1
|
Tan WH, Rücklin M, Larionova D, Ngoc TB, Joan van Heuven B, Marone F, Matsudaira P, Winkler C. A Collagen10a1 mutation disrupts cell polarity in a medaka model for metaphyseal chondrodysplasia type Schmid. iScience 2024; 27:109405. [PMID: 38510140 PMCID: PMC10952040 DOI: 10.1016/j.isci.2024.109405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/21/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
Heterozygous mutations in COL10A1 lead to metaphyseal chondrodysplasia type Schmid (MCDS), a skeletal disorder characterized by epiphyseal abnormalities. Prior analysis revealed impaired trimerization and intracellular retention of mutant collagen type X alpha 1 chains as cause for elevated endoplasmic reticulum (ER) stress. However, how ER stress translates into structural defects remained unclear. We generated a medaka (Oryzias latipes) MCDS model harboring a 5 base pair deletion in col10a1, which led to a frameshift and disruption of 11 amino acids in the conserved trimerization domain. col10a1Δ633a heterozygotes recapitulated key features of MCDS and revealed early cell polarity defects as cause for dysregulated matrix secretion and deformed skeletal structures. Carbamazepine, an ER stress-reducing drug, rescued this polarity impairment and alleviated skeletal defects in col10a1Δ633a heterozygotes. Our data imply cell polarity dysregulation as a potential contributor to MCDS and suggest the col10a1Δ633a medaka mutant as an attractive MCDS animal model for drug screening.
Collapse
Affiliation(s)
- Wen Hui Tan
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Martin Rücklin
- Naturalis Biodiversity Center, Postbus 9517, 2300 RA Leiden, the Netherlands
| | - Daria Larionova
- Department of Biology, Research Group Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
| | - Tran Bich Ngoc
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore 117543, Singapore
| | | | - Federica Marone
- Swiss Light Source, Paul Scherrer Institut, CH-5232 Villigen, Switzerland
| | - Paul Matsudaira
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Christoph Winkler
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
2
|
Feng Q, Cui N, Li S, Cao J, Chen Q, Wang H. Upregulation of SOX9 promotes the self-renewal and tumorigenicity of cervical cancer through activating the Wnt/β-catenin signaling pathway. FASEB J 2023; 37:e23174. [PMID: 37668416 DOI: 10.1096/fj.202201596rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 07/30/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Sry-box9 (SOX9) maintains stem cell properties and plays crucial roles in many cancers. However, whether SOX9 is correlated with cervical cancer cell stemness and its detailed mechanism remains obscure. We studied the relationship between SOX9 and prognosis of cervical cancer through public database, and SOX9 was related to poor prognosis of cervical cancer. Elevated SOX9 expression enhanced the self-renewal properties and promotes tumorigenicity in cervical cancer. Overexpression of SOX9 could promote the expression of stem cell-related factors in cervical cancer cells and xenografts. Meanwhile, overexpression of SOX9 could also enhance the expressions of FZD10, β-catenin, and c-Myc in cervical cancer cells and xenografts, while inhibiting the expression of DDK1. The activation of Wnt pathway by chir-99 021 raised the tumor spheroid ability of SOX9 knockdown HeLa cells. In addition, SOX9 could transcriptional inhibit DKK1 and activate FZD10 and MYC by binding to their promoters to affect the Wnt/β-catenin pathway. These results demonstrated SOX9 regulated the self-renewal and tumorigenicity of cervical cancer through Wnt/β-catenin pathway by directly transcriptional activation of FZD10, MYC and transcriptional inhibition of DKK1.
Collapse
Affiliation(s)
- Qian Feng
- Department of Reproductive Medicine, The First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Nan Cui
- Department of Reproductive Medicine, The First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Shan Li
- Department of Reproductive Medicine, The First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Jing Cao
- Department of Reproductive Medicine, The First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Qian Chen
- Department of Reproductive Medicine, The First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Haiyan Wang
- Department of Reproductive Medicine, The First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
3
|
Wang J, Yu H, Dong W, Zhang C, Hu M, Ma W, Jiang X, Li H, Yang P, Xiang D. N6-Methyladenosine-Mediated Up-Regulation of FZD10 Regulates Liver Cancer Stem Cells' Properties and Lenvatinib Resistance Through WNT/β-Catenin and Hippo Signaling Pathways. Gastroenterology 2023; 164:990-1005. [PMID: 36764493 DOI: 10.1053/j.gastro.2023.01.041] [Citation(s) in RCA: 88] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide, but there is a deficiency of early diagnosis biomarkers and therapeutic targets. Drug resistance accounts for most HCC-related deaths, yet the mechanisms underlying drug resistance remain poorly understood. METHODS Expression of Frizzled-10 (FZD10) in liver cancer stem cells (CSCs) was identified by means of RNA sequencing and validated by means of real-time polymerase chain reaction and immunohistochemistry. In vitro and in vivo experiments were used to assess the effect of FZD10 on liver CSC expansion and lenvatinib resistance. RNA sequencing, RNA binding protein immunoprecipitation, and luciferase report assays were applied to explore the mechanism underlying FZD10-mediated liver CSCs expansion and lenvatinib resistance. RESULTS Activation of FZD10 in liver CSCs was mediated by METTL3-dependent N6-methyladenosine methylation of FZD10 messenger RNA. Functional studies revealed that FZD10 promotes self-renewal, tumorigenicity, and metastasis of liver CSCs via activating β-catenin and YAP1. The FZD10-β-catenin/YAP1 axis is activated in liver CSCs and predicts poor prognosis. Moreover, FZD10-β-catenin/c-Jun axis transcriptionally activates METTL3 expression, forming a positive feedback loop. Importantly, the FZD10/β-catenin/c-Jun/MEK/ERK axis determines the responses of hepatoma cells to lenvatinib treatment. Analysis of patient cohort, patient-derived tumor organoids, and patient-derived xenografts further suggest that FZD10 might predict lenvatinib clinical benefit in patients with HCC. Furthermore, treatment of lenvatinib-resistant HCC with adeno-associated virus targeting FZD10 or a β-catenin inhibitor restored lenvatinib response. CONCLUSIONS Elevated FZD10 expression promotes expansion of liver CSCs and lenvatinib resistance, indicating that FZD10 expression is a novel prognostic biomarker and therapeutic target for human HCC.
