degradation. In addition, we observed that downregulation of YAP1 or RCHY1 weakened the role of irradiation exposure in cell viability inhibition and apoptosis promotion. Conclusion Collectively, this study emphasizes the vital role of Hippo/YAP signaling in radiation sensitivity of glioma, that RCHY1-mediated YAP1 protein downregulation is a main mechanism accounting for radiation-induced glioma cell apoptosis. Our study may enrich the theoretical basis of Hippo/YAP signaling as a new target for improving radiation sensitivity in glioma.
Collapse
Affiliation(s)
- Xiaofei Xu
- Department of Radiology, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| | - Yan Chen
- Department of Neurosurgery, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| | - Xi Wang
- Department of Radiology, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| | - Xingguo Mu
- Department of Radiology, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| |
Collapse
|
171
|
Yin L, Li W, Wang G, Shi H, Wang K, Yang H, Peng B. NR1B2 suppress kidney renal clear cell carcinoma (KIRC) progression by regulation of LATS 1/2-YAP signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:343. [PMID: 31391070 PMCID: PMC6686564 DOI: 10.1186/s13046-019-1344-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Kidney Renal Clear Cell Carcinoma (KIRC) accounts for 75% of all renal cancers. Previous study had conflict evidences regarding NR1B2 role in cancer, and its expression and biological role in KIRC remained unclear. Our aims were to characterize the role of NR1B2 in KIRC. METHODS NR1B2 expression in TCGA database were analyzed. Clinical KIRC samples were examined by RT-PCR, western blot and tissue microarray (TMA). The relationship between NR1B2 expression and the clinical characteristics were evaluated. KIRC cell line were stably overexpressed NR1B2 or with an NR1B2 knocked down using lentivirus system. The cells were analyzed by migration and invasion assay, then injected into nude mice to assess tumor growth and metastasis. EMT marker expression and LATS 1/2-YAP pathway demonstration were detected by the TCGA database and western blot. RESULTS The expression of NR1B2 in KIRC was significantly down-regulated in the TCGA database and our clinical samples. Moreover, NR1B2 expression negatively correlated with tumor stage and positively correlated with overall and disease-free survival rate. Univariate and multivariate analyses indicated the expression level of NR1B2 could be used as an independent factor for predicting the prognosis of KIRC. Overexpression NR1B2 significantly inhibited and knockdown NR1B2 markedly promoted KIRC cell invasion and metastasis both in vitro and in vivo. Mechanistic investigations revealed that NR1B2 might be a tumor suppressor to inhibit EMT through the LATS1/2-YAP pathway. CONCLUSIONS our results defined NR1B2 as a tumor suppressor in KIRC that restricted EMT by the LATS1/2-YAP pathway.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Wenjia Li
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangchun Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Heng Shi
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.,Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, China
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Huan Yang
- Department of Urology, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.
| |
Collapse
|
172
|
Hsu PC, Jablons DM, Yang CT, You L. Epidermal Growth Factor Receptor (EGFR) Pathway, Yes-Associated Protein (YAP) and the Regulation of Programmed Death-Ligand 1 (PD-L1) in Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2019; 20:ijms20153821. [PMID: 31387256 PMCID: PMC6695603 DOI: 10.3390/ijms20153821] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/02/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) pathway is a well-studied oncogenic pathway in human non-small cell lung cancer (NSCLC). A subset of advanced NSCLC patients (15–55%) have EGFR-driven mutations and benefit from treatment with EGFR-tyrosine kinase inhibitors (TKIs). Immune checkpoint inhibitors (ICIs) targeting the PD-1/PDL-1 axis are a new anti-cancer therapy for metastatic NSCLC. The anti-PD-1/PDL-1 ICIs showed promising efficacy (~30% response rate) and improved the survival of patients with metastatic NSCLC, but the role of anti-PD-1/PDL-1 ICIs for EGFR mutant NSCLC is not clear. YAP (yes-associated protein) is the main mediator of the Hippo pathway and has been identified as promoting cancer progression, drug resistance, and metastasis in NSCLC. Here, we review recent studies that examined the correlation between the EGFR, YAP pathways, and PD-L1 and demonstrate the mechanism by which EGFR and YAP regulate PD-L1 expression in human NSCLC. About 50% of EGFR mutant NSCLC patients acquire resistance to EGFR-TKIs without known targetable secondary mutations. Targeting YAP therapy is suggested as a potential treatment for NSCLC with acquired resistance to EGFR-TKIs. Future work should focus on the efficacy of YAP inhibitors in combination with immune checkpoint PD-L1/PD-1 blockade in EGFR mutant NSCLC without targetable resistant mutations.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - David M Jablons
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Liang You
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA.
| |
Collapse
|
173
|
Karvonen H, Barker H, Kaleva L, Niininen W, Ungureanu D. Molecular Mechanisms Associated with ROR1-Mediated Drug Resistance: Crosstalk with Hippo-YAP/TAZ and BMI-1 Pathways. Cells 2019; 8:cells8080812. [PMID: 31382410 PMCID: PMC6721603 DOI: 10.3390/cells8080812] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Signaling via the Wnt-related receptor tyrosine kinase-like orphan receptor 1 (ROR1) triggers tumorigenic features associated with cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT), while aberrant expression of ROR1 is strongly linked to advanced disease progression and chemoresistance. Several recent studies have shown that Wnt5a binding to ROR1 promotes oncogenic signaling by activating multiple pathways such as RhoA/Rac1 GTPases and PI3K/AKT, which in turn could induce transcriptional coactivator YAP/TAZ or polycomb complex protein BMI-1 signaling, respectively, to sustain stemness, metastasis and ultimately drug-resistance. These data point towards a new feedback loop during cancer development, linking Wnt5a-ROR1 signaling activation to YAP/TAZ or BMI-1 upregulation that could play an important role in disease progression and treatment resistance. This review focuses on the crosstalk between Wnt5a-ROR1 and YAP/TAZ or the BMI-1 signaling network, together with the current advancements in targeted strategies for ROR1-positive cancers.
Collapse
Affiliation(s)
- Hanna Karvonen
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland
| | - Harlan Barker
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Laura Kaleva
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland
| | - Wilhelmiina Niininen
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland
| | - Daniela Ungureanu
- Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland.