Collapse
Affiliation(s)
- Jinghan Wang
- Department of Hepatobiliary Surgery, East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongming Yu
- Department of Hepatic Surgery, Third Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Wei Dong
- Department of Pathology, Third Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Cheng Zhang
- Department of Gastroenterology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Mingtai Hu
- Department of Hepatobiliary Surgery, East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wencong Ma
- Department of Hepatobiliary Surgery, East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoqing Jiang
- Department of Hepatic Surgery, Third Affiliated Hospital of Naval Military Medical University, Shanghai, China.
| | - Hengyu Li
- Department of Breast and Thyroid Surgery, Changhai Hospital, Naval Military Medical University, Shanghai, China.
| | - Pinghua Yang
- Department of Hepatic Surgery, Third Affiliated Hospital of Naval Military Medical University, Shanghai, China.
| | - Daimin Xiang
- Department of Hepatobiliary Surgery, East Hospital, School of Medicine, Tongji University, Shanghai, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Tulalamba W, Ngernsombat C, Larbcharoensub N, Janvilisri T. Transcriptomic profiling revealed FZD10 as a novel biomarker for nasopharyngeal carcinoma recurrence. Front Oncol 2023; 12:1084713. [PMID: 36776376 PMCID: PMC9909960 DOI: 10.3389/fonc.2022.1084713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a type of cancers that develops in the nasopharynx, the very upper part of the throat behind the nose. NPC is typically diagnosed in later stages of the disease and has a high rate of recurrence due to the location of the tumor growth site. In this study, we compared the gene expression profiles of NPC tissues from patients with and without recurrence to identify potential molecular biomarkers of NPC recurrence. Methods Microarrays were used to analyze the expression of genes in 15 NPC tissues taken at the time of diagnosis and at the site of recurrence following therapeutic treatment. Pathway enrichment analysis was used to examine the biological interactions between the major differentially expressed genes. The target identified was then validated using immunohistochemistry on 86 NPC tissue samples. Results Our data showed that the Wnt signaling pathway was enhanced in NPC tissues with recurrence. FZD10, a component of the Wnt signaling pathway, was significantly expressed in NPC tissues, and was significantly associated with NPC recurrence. Conclusion Our study provides new insights into the pathogenesis of NPC and identifies FZD10 as a potential molecular biomarker for NPC recurrence. FZD10 may be a promising candidate for NPC recurrence and a potential therapeutic target.
Collapse
Affiliation(s)
- Warut Tulalamba
- Siriraj Center of Research Excellence in Advanced Gene and Cell Therapy (Si-CORE-AGCT) and Thalassemia Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand,Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Chawalit Ngernsombat
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Noppadol Larbcharoensub
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand,*Correspondence: Tavan Janvilisri,
| |
Collapse
|
5
|
Hua X, Xiang D, Guo M, Qian X, Chen R, Li T, Tian Z, Xu J, Huang C, Xie Q, Huang C. Induction of RAC1 protein translation and MKK7/JNK-dependent autophagy through dicer/miR-145/SOX2/miR-365a axis contributes to isorhapontigenin (ISO) inhibition of human bladder cancer invasion. Cell Death Dis 2022; 13:753. [PMID: 36045117 PMCID: PMC9433410 DOI: 10.1038/s41419-022-05205-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 01/21/2023]
Abstract
Although our previous studies have identified that isorhapontigenin (ISO) is able to initiate autophagy in human bladder cancer (BC) cells by activating JNK/C-Jun/SESN2 axis and possesses an inhibitory effect on BC cell growth, association of autophagy directly with inhibition of BC invasion has never been explored. Also, upstream cascade responsible for ISO activating JNK remains unknown. Thus, we explored both important questions in the current study and discovered that ISO treatment initiated RAC1 protein translation, and its downstream kinase MKK7/JNK phosphorylation/activation, and in turn promoted autophagic responses in human BC cells. Inhibition of autophagy abolished ISO inhibition of BC invasion, revealing that autophagy inhibition was crucial for ISO inhibition of BC invasion. Consistently, knockout of RAC1 also attenuated induction of autophagy and inhibition of BC invasion by ISO treatment. Mechanistic studies showed that upregulation of RAC1 translation was due to ISO inhibition of miR-365a transcription, which reduced miR-365a binding to the 3'-UTR of RAC1 mRNA. Further study indicated that inhibition of miR-365a transcription was caused by downregulation of its transcription factor SOX2, while ISO-promoted Dicer protein translation increased miR-145 maturation, and consequently downregulating SOX2 expression. These findings not only provide a novel insight into the understanding association of autophagy induction with BC invasion inhibition by ISO, but also identify an upstream regulatory cascade, Dicer/miR145/SOX2/miR365a/RAC1, leading to MKK7/JNKs activation and autophagy induction.
Collapse
Affiliation(s)
- Xiaohui Hua
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China ,grid.186775.a0000 0000 9490 772XDepartment of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032 China
| | - Daimin Xiang
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Mengxin Guo
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Xiaohui Qian
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Ruifan Chen
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Tengda Li
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Zhongxian Tian
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Jiheng Xu
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Chao Huang
- grid.33199.310000 0004 0368 7223Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Qipeng Xie
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Chuanshu Huang
- grid.268099.c0000 0001 0348 3990Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| |
Collapse
|
6
|
Kalaimani L, Devarajan B, Namperumalsamy VP, Veerappan M, Daniels JT, Chidambaranathan GP. Hsa-miR-143-3p inhibits Wnt-β-catenin and MAPK signaling in human corneal epithelial stem cells. Sci Rep 2022; 12:11432. [PMID: 35794158 PMCID: PMC9259643 DOI: 10.1038/s41598-022-15263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
Our previous study demonstrated hsa-miR-143-3p as one of the highly expressed miRNAs in enriched corneal epithelial stem cells (CESCs). Hence this study aims to elucidate the regulatory role of hsa-miR-143-3p in the maintenance of stemness in CESCs. The target genes of hsa-miR-143-3p were predicted and subjected to pathway analysis to select the targets for functional studies. Primary cultured limbal epithelial cells were transfected with hsa-miR-143-3p mimic, inhibitor or scrambled sequence using Lipofectamine 3000. The transfected cells were analysed for (i) colony forming potential, (ii) expression of stem cell (SC) markers/ transcription factors (ABCG2, NANOG, OCT4, KLF4, ΔNp63), (iii) differentiation marker (Cx43), (iv) predicted five targets of hsa-miR-143-3p (DVL3, MAPK1, MAPK14, KRAS and KAT6A), (v) MAPK signaling regulators and (vi) Wnt-β-catenin signaling regulators by qPCR, immunofluorescence staining and/or Western blotting. High expression of hsa-miR-143-3p increased the colony forming potential (10.04 ± 1.35%, p < 0.001) with the ability to form holoclone-like colonies in comparison to control (3.33 ± 0.71%). The mimic treated cells had increased expression of SC markers but reduced expression of Cx43 and hsa-miR-143-3p targets involved in Wnt-β-catenin and MAPK signaling pathways. The expression of β-catenin, active β-catenin and ERK2 in hsa-miR-143-3p inhibitor transfected cells were higher than the control cells and the localized nuclear expression indicated the activation of Wnt and MAPK signaling. Thus, the probable association of hsa-miR-143-3p in the maintenance of CESCs through inhibition of Wnt and MAPK signaling pathways was thus indicated.