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland.
| |
Collapse
|
174
|
Balasubramaniam SD, Balakrishnan V, Oon CE, Kaur G. Key Molecular Events in Cervical Cancer Development. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E384. [PMID: 31319555 PMCID: PMC6681523 DOI: 10.3390/medicina55070384] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 01/04/2023]
Abstract
Cervical cancer is the fourth most common cancer among women. Infection by high-risk human papillomavirus (HPV) is the main aetiology for the development of cervical cancer. Infection by high-risk human papillomavirus (HPV) and the integration of the HPV genome into the host chromosome of cervical epithelial cells are key early events in the neoplastic progression of cervical lesions. The viral oncoproteins, mainly E6 and E7, are responsible for the initial changes in epithelial cells. The viral proteins inactivate two main tumour suppressor proteins, p53, and retinoblastoma (pRb). Inactivation of these host proteins disrupts both the DNA repair mechanisms and apoptosis, leading to rapid cell proliferation. Multiple genes involved in DNA repair, cell proliferation, growth factor activity, angiogenesis, as well as mitogenesis genes become highly expressed in cervical intraepithelial neoplasia (CIN) and cancer. This genomic instability encourages HPV-infected cells to progress towards invasive carcinoma. The key molecular events involved in cervical carcinogenesis will be discussed in this review.
Collapse
Affiliation(s)
| | - Venugopal Balakrishnan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia.
| |
Collapse
|
175
|
Liu K, Du S, Gao P, Zheng J. Verteporfin suppresses the proliferation, epithelial-mesenchymal transition and stemness of head and neck squamous carcinoma cells via inhibiting YAP1. J Cancer 2019; 10:4196-4207. [PMID: 31413738 PMCID: PMC6691709 DOI: 10.7150/jca.34145] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/26/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose: Yes-associated protein 1 (YAP1) is overexpressed in head and neck squamous cell carcinoma (HNSCC). However, it is unknown whether verteporfin, a YAP1 inhibitor, can inhibit HNSCC cells as well as the molecular mechanisms involved. Methods: YAP1 expression was investigated by immunohistochemistry in human head and neck carcinoma tissues (n=70). CCK-8 assay, colony formation assay, flow cytometric analysis, wound-healing assay and Transwell migration and invasion assays were used to evaluated the effects of verteporfin on the six HNSCC cell lines (three HPV-positive and three HPV-negative). The transcription and protein expression levels of YAP1 and its associated genes were investigated by real-time PCR and Western blotting, respectively. The effects of verteporfin on HNSCC cells in vivo were assessed by a xenograft model. Results: YAP1 expression was significantly higher in carcinoma tissues than in tumor-adjacent normal tissues (n=10). A CCK-8 assay showed that the inhibitory effects of verteporfin on HNSCC cells were markedly enhanced by light activation. Verteporfin significantly inhibited HNSCC cell proliferation, migration and invasion, induced apoptosis, and arrested the cell cycle at the S/G2 phase. Verteporfin significantly attenuated the expression of genes related to epithelial-mesenchymal transition (YAP1, Snail, CTNNB1 and EGFR) and stemness (Oct4 and YAP1) and increased E-cadherin expression in HNSCC cells. Furthermore, verteporfin significantly inhibited PD-L1 expression in HNSCC cells. However, the expression levels of HPV-16 E6 and E7 did not change with VP treatment. The anticancer effect of verteporfin on HNSCC was confirmed by the inhibition of xenograft growth in vivo. Conclusions: Our results indicate that YAP1 overexpression is involved in HNSCC tumorigenesis and verteporfin is a potential therapeutic drug for HNSCC.
Collapse
Affiliation(s)
- Kui Liu
- Department of Pathology, Medical School of Southeast University, Nanjing 210009, China
| | - Shanmei Du
- Department of Pathology, Medical School of Southeast University, Nanjing 210009, China.,Zibo Vocational Institute, Zibo 255314, China
| | - Peng Gao
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA19104, USA.,Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jie Zheng
- Department of Pathology, Medical School of Southeast University, Nanjing 210009, China
| |
Collapse
|
176
|
Autocrine STAT3 activation in HPV positive cervical cancer through a virus-driven Rac1-NFκB-IL-6 signalling axis. PLoS Pathog 2019; 15:e1007835. [PMID: 31226168 PMCID: PMC6608985 DOI: 10.1371/journal.ppat.1007835] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/03/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
Persistent human papillomavirus (HPV) infection is the leading cause of cervical cancer. Although the fundamental link between HPV infection and oncogenesis is established, the specific mechanisms of virus-mediated transformation are not fully understood. We previously demonstrated that the HPV encoded E6 protein increases the activity of the proto-oncogenic transcription factor STAT3 in primary human keratinocytes; however, the molecular basis for STAT3 activation in cervical cancer remains unclear. Here, we show that STAT3 phosphorylation in HPV positive cervical cancer cells is mediated primarily via autocrine activation by the pro-inflammatory cytokine Interleukin 6 (IL-6). Antibody-mediated blockade of IL-6 signalling in HPV positive cells inhibits STAT3 phosphorylation, whereas both recombinant IL-6 and conditioned media from HPV positive cells leads to increased STAT3 phosphorylation within HPV negative cervical cancer cells. Interestingly, we demonstrate that activation of the transcription factor NFκB, involving the small GTPase Rac1, is required for IL-6 production and subsequent STAT3 activation. Our data provides new insights into the molecular re-wiring of cancer cells by HPV E6. We reveal that activation of an IL-6 signalling axis drives the autocrine and paracrine phosphorylation of STAT3 within HPV positive cervical cancers cells and that activation of this pathway is essential for cervical cancer cell proliferation and survival. Greater understanding of this pathway provides a potential opportunity for the use of existing clinically approved drugs for the treatment of HPV-mediated cervical cancer.
Collapse
|
177
|
Hsu PC, Tian B, Yang YL, Wang YC, Liu S, Urisman A, Yang CT, Xu Z, Jablons DM, You L. Cucurbitacin E inhibits the Yes‑associated protein signaling pathway and suppresses brain metastasis of human non‑small cell lung cancer in a murine model. Oncol Rep 2019; 42:697-707. [PMID: 31233205 PMCID: PMC6610039 DOI: 10.3892/or.2019.7207] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Human non-small cell lung cancer (NSCLC) is associated with an extremely poor prognosis especially for the 40% of patients who develop brain metastasis, and few treatment strategies exist. Cucurbitacin E (CuE), an oxygenated tetracyclic triterpenoid isolated from plants particularly of the family Cucurbitaceae, has shown anti-tumorigenic properties in several types of cancer, yet the mechanism remains unclear. Yes-associated protein (YAP), a main mediator of the Hippo signaling pathway, promotes tumorigenesis, drug resistance and metastasis in human NSCLC. The present study was designed to ascertain whether CuE inhibits YAP and its downstream gene expression in the human NSCLC cell lines H2030-BrM3 (K-rasG12C mutation) and PC9-BrM3 (EGFRΔexon19 mutation), which have high potential for brain metastasis. The efficacy of CuE in suppressing brain metastasis of H2030-BrM3 cells in a murine model was also investigated. It was found that after CuE treatment in H2030-BrM3 and PC9-BrM3 cells, YAP protein expression was decreased, and YAP signaling GTIIC reporter activity and expression of the downstream genes CTGF and CYR61 were significantly (P<0.01) decreased. CuE treatment also reduced the migration and invasion abilities of the H2030-BrM3 and PC9-BrM3 cells. Finally, our in vivo study showed that CuE treatment (0.2 mg/kg) suppressed H2030-BrM3 cell brain metastasis and that mice treated with CuE survived longer than the control mice treated with 10% DMSO (P=0.02). The present study is the first to demonstrate that CuE treatment inhibits YAP and the signaling downstream gene expression in human NSCLC in vitro, and suppresses brain metastasis of NSCLC in a murine model. More studies to verify the promising efficacy of CuE in inhibiting brain metastasis of NSCLC and various other cancers may be warranted.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Bo Tian
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Yi-Lin Yang
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Yu-Cheng Wang
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Shu Liu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Anatoly Urisman
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33305, Taiwan, R.O.C
| | - Zhidong Xu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - David M Jablons
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Liang You
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115, USA
| |
Collapse
|
178
|
Huh HD, Kim DH, Jeong HS, Park HW. Regulation of TEAD Transcription Factors in Cancer Biology. Cells 2019; 8:E600. [PMID: 31212916 PMCID: PMC6628201 DOI: 10.3390/cells8060600] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Transcriptional enhanced associate domain (TEAD) transcription factors play important roles during development, cell proliferation, regeneration, and tissue homeostasis. TEAD integrates with and coordinates various signal transduction pathways including Hippo, Wnt, transforming growth factor beta (TGFβ), and epidermal growth factor receptor (EGFR) pathways. TEAD deregulation affects well-established cancer genes such as KRAS, BRAF, LKB1, NF2, and MYC, and its transcriptional output plays an important role in tumor progression, metastasis, cancer metabolism, immunity, and drug resistance. To date, TEADs have been recognized to be key transcription factors of the Hippo pathway. Therefore, most studies are focused on the Hippo kinases and YAP/TAZ, whereas the Hippo-dependent and Hippo-independent regulators and regulations governing TEAD only emerged recently. Deregulation of the TEAD transcriptional output plays important roles in tumor progression and serves as a prognostic biomarker due to high correlation with clinicopathological parameters in human malignancies. In addition, discovering the molecular mechanisms of TEAD, such as post-translational modifications and nucleocytoplasmic shuttling, represents an important means of modulating TEAD transcriptional activity. Collectively, this review highlights the role of TEAD in multistep-tumorigenesis by interacting with upstream oncogenic signaling pathways and controlling downstream target genes, which provides unprecedented insight and rationale into developing TEAD-targeted anticancer therapeutics.
Collapse
Affiliation(s)
- Hyunbin D Huh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| | - Dong Hyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| | - Han-Sol Jeong
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea.
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| |
Collapse
|
179
|
Hamon A, García-García D, Ail D, Bitard J, Chesneau A, Dalkara D, Locker M, Roger JE, Perron M. Linking YAP to Müller Glia Quiescence Exit in the Degenerative Retina. Cell Rep 2019; 27:1712-1725.e6. [DOI: 10.1016/j.celrep.2019.04.045] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/25/2019] [Accepted: 04/09/2019] [Indexed: 12/30/2022] Open
|
180
|
Smith NJ, Fenton TR. The APOBEC3 genes and their role in cancer: insights from human papillomavirus. J Mol Endocrinol 2019; 62:R269-R287. [PMID: 30870810 DOI: 10.1530/jme-19-0011] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022]
Abstract
The interaction between human papillomaviruses (HPV) and the apolipoprotein-B mRNA-editing catalytic polypeptide-like (APOBEC)3 (A3) genes has garnered increasing attention in recent years, with considerable efforts focused on understanding their apparent roles in both viral editing and in HPV-driven carcinogenesis. Here, we review these developments and highlight several outstanding questions in the field. We consider whether editing of the virus and mutagenesis of the host are linked or whether both are essentially separate events, coincidentally mediated by a common or distinct A3 enzymes. We discuss the viral mechanisms and cellular signalling pathways implicated in A3 induction in virally infected cells and examine which of the A3 enzymes might play the major role in HPV-associated carcinogenesis and in the development of therapeutic resistance. We consider the parallels between A3 induction in HPV-infected cells and what might be causing aberrant A3 activity in HPV-independent cancers such as those arising in the bladder, lung and breast. Finally, we discuss the implications of ongoing A3 activity in tumours under treatment and the therapeutic opportunities that this may present.
Collapse
Affiliation(s)
- Nicola J Smith
- School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Tim R Fenton
- School of Biosciences, University of Kent, Canterbury, Kent, UK
| |
Collapse
|
181
|
Zhang C, Liu J, Tao F, Lu Y, He Q, Zhao L, Ou R, Xu Y, Li W. Retracted Article: The nuclear export of TR3 mediated gambogic acid-induced apoptosis in cervical cancer cells through mitochondrial dysfunction. RSC Adv 2019; 9:11855-11864. [PMID: 35516982 PMCID: PMC9063542 DOI: 10.1039/c8ra10542a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 03/29/2019] [Indexed: 12/02/2022] Open
Abstract
At present, chemotherapy is still the main treatment for cervical cancer. However, the drug resistance of chemotherapy drugs seriously restricts its use, so it is urgent to develop new drugs for cervical cancer. Some studies have shown that gambogic acid has a strong anti-tumor effect, while the anti-tumor effect and molecular mechanism of gambogic acid on cervical cancer need to be studied. Our study confirms that the cytotoxic effect of gambogic acid on cervical cancer cells depends on the expression of TR3 protein. Moreover, gambogic acid-induced apoptosis requires TR3 expression. In the mechanism, gambogic acid promoted nuclear export of TR3, resulting in up-regulation of p53, which leads to the decrease of mitochondrial membrane potential, eventually inducing apoptosis. These results suggest that the nuclear export of TR3 mediated gambogic acid-induced apoptosis through a p53-dependent apoptosis pathway.