Collapse
Affiliation(s)
- Lavanya Kalaimani
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India.,Department of Biotechnology, Aravind Medical Research Foundation-Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India.,Institute of Ophthalmology, University College London, London, UK
| | - Bharanidharan Devarajan
- Department of Bioinformatics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | | | - Muthukkaruppan Veerappan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India
| | - Julie T Daniels
- Institute of Ophthalmology, University College London, London, UK
| | - Gowri Priya Chidambaranathan
- Department of Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, 625020, India. .,Department of Biotechnology, Aravind Medical Research Foundation-Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
7
|
Wijesena N, Sun H, Kumburegama S, Wikramanayake AH. Distinct Frizzled receptors independently mediate endomesoderm specification and primary archenteron invagination during gastrulation in Nematostella. Dev Biol 2021; 481:215-225. [PMID: 34767794 DOI: 10.1016/j.ydbio.2021.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 11/03/2022]
Abstract
Endomesodermal cell fate specification and archenteron formation during gastrulation are tightly linked developmental processes in most metazoans. However, studies have shown that in the anthozoan cnidarian Nematostella vectensis, Wnt/β-catenin (cWnt) signalling-mediated endomesodermal cell fate specification can be experimentally uncoupled from Wnt/Planar Cell Polarity (PCP) signalling-mediated primary archenteron invagination. The upstream signalling mechanisms regulating cWnt signalling-dependent endomesoderm cell fate specification and Wnt/PCP signalling-mediated primary archenteron invagination in Nematostella embryos are not well understood. By screening for potential upstream mediators of cWnt and Wnt/PCP signalling, we identified two Nematostella Frizzled homologs that are expressed early in development. NvFzd1 is expressed maternally and in a broad pattern during early development while NvFzd10 is zygotically expressed at the animal pole in blastula stage embryos and is restricted to the invaginating cells of the presumptive endomesoderm. Molecular and morphological characterization of NvFzd1 and NvFzd10 knock-down phenotypes provide evidence for distinct regulatory roles for the two receptors in endomesoderm cell fate specification and primary archenteron invagination. These results provide further experimental evidence for the independent regulation of endomesodermal cell fate specification and primary archenteron invagination during gastrulation in Nematostella. Moreover, these results provide additional support for the previously proposed two-step model for the independent evolution of cWnt-mediated cell fate specification and Wnt/PCP-mediated primary archenteron invagination.
Collapse
Affiliation(s)
- Naveen Wijesena
- Department of Biology, University of Miami, Coral Gables, FL33146, USA; Department of Biology, University of Bergen, Bergen, Norway
| | - Hongyan Sun
- Department of Biology, University of Miami, Coral Gables, FL33146, USA
| | - Shalika Kumburegama
- Department of Biology, University of Miami, Coral Gables, FL33146, USA; Department of Zoology, University of Peradeniya, Peradeniya, Sri Lanka
| | | |
Collapse
|
8
|
WNT/β-Catenin Pathway in Soft Tissue Sarcomas: New Therapeutic Opportunities? Cancers (Basel) 2021; 13:cancers13215521. [PMID: 34771683 PMCID: PMC8583315 DOI: 10.3390/cancers13215521] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The WNT/β-catenin signaling pathway is involved in fundamental processes for the proliferation and differentiation of mesenchymal stem cells. However, little is known about its relevance for mesenchymal neoplasms, such us soft tissue sarcomas (STS). Chemotherapy based on doxorubicin (DXR) still remains the standard first-line treatment for locally advanced unresectable or metastatic STS, although overall survival could not be improved by combination with other chemotherapeutics. In this sense, the development of new therapeutic approaches continues to be an unmatched goal. This review covers the most important molecular alterations of the WNT signaling pathway in STS, broadening the current knowledge about STS as well as identifying novel drug targets. Furthermore, the current therapeutic options and drug candidates to modulate WNT signaling, which are usually classified by their interaction site upstream or downstream of β-catenin, and their presumable clinical impact on STS are discussed. Abstract Soft tissue sarcomas (STS) are a very heterogeneous group of rare tumors, comprising more than 50 different histological subtypes that originate from mesenchymal tissue. Despite their heterogeneity, chemotherapy based on doxorubicin (DXR) has been in use for forty years now and remains the standard first-line treatment for locally advanced unresectable or metastatic STS, although overall survival could not be improved by combination with other chemotherapeutics. In this sense, the development of new therapeutic approaches continues to be a largely unmatched goal. The WNT/β-catenin signaling pathway is involved in various fundamental processes for embryogenic development, including the proliferation and differentiation of mesenchymal stem cells. Although the role of this pathway has been widely researched in neoplasms of epithelial origin, little is known about its relevance for mesenchymal neoplasms. This review covers the most important molecular alterations of the WNT signaling pathway in STS. The detection of these alterations and the understanding of their functional consequences for those pathways controlling sarcomagenesis development and progression are crucial to broaden the current knowledge about STS as well as to identify novel drug targets. In this regard, the current therapeutic options and drug candidates to modulate WNT signaling, which are usually classified by their interaction site upstream or downstream of β-catenin, and their presumable clinical impact on STS are also discussed.
Collapse
|
9
|
Scavo MP, Rizzi F, Depalo N, Armentano R, Coletta S, Serino G, Fanizza E, Pesole PL, Cervellera A, Carella N, Curri ML, Giannelli G. Exosome Released FZD10 Increases Ki-67 Expression via Phospho-ERK1/2 in Colorectal and Gastric Cancer. Front Oncol 2021; 11:730093. [PMID: 34671555 PMCID: PMC8522497 DOI: 10.3389/fonc.2021.730093] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/03/2021] [Indexed: 12/18/2022] Open
Abstract
Frizzled (FZD) proteins are primary receptors for Wnt signaling that activates the mitogen-activated protein kinase (MAPK) pathways. Dysfunction of Wnt signals with consequently abnormal activation of MAPK3 pathways was found in colorectal cancer (CRC) and gastric cancer (GC). Upregulation of FZD10 protein, localized in the exosomes isolated from plasma of CRC and GC patients, was associated with a poor prognosis. Herein, the expression levels of circulating FZD10 were found to be strongly correlated to their expression levels in the corresponding tissues in CRC and GC patients. Bioinformatic prediction revealed a link between FZD10 and Ki-67 through MAPK3. In both CRC and GC tissues, pERK1/2 levels were significantly increased at more advanced disease stages, and pERK1/2 and Ki-67 were correlated. Silencing of FZD10 in CRC and GC cells resulted in a significant reduction of pERK1/2 and Ki-67 expression, while subsequent treatment with exogenous exosomes partially restored their expression levels. The strong correlation between the expression of Ki-67 in tissues and of FZD10 in exosomes suggests that the exosome-delivered FZD10 may be a promising novel prognostic and diagnostic biomarker for CRC and GC.