Collapse
Affiliation(s)
- Chunhong Zhang
- Department of Pharmacy, The First Affliated Hospital of Wenzhou Medical University Wenzhou Zhejiang Province China
| | - Jia Liu
- Department of Dermatovenereology, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
- Plastic and Cosmetic Center, The Affiliated Eye Hospital of Wenzhou Medical University Wenzhou Zhejiang Province China
| | - Fengxing Tao
- Department of Dermatovenereology, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
| | - Yiyi Lu
- Department of Dermatovenereology, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
| | - Qin He
- Department of Dermatovenereology, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
| | - Liang Zhao
- Laboratory for Advanced Interdisciplinary Research, Institute of Translational Medicine, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
| | - Rongying Ou
- Department of Gynaecology and Obstetrics, The First Affliated Hospital of Wenzhou Medical University Wenzhou Zhejiang Province China
| | - Yunsheng Xu
- Department of Dermatovenereology, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
| | - Wenfeng Li
- Laboratory for Advanced Interdisciplinary Research, Institute of Translational Medicine, The First Affliated Hospital of Wenzhou Medical University Nanbaixiang Street Wenzhou Zhejiang Province China
| |
Collapse
|
182
|
Deng H, Sun Y, Wang W, Li M, Yuan T, Kong W, Huang X, Long Z, Chen Z, Wang D, Yang Q. The hippo pathway effector Yes-associated protein promotes epithelial proliferation and remodeling in chronic rhinosinusitis with nasal polyps. Allergy 2019; 74:731-742. [PMID: 30362580 DOI: 10.1111/all.13647] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/18/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hippo-Yes-associated protein (YAP) pathway plays an important role in epithelial cell proliferation and development. However, its possible role in chronic rhinosinusitis with nasal polyps (CRSwNP) remains unknown. We aim to investigate it on nasal epithelial proliferation and remodeling in CRSwNP. METHODS The expressions of hippo pathway components as well as Ki-67 and E-cadherin in the sinonasal mucosa and nasal epithelial cells were analyzed in 14 controls, 14 eosinophilic CRSwNP, and 14 noneosinophilic CRSwNP. Nasal epithelial cells from 6 controls, 6 eosinophilic CRSwNP, and 6 noneosinophilic CRSwNP were cultured and treated with lipopolysaccharide (LPS), Poly(I:C), or a selective YAP inhibitor verteporfin (VP). RESULTS The hippo pathway components MST1, LATS1/2, YAP, and TEAD1 were increased in both eosinophilic and noneosinophilic CRSwNP, particularly in nasal epithelial cells, along with upregulation of Ki-67 and downregulation of E-cadherin. The mRNA levels of YAP positively correlated with the Ki-67 mRNA levels, and negatively associated with the E-cadherin mRNA levels in polyp tissues and epithelial cells from nasal polyps (NPECs). LPS and Poly(I:C) upregulated the YAP expression in nasal epithelial cells accompanied by increased TEAD1 and Ki-67 expression. Conversely, YAP inhibition by VP decreased TEAD1 and Ki-67 expression in NPECs. CONCLUSIONS Hippo pathway components are abnormally upregulated in NPECs, and its effector YAP promotes nasal epithelial cells proliferation and remodeling in CRSwNP. It provides a rationale to explore inhibition of YAP as a novel therapeutic strategy for reducing the epithelial proliferation and remodeling in CRSwNP.
Collapse
Affiliation(s)
- Huiyi Deng
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Yueqi Sun
- Otorhinolaryngology Hospital The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Weihao Wang
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Meijiao Li
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Tian Yuan
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Weifeng Kong
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Xuekun Huang
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Zijie Long
- Department of Hematology The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Zhuanggui Chen
- Department of Pediatrics The Third Affiliated Hospital Sun Yat‐Sen University Guangzhou China
| | - Deyun Wang
- Department of Otolaryngology National University Health System National University of Singapore Singapore Singapore
| | - Qintai Yang
- Department of Otorhinolaryngology‐Head and Neck Surgery The Third Affiliated Hospital Sun Yat‐sen University Guangzhou China
| |
Collapse
|
183
|
He C, Lv X, Huang C, Hua G, Ma B, Chen X, Angeletti PC, Dong J, Zhou J, Wang Z, Rueda BR, Davis JS, Wang C. YAP1-LATS2 feedback loop dictates senescent or malignant cell fate to maintain tissue homeostasis. EMBO Rep 2019; 20:e44948. [PMID: 30755404 PMCID: PMC6399607 DOI: 10.15252/embr.201744948] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/16/2022] Open
Abstract
Dysfunction of the homeostasis-maintaining systems in specific cell types or tissues renders the organism susceptible to a range of diseases, including cancers. One of the emerging mechanisms for maintaining tissue homeostasis is cellular senescence. Here, we report that the Hippo pathway plays a critical role in controlling the fate of ovarian cells. Hyperactivation of Yes-associated protein 1 (YAP1), the major effector of the Hippo pathway, induces senescence in cultured primary human ovarian surface epithelial cells (hOSEs). Large tumor suppressor 2 (LATS2), the primary upstream negative regulator of YAP1, is elevated in both YAP1-induced and natural replicative-triggered senescence. Deletion of LATS2 in hOSEs prevents these cells from natural replicative and YAP1-induced senescence. Most importantly, loss of LATS2 switches ovarian cells from YAP-induced senescence to malignant transformation. Our results demonstrate that LATS2 and YAP1, two major components of the Hippo/YAP signaling pathway, form a negative feedback loop to control YAP1 activity and prevent ovarian cells from malignant transformation. Human cancer genomic data extracted from TCGA datasets further confirm the clinical relevance of our finding.
Collapse
Affiliation(s)
- Chunbo He
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cong Huang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Guohua Hua
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bowen Ma
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter C Angeletti
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jixin Dong
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jin Zhou
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Department of Obstetrics and gynecology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Zhengfeng Wang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Omaha Veterans Affairs Medical Center, Omaha, NE, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
184
|
Establishment, molecular and biological characterization of HCB-514: a novel human cervical cancer cell line. Sci Rep 2019; 9:1913. [PMID: 30760827 PMCID: PMC6374403 DOI: 10.1038/s41598-018-38315-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/19/2018] [Indexed: 01/28/2023] Open
Abstract
Cervical cancer is the fourth most common cancer in women. Although cure rates are high for early stage disease, clinical outcomes for advanced, metastatic, or recurrent disease remain poor. To change this panorama, a deeper understanding of cervical cancer biology and novel study models are needed. Immortalized human cancer cell lines such as HeLa constitute crucial scientific tools, but there are few other cervical cancer cell lines available, limiting our understanding of a disease known for its molecular heterogeneity. This study aimed to establish novel cervical cancer cell lines derived from Brazilian patients. We successfully established one (HCB-514) out of 35 cervical tumors biopsied. We confirmed the phenotype of HCB-514 by verifying its’ epithelial and tumor origin through cytokeratins, EpCAM and p16 staining. It was also HPV-16 positive. Whole-exome sequencing (WES) showed relevant somatic mutations in several genes including BRCA2, TGFBR1 and IRX2. A copy number variation (CNV) analysis by nanostring and WES revealed amplification of genes mainly related to kinases proteins involved in proliferation, migration and cell differentiation, such as EGFR, PIK3CA, and MAPK7. Overexpression of EGFR was confirmed by phospho RTK-array and validated by western blot analysis. Furthermore, the HCB-514 cell line was sensitive to cisplatin. In summary, this novel Brazilian cervical cancer cell line exhibits relevant key molecular features and constitutes a new biological model for pre-clinical studies.
Collapse
|
185
|
Du F, Yu C, Li R, Ding D, He L, Wen G. Expression of miR-141 and YAP1 in gastric carcinoma and modulation of cancer cell proliferation and apoptosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:559-567. [PMID: 31933860 PMCID: PMC6945079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/07/2018] [Indexed: 06/10/2023]
Abstract
Yes-associated protein 1 (YAP1) plays important roles in facilitating cell proliferation and decreasing apoptosis and is related to gastric cancer. Abnormal down-regulation of miR-141 is associated with gastric cancer pathogenesis, suggesting a potentially tumor suppressor role. Bioinformatics analysis found complementary binding sites between miR-141 and YP1. This study investigated the role of miR-141 in mediating YAP1 expression and biological behavior of gastric cancer cells. Gastric cancer tissues were collected using normal mucosal tissues as the control. qRT-PCR compared expression of miR-141 and YAP1 mRNA, and western blot quantified YAP1 protein expression. Spearman approach analyzed the correlation between miR-141 and YAP1 mRNA in cancer tissues. Dual luciferase reporter gene assay confirmed the targeted regulation between miR-141 and YAP1. Using GES-1 cell as the control, miR-141 and YAP1 expression were measured in gastric cancer cell lines SGC7901 and MGC03. Those cells were transfected with miR-141 mimic in the presence or absence of miR-YAP1 mimic followed by flow cytometry for apoptosis and EdU staining for proliferation. Cancer tissues had decreased miR-141 and higher YAP1 expression, which was associated with TNM stage. YAP1 mRNA and miR-141 were positively correlated (r=-0.623, P<0.001). Dual luciferase assay demonstrated targeted regulation between miR-141 and YAP1. Comparing to GES-1 cells, SGC7901 and MGC803 cells had decreased miR-141 and increased YAP1 expression. Transfection of miR-141 mimic inhibited YAP1 expression or cell proliferation and facilitated apoptosis. However, overexpression of YAP1 decreased the effect of miR-141 mimic on cell proliferation and apoptosis. miR-141 down-regulation and YAP1 up-regulation are correlated with gastric cancer pathogenesis. miR-141 targets and inhibits YAP1 expression, to suppress gastric cancer cell proliferation and induce apoptosis.
Collapse
Affiliation(s)
- Fangchao Du
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University Hefei 230061, Anhui, China
| | - Chao Yu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University Hefei 230061, Anhui, China
| | - Rui Li
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University Hefei 230061, Anhui, China
| | - Ding Ding
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University Hefei 230061, Anhui, China
| | - Lei He
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University Hefei 230061, Anhui, China
| | - Gang Wen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University Hefei 230061, Anhui, China
| |
Collapse
|
186
|
The Ambivalent Function of YAP in Apoptosis and Cancer. Int J Mol Sci 2018; 19:ijms19123770. [PMID: 30486435 PMCID: PMC6321280 DOI: 10.3390/ijms19123770] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/16/2018] [Accepted: 11/23/2018] [Indexed: 02/07/2023] Open
Abstract
Yes-associated protein, a core regulator of the Hippo-YAP signaling pathway, plays a vital role in inhibiting apoptosis. Thus, several studies and reviews suggest that yes-associated protein is a good target for treating cancer. Unfortunately, more and more evidence demonstrates that this protein is also an essential contributor of p73-mediated apoptosis. This questions the concept that yes-associated protein is always a good target for developing novel anti-cancer drugs. Thus, the aim of this review was to evaluate the clinical relevance of yes-associated protein for cancer pathophysiology. This review also summarized the molecules, processes and drugs, which regulate Hippo-YAP signaling and discusses their effect on apoptosis. In addition, issues are defined, which should be addressed in the future in order to provide a solid basis for targeting the Hippo-YAP signaling pathway in clinical trials.
Collapse
|
187
|
Obasi TC, Braicu C, Iacob BC, Bodoki E, Jurj A, Raduly L, Oniga I, Berindan-Neagoe I, Oprean R. Securidaca-saponins are natural inhibitors of AKT, MCL-1, and BCL2L1 in cervical cancer cells. Cancer Manag Res 2018; 10:5709-5724. [PMID: 30532593 PMCID: PMC6245348 DOI: 10.2147/cmar.s163328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Introduction Scientific research is beginning to prove the connection between claims by African traditional medicine and the natural chemical specifics contained in medicinal plant Securidaca longipedunculata. Our previous studies showed that two natural saponin fractions (4A3 and 4A4) identified in the plant as triterpenoid glycosides are capable of activating apoptosis on cervical tumor cell lines. Considering this and some critical roles of human papillomavirus (HPV) E6 oncogene on cervical cells, by promoting carcinogenesis and cell survival, it became necessary to investigate the possible pathways for apoptosis transmission. Methods Tests conducted on relevant cervical tumor cell lines such as Caski and Bu25TK included the following: MTT assay; scratch assay (to determine cell migration/invasion); fluorescence microscopy with Annexin V–fluorescein isothiocyanate, muscle progenitor cell) and propidium iodide staining; and finally reverse transcriptase quantitative PCR (RT-qPCR) for gene analysis. Results Reduced cell proliferation was observed due to activities of 4A3 and 4A4 fractions, with half-maximal inhibitory concentration (IC50) of 7.03 and 16.39 μg/mL, respectively, on Caski cell line. A significant reduction in cell migration occurred within 48 and 72 hours, respectively, for Caski and Bu25TK cell lines. Late apoptosis was activated by 4A3, staining both Annexin V and PI, in contrast to 4A4’s early apoptosis. RT-qPCR data revealed a fold change (FC) inhibition of antiapoptotic proteins such as MCL-1 and BCL2L1, with diminished level of AKT-3, VEGFA, MALAT1, etc. The expression of p53, proapoptotic BAD, and caspase-8 was nonsignificant. Conclusion The low expression of AKT-3 and antiapoptotic proteins (MCL-1 and BCL2L1), as well as VEGFA, could simply be an indication for possible suppression of cell survival mechanisms via multiple channels. We therefore conclude that 4A3 and 4A4 fractions mediate activity via the inhibition of phosphatidylinositol-3-OH kinase (PI3K)-AKT/mTOR/NF-kB-dependent antiapoptotic stimuli. Further studies are ongoing to reveal the chemical structures and compositions of these two fractions.