Collapse
Affiliation(s)
- Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. De Bellis” Research Hospital, Bari, Italy
| | - Federica Rizzi
- University of Bari “A. Moro,” Chemistry Department, Bari, Italy
- Institute for Chemical–Physical Processes (IPCF)–National Research Council Secondary Site (CNR SS) Bari, Bari, Italy
| | - Nicoletta Depalo
- Institute for Chemical–Physical Processes (IPCF)–National Research Council Secondary Site (CNR SS) Bari, Bari, Italy
| | - Raffaele Armentano
- Department of Pathology, National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Bari, Italy
| | - Sergio Coletta
- Department of Pathology, National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Bari, Italy
| | - Grazia Serino
- Experimental Immunopathology Laboratory, National Institute of Gastroenterology “S. de Bellis,” Research Hospital, Bari, Italy
| | - Elisabetta Fanizza
- University of Bari “A. Moro,” Chemistry Department, Bari, Italy
- Institute for Chemical–Physical Processes (IPCF)–National Research Council Secondary Site (CNR SS) Bari, Bari, Italy
| | - Pasqua Letizia Pesole
- Laboratory of Clinical Pathology, National Institute of Gastroenterology, “S de Bellis” Research Hospital, Bari, Italy
| | - Alessandra Cervellera
- Laboratory of Clinical Pathology, National Institute of Gastroenterology, “S de Bellis” Research Hospital, Bari, Italy
| | - Nicola Carella
- Laboratory of Clinical Pathology, National Institute of Gastroenterology, “S de Bellis” Research Hospital, Bari, Italy
| | - Maria Lucia Curri
- University of Bari “A. Moro,” Chemistry Department, Bari, Italy
- Institute for Chemical–Physical Processes (IPCF)–National Research Council Secondary Site (CNR SS) Bari, Bari, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology “S. De Bellis” Research Hospital, Bari, Italy
| |
Collapse
|
10
|
Sompel K, Elango A, Smith AJ, Tennis MA. Cancer chemoprevention through Frizzled receptors and EMT. Discov Oncol 2021; 12:32. [PMID: 34604862 PMCID: PMC8429367 DOI: 10.1007/s12672-021-00429-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Frizzled (FZD) transmembrane receptors are well known for their role in β-catenin signaling and development and now understanding of their role in the context of cancer is growing. FZDs are often associated with the process of epithelial to mesenchymal transition (EMT) through β-catenin, but some also influence EMT through non-canonical pathways. With ten different FZDs, there is a wide range of activity from oncogenic to tumor suppressive depending on the tissue context. Alterations in FZD signaling can occur during development of premalignant lesions, supporting their potential as targets of chemoprevention agents. Agonizing or antagonizing FZD activity may affect EMT, which is a key process in lesion progression often targeted by chemoprevention agents. Recent studies identified a specific FZD as important for activity of an EMT inhibiting chemopreventive agent and other studies have highlighted the previously unrecognized potential for targeting small molecules to FZD receptors. This work demonstrates the value of investigating FZDs in chemoprevention and here we provide a review of FZDs in cancer EMT and their potential as chemoprevention targets.
Collapse
Affiliation(s)
- K. Sompel
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - A. Elango
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - A. J. Smith
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| | - M. A. Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12700 E 19th AVE, RC2 Box C272, Aurora, CO 80045 USA
| |
Collapse
|
11
|
Li XS, Nie KC, Zheng ZH, Zhou RS, Huang YS, Ye ZJ, He F, Tang Y. Molecular subtypes based on DNA methylation predict prognosis in lung squamous cell carcinoma. BMC Cancer 2021; 21:96. [PMID: 33485313 PMCID: PMC7825161 DOI: 10.1186/s12885-021-07807-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/12/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Due to tumor heterogeneity, the diagnosis, treatment, and prognosis of patients with lung squamous cell carcinoma (LUSC) are difficult. DNA methylation is an important regulator of gene expression, which may help the diagnosis and therapy of patients with LUSC. METHODS In this study, we collected the clinical information of LUSC patients in the Cancer Genome Atlas (TCGA) database and the relevant methylated sequences of the University of California Santa Cruz (UCSC) database to construct methylated subtypes and performed prognostic analysis. RESULTS Nine hundred sixty-five potential independent prognosis methylation sites were finally identified and the genes were identified. Based on consensus clustering analysis, seven subtypes were identified by using 965 CpG sites and corresponding survival curves were plotted. The prognostic analysis model was constructed according to the methylation sites' information of the subtype with the best prognosis. Internal and external verifications were used to evaluate the prediction model. CONCLUSIONS Models based on differences in DNA methylation levels may help to classify the molecular subtypes of LUSC patients, and provide more individualized treatment recommendations and prognostic assessments for different clinical subtypes. GNAS, FZD2, FZD10 are the core three genes that may be related to the prognosis of LUSC patients.
Collapse
Affiliation(s)
- Xiu-Shen Li
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Ke-Chao Nie
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Zhi-Hua Zheng
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Rui-Sheng Zhou
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Yu-Sheng Huang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Zeng-Jie Ye
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Fan He
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| | - Ying Tang
- Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, Guangdong China
| |
Collapse
|
12
|
Wang J, Feng D, Gao B. An Overview of Potential Therapeutic Agents Targeting WNT/PCP Signaling. Handb Exp Pharmacol 2021; 269:175-213. [PMID: 34463852 DOI: 10.1007/164_2021_533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Since the discovery of the proto-oncogene Wnt1 (Int1) in 1982, WNT signaling has been identified as one of the most important pathways that regulates a wide range of fundamental developmental and physiological processes in multicellular organisms. The canonical WNT signaling pathway depends on the stabilization and translocation of β-catenin and plays important roles in development and homeostasis. The WNT/planar cell polarity (WNT/PCP) signaling, also known as one of the β-catenin-independent WNT pathways, conveys directional information to coordinate polarized cell behaviors. Similar to WNT/β-catenin signaling, disruption or aberrant activation of WNT/PCP signaling also underlies a variety of developmental defects and cancers. However, the pharmacological targeting of WNT/PCP signaling for therapeutic purposes remains largely unexplored. In this review, we briefly discuss WNT/PCP signaling in development and disease and summarize the known drugs/inhibitors targeting this pathway.
Collapse
Affiliation(s)
- Jin Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Di Feng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
13
|
Synovial Sarcoma: A Complex Disease with Multifaceted Signaling and Epigenetic Landscapes. Curr Oncol Rep 2020; 22:124. [PMID: 33025259 DOI: 10.1007/s11912-020-00985-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Aside from a characteristic SS18-SSX translocation identified in almost all cases, no genetic anomalies have been reliably isolated yet to drive the pathogenesis of synovial sarcoma. In the following review, we explore the structural units of wild-type SS18 and SSX, particularly as they relate to the transcriptional alterations and cellular pathway changes imposed by SS18-SSX. RECENT FINDINGS Native SS18 and SSX contribute recognizable domains to the SS18-SSX chimeric proteins, which inflict transcriptional and epigenetic changes through selective protein interactions involving the SWI/SNF and Polycomb chromatin remodeling complexes. Multiple oncogenic and developmental pathways become altered, collectively reprogramming the cellular origin of synovial sarcoma and promoting its malignant transformation. Synovial sarcoma is characterized by complex epigenetic and signaling landscapes. Identifying the operational pathways and concomitant genetic changes induced by SS18-SSX fusions could help develop tailored therapeutic strategies to ultimately improve disease control and patient survivorship.