Collapse
Affiliation(s)
- Titus Chukwuemeka Obasi
- Department of Analytical Chemistry and Instrumental Analysis, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| | - Bogdan Cezar Iacob
- Department of Analytical Chemistry and Instrumental Analysis, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| | - Ede Bodoki
- Department of Analytical Chemistry and Instrumental Analysis, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| | - Ilioara Oniga
- Department of Pharmacognosy, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania, .,MEDFUTURE - Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Cluj-Napoca, Romania, .,Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania,
| | - Radu Oprean
- Department of Analytical Chemistry and Instrumental Analysis, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania,
| |
Collapse
|
188
|
Li G, Yin Q, Ji H, Wang Y, Liu H, Jiang L, Zhu F, Li B. A study on screening and antitumor effect of CD55-specific ligand peptide in cervical cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3899-3912. [PMID: 30519000 PMCID: PMC6239109 DOI: 10.2147/dddt.s182337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background To improve the targeting ability of antitumor drugs, we identified the antigens with high expression on the surface of tumor cells associated with tumor escape, such as the complement regulatory protein CD55 molecule, which is also known as the decay accelerating factor. In this study, phage display technology was used to screen and identify CD55-specific ligand peptide (CD55sp) bound to CD55 molecule on the surface of cervical cancer HeLa cells. We then explored the role of this peptide in inhibiting the growth of cervical cancer cells in vitro. Our characterization of CD55sp will provide implication for tumor target therapy. Methods The phage bound to the surface of HeLa cells were isolated by phage display technology. Positive phage clones were identified by ELISA. Phage was then amplified and determined by agarose gel electrophoresis after monoclonal DNA extraction. DNA sequencing and bioinformatical analysis were conducted to obtain specific ligand peptides. Flow cytometry and immunofluorescence were used to measure the expression of CD55 molecule on the surface of tumor and normal cells. Subsequently, the effects of CD55sp on the proliferation and apoptosis of HeLa and SiHa cells were determined by Cell Counting Kit-8 (CCK-8), flow cytometry, and TUNEL assay, respectively. The morphology of apoptotic cells was examined by electron microscope. The distribution of Cleaved caspase-3 was detected by immunofluorescence. The expression of bcl-2 and Cleaved caspase-3 were determined by Western blot. Results The results showed that the peptide (QVNGLGERSQQM) can bind to the CD55 molecule on the surface of cervical cancer HeLa and SiHa cells as a ligand peptide. It can also effectively inhibit the proliferation of cervical cancer cells and induce cell apoptosis. Conclusion This study demonstrates that CD55sp screened by phage display technology plays a strong antitumor role.
Collapse
Affiliation(s)
- Guoxiang Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Qifeng Yin
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Huanhuan Ji
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Yujuan Wang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Huihui Liu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Liangqian Jiang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Feng Zhu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266071, People's Republic of China,
| |
Collapse
|
189
|
Subat S, Inamura K, Ninomiya H, Nagano H, Okumura S, Ishikawa Y. Unique MicroRNA and mRNA Interactions in EGFR-Mutated Lung Adenocarcinoma. J Clin Med 2018; 7:E419. [PMID: 30404194 PMCID: PMC6262391 DOI: 10.3390/jcm7110419] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022] Open
Abstract
The EGFR gene was one of the first molecules to be selected for targeted gene therapy. EGFR-mutated lung adenocarcinoma, which is responsive to EGFR inhibitors, is characterized by a distinct oncogenic pathway in which unique microRNA (miRNA)⁻mRNA interactions have been observed. However, little information is available about the miRNA⁻mRNA regulatory network involved. Both miRNA and mRNA expression profiles were investigated using microarrays in 155 surgically resected specimens of lung adenocarcinoma with a known EGFR mutation status (52 mutated and 103 wild-type cases). An integrative analysis of the data was performed to identify the unique miRNA⁻mRNA regulatory network in EGFR-mutated lung adenocarcinoma. Expression profiling of miRNAs and mRNAs yielded characteristic miRNA/mRNA signatures (19 miRNAs/431 mRNAs) in EGFR-mutated lung adenocarcinoma. Five of the 19 miRNAs were previously listed as EGFR-mutation-specific miRNAs (i.e., miR-532-3p, miR-500a-3p, miR-224-5p, miR-502-3p, and miR-532-5p). An integrative analysis of miRNA and mRNA expression revealed a refined list of putative miRNA⁻mRNA interactions, of which 63 were potentially involved in EGFR-mutated tumors. Network structural analysis provided a comprehensive view of the complex miRNA⁻mRNA interactions in EGFR-mutated lung adenocarcinoma, including DUSP4 and MUC4 axes. Overall, this observational study provides insight into the unique miRNA⁻mRNA regulatory network present in EGFR-mutated tumors. Our findings, if validated, would inform future research examining the interplay of miRNAs and mRNAs in EGFR-mutated lung adenocarcinoma.