Collapse
|
14
|
Li L, Li H, Wang L, Wu S, Lv L, Tahir A, Xiao X, Wong CKC, Sun F, Ge R, Cheng CY. Role of cell polarity and planar cell polarity (PCP) proteins in spermatogenesis. Crit Rev Biochem Mol Biol 2020; 55:71-87. [PMID: 32207344 DOI: 10.1080/10409238.2020.1742091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Studies on cell polarity proteins and planar cell polarity (PCP) proteins date back to almost 40 years ago in Drosophila and C. elegans when these proteins were shown to be crucial to support apico-basal polarity and also directional alignment of polarity cells across the plane of an epithelium during morphogenesis. In adult mammals, cell polarity and PCP are most notable in cochlear hair cells. However, the role of these two groups of proteins to support spermatogenesis was not explored until a decade earlier when several proteins that confer cell polarity and PCP proteins were identified in the rat testis. Since then, there are several reports appearing in the literature to examine the role of both cell polarity and PCP in supporting spermatogenesis. Herein, we provide an overview regarding the role of cell polarity and PCP proteins in the testis, evaluating these findings in light of studies in other mammalian epithelial cells/tissues. Our goal is to provide a timely evaluation of these findings, and provide some thought provoking remarks to guide future studies based on an evolving concept in the field.
Collapse
Affiliation(s)
- Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, New York, USA
| | - Huitao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, New York, USA
| | - Lingling Wang
- Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, New York, USA
| | - Siwen Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, New York, USA
| | - Lixiu Lv
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Anam Tahir
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiang Xiao
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Chris K C Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Hong Kong, China
| | - Fei Sun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Center for Biomedical Research, The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, New York, USA
| |
Collapse
|
15
|
Scavo MP, Depalo N, Rizzi F, Ingrosso C, Fanizza E, Chieti A, Messa C, Denora N, Laquintana V, Striccoli M, Curri ML, Giannelli G. FZD10 Carried by Exosomes Sustains Cancer Cell Proliferation. Cells 2019; 8:E777. [PMID: 31349740 PMCID: PMC6721576 DOI: 10.3390/cells8080777] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/21/2019] [Accepted: 07/23/2019] [Indexed: 01/10/2023] Open
Abstract
Extracellular vesicles (EVs) are involved in intercellular communication during carcinogenesis, and cancer cells are able to secrete EVs, in particular exosomes containing molecules, that can be transferred to recipient cells to induce pathological processes and significant modifications, as metastasis, increase of proliferation, and carcinogenesis evolution. FZD proteins, a family of receptors comprised in the Wnt signaling pathway, play an important role in carcinogenesis of the gastroenteric tract. Here, a still unknown role of Frizzled 10 (FZD10) protein was identified. In particular, the presence of FZD10 and FZD10-mRNA in exosomes extracted from culture medium of the untreated colorectal, gastric, hepatic, and cholangio cancer cell lines, was detected. A substantial reduction in the FZD10 and FZD10-mRNA level was achieved in FZD10-mRNA silenced cells and in their corresponding exosomes. Concomitantly, a significant decrease in viability of the silenced cells compared to their respective controls was observed. Notably, the incubation of silenced cells with the exosomes extracted from culture medium of the same untreated cells promoted the restoration of the cell viability and, also, of the FZD10 and FZD10-mRNA level, thus indicating that the FZD10 and FZD10-mRNA delivering exosomes may be potential messengers of cancer reactivation and play an active role in long-distance metastatization.
Collapse
Affiliation(s)
- Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. De Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy.
| | - Nicoletta Depalo
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Federica Rizzi
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Chiara Ingrosso
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Elisabetta Fanizza
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Annarita Chieti
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. De Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Caterina Messa
- Laboratory of Clinical Biochemistry, National Institute of Gastroenterology "S. De Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Nunzio Denora
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
- Dipartimento di Farmacia, Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Valentino Laquintana
- Dipartimento di Farmacia, Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Marinella Striccoli
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Maria Lucia Curri
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, Via Orabona 4, 70126 Bari, Italy
| | - Gianluigi Giannelli
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. De Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy.
- National Institute of Gastroenterology "S. De Bellis", Scientific Direction, Via Turi 27, Castellana Grotte 70013 Bari, Italy.
| |
Collapse
|
16
|
Huo R, Fu W, Li H, Jiao Y, Yan Z, Wang L, Wang J, Wang S, Cao Y, Zhao J. RNA Sequencing Reveals the Activation of Wnt Signaling in Low Flow Rate Brain Arteriovenous Malformations. J Am Heart Assoc 2019; 8:e012746. [PMID: 31170876 PMCID: PMC6645621 DOI: 10.1161/jaha.119.012746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background The blood flow rate of brain arteriovenous malformations (bAVMs) is an important clinical characteristic closely associated with the hemorrhage risk and radiosurgery obliteration rate of bAVMs. However, the underlying molecular properties remain unclear. To identify potential key molecules, signaling pathways, and vascular cell types involved, we compared gene expression profiles between bAVMs with high flow rates and low flow rates (LFR) and validated the functions of selected key molecules in vitro. Methods and Results We performed RNA‐sequencing analysis on 51 samples, including 14 high flow rate bAVMs and 37 LFR bAVMs. Functional pathway analysis was performed to identify potential signals influencing the flow rate phenotype of bAVMs. Candidate genes were investigated in bAVM specimens by immunohistochemical staining. Migration, tube formation, and proliferation assays were used to test the effects of candidate genes on the phenotypic properties of cultured human umbilical vein endothelial cells and human brain vascular smooth muscle cells. We identified 250 upregulated and 118 downregulated genes in LFR bAVMs compared with high flow rate bAVMs. Wnt signaling was activated in the LFR group via upregulation of FZD10 and MYOC. Immunohistochemical staining showed that vascular endothelial and smooth muscle cells of LFR bAVMs exhibited increased FZD10 and MYOC expression. Experimentally elevating these genes promoted human umbilical vein endothelial cells and migration and tube formation by activating canonical Wnt signaling in vitro. Conclusions Our results suggest that canonical Wnt signaling mediated by FZD10 and MYOC is activated in vascular endothelial and smooth muscle cells in LFR bAVMs.