Collapse
Affiliation(s)
- Sophia Subat
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo 135-8550, Japan.
| | - Kentaro Inamura
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo 135-8550, Japan.
| | - Hironori Ninomiya
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo 135-8550, Japan.
| | - Hiroko Nagano
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo 135-8550, Japan.
| | - Sakae Okumura
- Department of Thoracic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo 135-8550, Japan.
| | - Yuichi Ishikawa
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo 135-8550, Japan.
| |
Collapse
|
190
|
Liu Y, Ren M, Tan X, Hu L. Distinct Changes in the Expression TAZ are Associated with Normal Cervix and Human Cervical Cancer. J Cancer 2018; 9:4263-4270. [PMID: 30519328 PMCID: PMC6277613 DOI: 10.7150/jca.26623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
The transcriptional coactivator with the PDZ-binding motif (TAZ) has been associated with different types of cancer. In this study, we examined the TAZ protein expression and cellular localization in 194 cases of human cervical squamous cell carcinoma (SCC). We observed that a normal cervix is characterized by higher expression levels of both nuclear and cytosolic TAZ compared to cervical SCC. Lower membranous and cytosolic TAZ expression levels are associated with lymph node involvement. We observed that TAZ expression levels are associated with β1 integrin and Src in SCC and cell lines derived from human cervical cancers. Of note, knock down of TAZ increased the expression of β1 integrin and Src in both normal and human cervical cancer cells. Our data indicate that the expression and cellular localization of TAZ are inversely associated with the development and progression of cervical SCC, and TAZ-mediated transcription may be involved in the activation of the integrin-Src signalling pathway.
Collapse
Affiliation(s)
- Yaofang Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China
| | - Meiping Ren
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Xiaoyong Tan
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Lina Hu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P.R. China
| |
Collapse
|
191
|
Zhang RQ, Sun XF, Wu RY, Cheng SF, Zhang GL, Zhai QY, Liu XL, Zhao Y, Shen W, Li L. Zearalenone exposure elevated the expression of tumorigenesis genes in mouse ovarian granulosa cells. Toxicol Appl Pharmacol 2018; 356:191-203. [DOI: 10.1016/j.taap.2018.08.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 01/13/2023]
|
192
|
Liu S, Song L, Yao H, Zhang L, Xu D, Li Q, Li Y. Preserved miR-361-3p Expression Is an Independent Prognostic Indicator of Favorable Survival in Cervical Cancer. DISEASE MARKERS 2018; 2018:8949606. [PMID: 30344797 PMCID: PMC6174793 DOI: 10.1155/2018/8949606] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/09/2018] [Accepted: 07/26/2018] [Indexed: 01/12/2023]
Abstract
In this study, we aimed to assess the independent prognostic value of miR-361-3p in terms of overall survival (OS) and recurrence-free survival (RFS) in cervical cancer, as well as its possible regulative network. A retrospective analysis was performed by using data from the Cancer Genome Atlas-Cervical Cancer (TCGA-CESC). Results showed that decreased miR-361-3p expression was associated with lymphovascular invasion and poor responses to primary therapy. The patients with recurrence and the deceased cases had substantially lower miR-361-3p expression compared to their respective controls. By generating Kaplan-Meier curves of OS and RFS, we found that high miR-361-3p expression was associated with better survival outcome. More importantly, univariate and multivariate analysis confirmed that high miR-361-3p expression was an independent indicator of favorable OS (HR: 0.377, 95% CI: 0.233-0.608, p < 0.001) and RFS (HR: 0.398, 95% CI: 0.192-0.825, p = 0.013). By performing bioinformatic analysis, we identified 24 genes that were negatively correlated with miR-361-3p expression. Among the potential targeting genes, SOST, MTA1, TFRC, and YAP1 are involved in some important signaling pathways modulating cervical cancer cell invasion, migration, and drug sensitivity. Therefore, it is meaningful to verify the potential regulative effect of miR-361-3p on the expression of these genes in the future.
Collapse
Affiliation(s)
- Shikai Liu
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| | - Lili Song
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| | - Hairong Yao
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| | - Liang Zhang
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| | - Dongkui Xu
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| | - Qian Li
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| | - Ying Li
- Department of Obstetrics & Gynecology, Cangzhou Central Hospital, Hebei 061001, China
| |
Collapse
|
193
|
Liu BS, Dai XY, Xia HW, Xu HJ, Tang QL, Gong QY, Nie YZ, Bi F. Geranylgeranyl transferase 1 inhibitor GGTI‑298 enhances the anticancer effect of gefitinib. Mol Med Rep 2018; 18:4023-4029. [PMID: 30106149 DOI: 10.3892/mmr.2018.9371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/11/2018] [Indexed: 02/05/2023] Open
Abstract
Dysregulation of epidermal growth factor receptor (EGFR) signaling is responsible for the resistance to EGFR tyrosine kinase inhibitors (TKIs), such as gefitinib and erlotinib, and is thereby associated with the progression of tumors in non‑small cell lung cancers (NSCLCs). Immunoblotting results revealed that geranylgeranyl transferase 1 inhibitor (GGTI)‑298, a geranylgeranyl transferase 1 inhibitor with potential antitumor effects, effectively inhibited the phosphorylation of EGFR and its downstream target protein kinase B (AKT). A combination of gefitinib and GGTI‑298 amplified the inhibition of the EGFR‑AKT signaling pathway. In addition, GGTI‑298 treatment produced a synergistic effect on the inhibition of proliferation as indicated by the combination index values of <1 when combined with gefitinib in the NSCLC cell lines HCC827 and A549. These synergistic effects were also observed to induce apoptosis and migration inhibition. Further mechanistic studies demonstrated that GGTI‑298 inhibited the activity of Ras homolog family member A (RhoA), and downregulation of RhoA with small interfering RNA impaired the phosphorylation of EGFR, which suggested that EGFR inhibition by GGTI‑298 may be exerted mainly through RhoA mediation. These results presented a novel, promising therapeutic strategy involving a combination of two drugs for targeting EGFR signaling in lung cancer.
Collapse
Affiliation(s)
- Bi-Sheng Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xin-Yu Dai
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong-Wei Xia
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Huan-Ji Xu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qiu-Lin Tang
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qi-Yong Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yong-Zhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digest Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Feng Bi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
194
|
Deng Q, Jiang G, Wu Y, Li J, Liang W, Chen L, Su Q, Li W, Du J, Wong CKC, Chen Z, Wang H. GPER/Hippo-YAP signal is involved in Bisphenol S induced migration of triple negative breast cancer (TNBC) cells. JOURNAL OF HAZARDOUS MATERIALS 2018; 355:1-9. [PMID: 29758456 DOI: 10.1016/j.jhazmat.2018.05.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 05/20/2023]
Abstract
Nowadays, risk factors of triple-negative breast cancer (TNBC) metastasis are not well identified. Our present study reveals that an industrial chemical, bisphenol S (BPS), can promote the migration, but not the proliferation, of TNBC cells in vitro. BPS activates YAP, a key effector of Hippo pathway, by inhibiting its phosphorylation, which promotes YAP nuclear accumulation and up-regulates its downstream genes such as CTGF and ANKRD1. Inhibition of YAP blocks the BPS-triggered cell migration and up-regulation of fibronectin (FN) and vimentin (Vim). BPS rapidly decreases the phosphorylation levels of LATS1 (Ser909) in TNBC cells, which regulates the activation and functions of YAP. Silencing LATS1/2 by siRNA increases BPS-induced dephosphorylation of YAP and extended the half-life of YAP protein. Inhibition of G protein-coupled estrogen receptor 1 (GPER) and its downstream PLCβ/PKC signals attenuate the effects of BPS-induced YAP dephosphorylation and CTGF up-regulation. Targeted inhibition of GPER/YAP inhibits BPS-induced migration of TNBC cells. Collectively, we reveal that GPER/Hippo-YAP signal is involved in BPS-induced migration of TNBC cells.