Collapse
Affiliation(s)
- Ran Huo
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Weilun Fu
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Hao Li
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Yuming Jiao
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Zihan Yan
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Linjian Wang
- 5 Savaid Medical School University of the Chinese Academy of Sciences Beijing China
| | - Jie Wang
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Shuo Wang
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Yong Cao
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China
| | - Jizong Zhao
- 1 Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University Beijing China.,2 China National Clinical Research Center for Neurological Diseases Beijing China.,3 Center of Stroke Beijing Institute for Brain Disorders Beijing China.,4 Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease Beijing China.,5 Savaid Medical School University of the Chinese Academy of Sciences Beijing China
| |
Collapse
|
17
|
Chakravarthi BVSK, Chandrashekar DS, Hodigere Balasubramanya SA, Robinson AD, Carskadon S, Rao U, Gordetsky J, Manne U, Netto GJ, Sudarshan S, Palanisamy N, Varambally S. Wnt receptor Frizzled 8 is a target of ERG in prostate cancer. Prostate 2018; 78:1311-1320. [PMID: 30051493 DOI: 10.1002/pros.23704] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022]
Abstract
Prostate cancer (PCa) is one of the most frequently diagnosed cancers among men. Many molecular changes have been detailed during PCa progression. The gene encoding the transcription factor ERG shows recurrent rearrangement, resulting in the overexpression of ERG in the majority of prostate cancers. Overexpression of ERG plays a critical role in prostate oncogenesis and development of metastatic disease. Among the downstream effectors of ERG, Frizzled family member FZD4 has been shown to be a target of ERG. Frizzled-8 (FZD8) has been shown to be involved in PCa bone metastasis. In the present study, we show that the expression of FZD8 is directly correlated with ERG expression in PCa. Furthermore, we show that ERG directly targets and activates FZD8 by binding to its promoter. This activation is specific to ETS transcription factor ERG and not ETV1. We propose that ERG overexpression in PCa leads to induction of Frizzled family member FZD8, which is known to activate the Wnt pathway. Taken together, these findings uncover a novel mechanism for PCa metastasis, and indicate that FZD8 may represent a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Balabhadrapatruni V S K Chakravarthi
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Darshan S Chandrashekar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sai Akshaya Hodigere Balasubramanya
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Alyncia D Robinson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shannon Carskadon
- Vattikuti Urology Institute, Department of Urology, Henry Ford Health System, Detroit, Michigan
| | - Uttam Rao
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sunil Sudarshan
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Nallasivam Palanisamy
- Vattikuti Urology Institute, Department of Urology, Henry Ford Health System, Detroit, Michigan
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
18
|
Wang J, Duan Y, Meng QH, Gong R, Guo C, Zhao Y, Zhang Y. Integrated analysis of DNA methylation profiling and gene expression profiling identifies novel markers in lung cancer in Xuanwei, China. PLoS One 2018; 13:e0203155. [PMID: 30286088 PMCID: PMC6171826 DOI: 10.1371/journal.pone.0203155] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 08/15/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Aberrant DNA methylation occurs frequently in cancer. The aim of this study was to identify novel methylation markers in lung cancer in Xuanwei, China, through integrated genome-wide DNA methylation and gene expression studies. METHODS Differentially methylated regions (DMRs) and differentially expressed genes (DEGs) were detected on 10 paired lung cancer tissues and noncancerous lung tissues by methylated DNA immunoprecipitation combined with microarray (MeDIP-chip) and gene expression microarray analyses, respectively. Integrated analysis of DMRs and DEGs was performed to screen out candidate methylation-related genes. Both methylation and expression changes of the candidate genes were further validated and analyzed. RESULTS Compared with normal lung tissues, lung cancer tissues expressed a total of 6,899 DMRs, including 5,788 hypermethylated regions and 1,111 hypomethylated regions. Integrated analysis of DMRs and DEGs identified 45 tumor-specific candidate genes: 38 genes whose DMRs were hypermethylated and expression was downregulated, and 7 genes whose DMRs were hypomethylated and expression was upregulated. The methylation and expression validation results identified 4 candidate genes (STXBP6, BCL6B, FZD10, and HSPB6) that were significantly hypermethylated and downregulated in most of the tumor tissues compared with the noncancerous lung tissues. CONCLUSIONS This integrated analysis of genome-wide DNA methylation and gene expression in lung cancer in Xuanwei revealed several genes regulated by promoter methylation that have not been described in lung cancer before. These results provide new insight into the carcinogenesis of lung cancer in Xuanwei and represent promising new diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Juan Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Institute of Experimental Diagnosis, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Yong Duan
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Institute of Experimental Diagnosis, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Qing-He Meng
- Department of Laboratory Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Rong Gong
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Institute of Experimental Diagnosis, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Chong Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Institute of Experimental Diagnosis, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Ying Zhao
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Institute of Experimental Diagnosis, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Yanliang Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Institute of Experimental Diagnosis, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
- * E-mail:
| |
Collapse
|
19
|
Zhao Y, Song K, Zhang Y, Xu H, Zhang X, Wang L, Fan C, Jiang G, Wang E. TMEM17 promotes malignant progression of breast cancer via AKT/GSK3β signaling. Cancer Manag Res 2018; 10:2419-2428. [PMID: 30122991 PMCID: PMC6080873 DOI: 10.2147/cmar.s168723] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Current knowledge of TMEM17, a recently identified protein of the transmembrane (TMEM) family, is limited, especially with respect to its expression and biological functions in malignant tumors. This study analyzed TMEM17 expression in invasive breast cancer tissue and breast cell lines and its relevance to clinicopathological factors, and investigated the mechanisms underlying the biological effects of TMEM17 on breast cancer cells. Patients and methods TMEM17 protein expression was determined in 20 freshly harvested specimens (tumor and paired normal tissues) by Western blotting. Immunohistochemical analysis was performed to determine the expression and subcellular localization of TMEM17 in samples from 167 patients (mean age, 49 years) diagnosed with invasive ductal carcinoma (38 with triple-negative breast cancer; 129 with non-triple-negative breast cancer) who underwent complete resection in the First Affiliated Hospital of China Medical University between 2011 and 2013. Furthermore, TMEM17 was knocked down by small interfering RNAs in breast cancer cell lines. Results TMEM17 was found to be significantly upregulated in breast cancer tissues compared to the corresponding normal breast tissues by Western blotting (p=0.015). Immunohistochemical analysis revealed that TMEM was significantly upregulated in invasive breast cancer cells compared to adjacent normal breast duct glandular epithelial cells (10.78% vs 76.05%, p<0.001), and its expression was closely related to the patient’s T-stage (p=0.022), advanced TNM stages (p=0.007), and lymph node metastasis (p=0.012). After TMEM17 knockdown or overexpression in breast cancer cell lines, TMEM17 upregulated p-AKT, p-GSK3β, active β-catenin, and Snail, and downstream target proteins c-myc and cyclin D1, and downregulated E-cadherin, resulting in increased cancer cell proliferation, invasion, and migration. These effects were reversed by the AKT inhibitor LY294002. Conclusion Our results indicate that TMEM17 is upregulated in breast cancer tissues and can promote malignant progression of breast cancer cells by activating the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Kuiyuan Song
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Yong Zhang
- Departments of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Hongtao Xu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Xiupeng Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Liang Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Guiyang Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Enhua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| |
Collapse
|
20
|
Chen X, Shi C, Cao H, Chen L, Hou J, Xiang Z, Hu K, Han X. The hedgehog and Wnt/β-catenin system machinery mediate myofibroblast differentiation of LR-MSCs in pulmonary fibrogenesis. Cell Death Dis 2018; 9:639. [PMID: 29844390 PMCID: PMC5974360 DOI: 10.1038/s41419-018-0692-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and fatal lung disease that is characterized by enhanced changes in stem cell differentiation and fibroblast proliferation. Resident mesenchymal stem cells (LR-MSCs) can undergo phenotype conversion to myofibroblasts to augment extracellular matrix production, impairing function and contributing to pulmonary fibrosis. Hedgehog and Wnt signaling are developmental signal cascades that play an essential role in regulating embryogenesis and tissue homeostasis. Recently, it has been reported that both hedgehog and Wnt signaling play important roles in pulmonary fibrogenesis. Thus, the identification of specific target regulators may yield new strategy for pulmonary fibrosis therapies. In our work, we demonstrated the critical role of Gli1, Wnt7b, Wnt10a and Fzd10 in the process of pulmonary fibrogenesis in vitro and in vivo. Gli1 was induced in LR-MSCs following TGF-β1 treatment and fibrotic lung tissues. Inhibition of Gli1 suppressed myofibroblast differentiation of LR-MSCs and pulmonary fibrosis, and decreased the expression of Wnt7b, Wnt10a and β-catenin. Gli1 bound to and increased promoter activity of the Wnt7b and Wnt10a genes, and Wnt7b and Wnt10a were critical activators of Wnt/β-catenin signaling. It was noteworthy that Fzd10 knockdown reduced Wnt7b and Wnt10a-induced activation of Wnt/β-catenin signaling, which imply that Wnt7b and Wnt10a may be the ligands for Fzd10. Moreover, siRNA-mediated inhibition of Fzd10 prevented TGF-β1-induced myofibroblast differentiation of LR-MSCs in vitro and impaired bleomycin-induced pulmonary fibrosis. We conclude that hedgehog and Wnt/β-catenin signaling play a critical role in promoting myofibroblast differentiation of LR-MSCs and development of pulmonary fibrosis. These findings elucidate a therapeutic approach to attenuate pulmonary fibrosis through targeted inhibition of Gli1 or Fzd10.
Collapse
Affiliation(s)
- Xiang Chen
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China
| | - Chaowen Shi
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China
| | - Honghui Cao
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China
| | - Ling Chen
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China
| | - Jiwei Hou
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China
| | - Zou Xiang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Kebin Hu
- Department of Medicine, Division of Nephrology, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| |
Collapse
|
21
|
Frizzled Receptors as Potential Therapeutic Targets in Human Cancers. Int J Mol Sci 2018; 19:ijms19051543. [PMID: 29789460 PMCID: PMC5983605 DOI: 10.3390/ijms19051543] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/12/2018] [Accepted: 05/19/2018] [Indexed: 12/14/2022] Open
Abstract
Frizzled receptors (FZDs) are a family of seven-span transmembrane receptors with hallmarks of G protein-coupled receptors (GPCRs) that serve as receptors for secreted Wingless-type (WNT) ligands in the WNT signaling pathway. Functionally, FZDs play crucial roles in regulating cell polarity, embryonic development, cell proliferation, formation of neural synapses, and many other processes in developing and adult organisms. In this review, we will introduce the basic structural features and review the biological function and mechanism of FZDs in the progression of human cancers, followed by an analysis of clinical relevance and therapeutic potential of FZDs. We will focus on the development of antibody-based and small molecule inhibitor-based therapeutic strategies by targeting FZDs for human cancers.
Collapse
|
22
|
Tomar T, Alkema NG, Schreuder L, Meersma GJ, de Meyer T, van Criekinge W, Klip HG, Fiegl H, van Nieuwenhuysen E, Vergote I, Widschwendter M, Schuuring E, van der Zee AGJ, de Jong S, Wisman GBA. Methylome analysis of extreme chemoresponsive patients identifies novel markers of platinum sensitivity in high-grade serous ovarian cancer. BMC Med 2017; 15:116. [PMID: 28641578 PMCID: PMC5481993 DOI: 10.1186/s12916-017-0870-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/06/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Despite an early response to platinum-based chemotherapy in advanced stage high-grade serous ovarian cancer (HGSOC), the majority of patients will relapse with drug-resistant disease. Aberrant epigenetic alterations like DNA methylation are common in HGSOC. Differences in DNA methylation are associated with chemoresponse in these patients. The objective of this study was to identify and validate novel epigenetic markers of chemoresponse using genome-wide analysis of DNA methylation in extreme chemoresponsive HGSOC patients. METHODS Genome-wide next-generation sequencing was performed on methylation-enriched tumor DNA of two HGSOC patient groups with residual disease, extreme responders (≥18 months progression-free survival (PFS), n = 8) and non-responders (≤6 months PFS, n = 10) to platinum-based chemotherapy. DNA methylation and expression data of the same patients were integrated to create a gene list. Genes were validated on an independent cohort of extreme responders (n = 21) and non-responders (n = 31) using pyrosequencing and qRT-PCR. In silico validation was performed using publicly available DNA methylation (n = 91) and expression (n = 208) datasets of unselected advanced stage HGSOC patients. Functional validation of FZD10 on chemosensitivity was carried out in ovarian cancer cell lines using siRNA-mediated silencing. RESULTS Integrated genome-wide methylome and expression analysis identified 45 significantly differentially methylated and expressed genes between two chemoresponse groups. Four genes FZD10, FAM83A, MYO18B, and MKX were successfully validated in an external set of extreme chemoresponsive HGSOC patients. High FZD10 and MKX methylation were related with extreme responders and high FAM83A and MYO18B methylation with non-responders. In publicly available advanced stage HGSOC datasets, FZD10 and MKX methylation levels were associated with PFS. High FZD10 methylation was strongly associated with improved PFS in univariate analysis (hazard ratio (HR) = 0.43; 95% CI, 0.27-0.71; P = 0.001) and multivariate analysis (HR = 0.39; 95% CI, 0.23-0.65; P = 0.003). Consistently, low FZD10 expression was associated with improved PFS (HR = 1.36; 95% CI, 0.99-1.88; P = 0.058). FZD10 silencing caused significant sensitization towards cisplatin treatment in survival assays and apoptosis assays. CONCLUSIONS By applying genome-wide integrated methylome analysis on extreme chemoresponsive HGSOC patients, we identified novel clinically relevant, epigenetically-regulated markers of platinum-sensitivity in HGSOC patients. The clinical potential of these markers in predictive and therapeutic approaches has to be further validated in prospective studies.
Collapse
Affiliation(s)
- Tushar Tomar
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Nicolette G Alkema
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Leroy Schreuder
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Gert Jan Meersma
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Tim de Meyer
- Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Wim van Criekinge
- Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Harry G Klip
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Heidi Fiegl
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Els van Nieuwenhuysen
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Ignace Vergote
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Martin Widschwendter
- Department of Women's Cancer, UCL Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
| | - Ed Schuuring
- Department of Medical Biology and Pathology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ate G J van der Zee
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands.