Collapse
Affiliation(s)
- Qianqian Deng
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yingmin Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiexin Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Weiting Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Likun Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wuguo Li
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chris K C Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Zhuojia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Hongsheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
195
|
Seo HM, Moon GT, Song YM, Gee HY, Park YM, Lee JY, Lee JH. Expression of YAP and TAZ in molluscum contagiosum virus infected skin. Br J Dermatol 2018; 179:188-189. [PMID: 29330849 DOI: 10.1111/bjd.16333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- H-M Seo
- Department of Dermatology, Hanyang University Guri Hospital, Seoul, Korea
| | - G T Moon
- Department of Dermatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Y M Song
- Department of Dermatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - H Y Gee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Y M Park
- Department of Dermatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - J Y Lee
- Department of Dermatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - J H Lee
- Department of Dermatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
196
|
Debaugnies M, Sánchez-Danés A, Rorive S, Raphaël M, Liagre M, Parent MA, Brisebarre A, Salmon I, Blanpain C. YAP and TAZ are essential for basal and squamous cell carcinoma initiation. EMBO Rep 2018; 19:embr.201845809. [PMID: 29875149 DOI: 10.15252/embr.201845809] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/04/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023] Open
Abstract
YAP and TAZ are key downstream regulators of the Hippo pathway, regulating cell proliferation and differentiation. YAP and TAZ activation has been reported in different cancer types. However, it remains unclear whether they are required for the initiation of major skin malignancies like basal cell carcinoma (BCC) and squamous cell carcinoma (SCC). Here, we analyze the expression of YAP and TAZ in these skin cancers and evaluate cancer initiation in knockout mouse models. We show that YAP and TAZ are nuclear and highly expressed in different BCC types in both human and mice. Further, we find that cells with nuclear YAP and TAZ localize to the invasive front in well-differentiated SCC, whereas nuclear YAP is homogeneously expressed in spindle cell carcinoma undergoing EMT We also show that mouse BCC and SCC are enriched for YAP gene signatures. Finally, we find that the conditional deletion of YAP and TAZ in mouse models of BCC and SCC prevents tumor formation. Thus, YAP and TAZ are key determinants of skin cancer initiation, suggesting that targeting the YAP and TAZ signaling pathway might be beneficial for the treatment of skin cancers.
Collapse
Affiliation(s)
- Maud Debaugnies
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Adriana Sánchez-Danés
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme University Hospital, University Libre de Bruxelles, Brussels, Belgium
| | - Maylis Raphaël
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Mélanie Liagre
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie-Astrid Parent
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Audrey Brisebarre
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme University Hospital, University Libre de Bruxelles, Brussels, Belgium
| | - Cédric Blanpain
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium .,WELBIO, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
197
|
|
198
|
Kim N, Park YY, Joo CH, Kim HS. Relief of YAP-mediated inhibition by IKKɛ promotes innate antiviral immunity. Cell Mol Immunol 2018; 15:642-644. [PMID: 29057972 PMCID: PMC6078990 DOI: 10.1038/cmi.2017.97] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 12/13/2022] Open
Affiliation(s)
- Nayoung Kim
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea.
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea.
| | - Yun-Yong Park
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea
| | - Chul Hyun Joo
- Department of Microbiology, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea
| | - Hun Sik Kim
- Department of Microbiology, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea.
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 05505, Seoul, Korea.
| |
Collapse
|
199
|
Zhang J, Zhao X, Zhang J, Zheng X, Li F. Circular RNA hsa_circ_0023404 exerts an oncogenic role in cervical cancer through regulating miR-136/TFCP2/YAP pathway. Biochem Biophys Res Commun 2018; 501:428-433. [DOI: 10.1016/j.bbrc.2018.05.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/30/2022]
|
200
|
Park S, Choe M, Yeo H, Han H, Kim J, Chang W, Yun S, Lee H, Lee M. Yes-associated protein mediates human embryonic stem cell-derived cardiomyocyte proliferation: Involvement of epidermal growth factor receptor signaling. J Cell Physiol 2018; 233:7016-7025. [PMID: 29693249 DOI: 10.1002/jcp.26625] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 03/28/2018] [Indexed: 11/09/2022]
Abstract
Unlike mature cardiomyocytes, human pluripotent stem cell-derived cardiomyocytes exhibit higher proliferative capacity; however, the underlying mechanisms involved are yet to be elucidated. Here, we revealed that the Yes-associated protein (YAP) plays a critical role in regulating cell proliferation in association with epidermal growth factor receptor (EGFR) in human embryonic stem cell-derived cardiomyocytes (hESC-CMs). Our results show that low-density culture significantly promotes the proliferation of hESC-CMs via YAP. Interestingly, the low-density culture-induced YAP expression further induced EGFR expression, without any alterations in the activity of EGFR and its two major downstream kinases, ERK, and AKT. However, treatment of a low-density-culture of hESC-CMs with epidermal growth factor (EGF) increased proliferation via phosphorylation of EGFR, ERK, and AKT, and the EGF-induced phosphorylation of EGFR, ERK, and AKT was significantly higher in low-density hESC-CMs than in high-density hESC-CMs. Furthermore, the EGF-induced activation of EGFR, ERK, and AKT increased YAP expression and subsequently proliferation. In conclusion, YAP mediates both low-density culture-induced and EGF-induced proliferation of hESC-CMs in low-density culture conditions.
Collapse
Affiliation(s)
- Somi Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Museog Choe
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Hancheol Yeo
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Hojae Han
- College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Joongsun Kim
- K-herb Research Center, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Woocheol Chang
- Department of Biology Education, College of Education, Pusan National University, Busan, South Korea
| | - Seungpil Yun
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hojin Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Minyoung Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| |
Collapse
|