| | - G Bea A Wisman
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
23
|
Zhao HY, Han Y, Wang J, Yang LH, Zheng XY, Du J, Wu GP, Wang EH. IQ-domain GTPase-activating protein 1 promotes the malignant phenotype of invasive ductal breast carcinoma via canonical Wnt pathway. Tumour Biol 2017; 39:1010428317705769. [PMID: 28618949 DOI: 10.1177/1010428317705769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
IQ-domain GTPase-activating protein 1 is a scaffolding protein with multidomain which plays a role in modulating dishevelled (Dvl) nuclear translocation in canonical Wnt pathway. However, the biological function and mechanism of IQ-domain GTPase-activating protein 1 in invasive ductal carcinoma (IDC) remain unknown. In this study, we found that IQ-domain GTPase-activating protein 1 expression was elevated in invasive ductal carcinoma, which was positively correlated with tumor grade, lymphatic metastasis, and poor prognosis. Coexpression of IQ-domain GTPase-activating protein 1 and Dvl in the nucleus and cytoplasm of invasive ductal carcinoma was significantly correlated but not in the membrane. Postoperative survival in the patients with their coexpression in the nucleus and cytoplasm was obviously lower than that without coexpression. The positive expression rates of c-myc and cyclin D1 were significantly higher in the patients with nuclear coexpression of Dvl and IQ-domain GTPase-activating protein 1 than that with cytoplasmic coexpression, correlating with poor prognosis. IQ-domain GTPase-activating protein 1 significantly enhanced cell proliferation and invasion in invasive ductal carcinoma cell lines by interacting with Dvl in cytoplasm to promote Dvl nuclear translocation so as to upregulate the expression of c-myc and cyclin D1. Collectively, our data suggest that IQ-domain GTPase-activating protein 1 may promote the malignant phenotype of invasive ductal carcinoma via canonical Wnt signaling, and it could be used as a potential prognostic biomarker for breast cancer patients.
Collapse
Affiliation(s)
- Huan-Yu Zhao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yang Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jian Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lian-He Yang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xiao-Ying Zheng
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jiang Du
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Guang-Ping Wu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - En-Hua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
24
|
Hot B, Valnohova J, Arthofer E, Simon K, Shin J, Uhlén M, Kostenis E, Mulder J, Schulte G. FZD 10-Gα 13 signalling axis points to a role of FZD 10 in CNS angiogenesis. Cell Signal 2017; 32:93-103. [PMID: 28126591 DOI: 10.1016/j.cellsig.2017.01.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/09/2017] [Accepted: 01/21/2017] [Indexed: 12/20/2022]
Abstract
Among the 10 Frizzled (FZD) isoforms belonging to the Class F of G protein-coupled receptors (GPCRs), FZD10 remains the most enigmatic. FZD10 shows homology to FZD4 and FZD9 and was previously implicated in both β-catenin-dependent and -independent signalling. In normal tissue, FZD10 levels are generally very low; however, its upregulation in synovial carcinoma has attracted some attention for therapy. Our findings identify FZD10 as a receptor interacting with and signalling through the heterotrimeric G protein Gα13 but not Gα12, Gαi1, GαoA, Gαs, or Gαq. Stimulation with the FZD agonist WNT induced the dissociation of the Gα13 protein from FZD10, and led to global Gα12/13-dependent cell changes assessed by dynamic mass redistribution measurements. Furthermore, we show that FZD10 mediates Gα12/13 activation-dependent induction of YAP/TAZ transcriptional activity. In addition, we show a distinct expression of FZD10 in embryonic CNS endothelial cells at E11.5-E14.5. Given the well-known importance of Gα13 signalling for the development of the vascular system, the selective expression of FZD10 in brain vascular endothelial cells points at a potential role of FZD10-Gα13 signalling in CNS angiogenesis.
Collapse
Affiliation(s)
- Belma Hot
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S17177 Stockholm, Sweden
| | - Jana Valnohova
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S17177 Stockholm, Sweden
| | - Elisa Arthofer
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S17177 Stockholm, Sweden; Section on Molecular Signal Transduction Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 35A Convent Drive, MSC 3752, Bethesda, MD 20892-3752, USA
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Jaekyung Shin
- Science for Life Laboratory, Department of Neuroscience, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Jan Mulder
- Science for Life Laboratory, Department of Neuroscience, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Gunnar Schulte
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S17177 Stockholm, Sweden; Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
25
|
Chen H, Cheng CY. Planar cell polarity (PCP) proteins and spermatogenesis. Semin Cell Dev Biol 2016; 59:99-109. [PMID: 27108805 PMCID: PMC5071175 DOI: 10.1016/j.semcdb.2016.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/18/2016] [Indexed: 11/24/2022]
Abstract
In adult mammalian testes, spermatogenesis is comprised of several discrete cellular events that work in tandem to support the transformation and differentiation of diploid spermatogonia to haploid spermatids in the seminiferous epithelium during the seminiferous epithelial cycle. These include: self-renewal of spermatogonial stem cells via mitosis and their transformation into differentiated spermatogonia, meiosis I/II, spermiogenesis and the release of sperms at spermiation. Studies have shown that these cellular events are under precise and coordinated controls of multiple proteins and signaling pathways. These events are also regulated by polarity proteins that are known to confer classical apico-basal (A/B) polarity in other epithelia. Furthermore, spermatid development is likely supported by planar cell polarity (PCP) proteins since polarized spermatids are aligned across the plane of seminiferous epithelium in an orderly fashion, analogous to hair cells in the cochlea of the inner ear. Thus, the maximal number of spermatids can be packed and supported by a fixed population of differentiated Sertoli cells in the limited space of the seminiferous epithelium in adult testes. In this review, we briefly summarize recent findings regarding the role of PCP proteins in the testis. This information should be helpful in future studies to better understand the role of PCP proteins in spermatogenesis.
Collapse
Affiliation(s)
- Haiqi Chen
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, United States
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, United States.
| |
Collapse
|
26
|
Wnt Signaling in Cell Motility and Invasion: Drawing Parallels between Development and Cancer. Cancers (Basel) 2016; 8:cancers8090080. [PMID: 27589803 PMCID: PMC5040982 DOI: 10.3390/cancers8090080] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/09/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022] Open
Abstract
The importance of canonical and non-canonical Wnt signal transduction cascades in embryonic development and tissue homeostasis is well recognized. The aberrant activation of these pathways in the adult leads to abnormal cellular behaviors, and tumor progression is frequently a consequence. Here we discuss recent findings and analogies between Wnt signaling in developmental processes and tumor progression, with a particular focus on cell motility and matrix invasion and highlight the roles of the ARF (ADP-Ribosylation Factor) and Rho-family small GTP-binding proteins. Wnt-regulated signal transduction from cell surface receptors, signaling endosomes and/or extracellular vesicles has the potential to profoundly influence cell movement, matrix degradation and paracrine signaling in both development and disease.
Collapse
|
27
|
Fu X, Li H, Liu C, Hu B, Li T, Wang Y. Long noncoding RNA AK126698 inhibits proliferation and migration of non-small cell lung cancer cells by targeting Frizzled-8 and suppressing Wnt/β-catenin signaling pathway. Onco Targets Ther 2016; 9:3815-27. [PMID: 27445486 PMCID: PMC4928661 DOI: 10.2147/ott.s100633] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies indicate that long noncoding RNAs (lncRNAs) play a key role in the control of cellular processes such as proliferation, metastasis, and differentiation. The lncRNA dysregulation has been identified in all types of cancer. We previously found that lncRNA AK126698 suppresses cisplatin resistance in A549 cells through the Wnt/β-catenin signaling pathway. However, the clinical significance of lncRNA AK126698 and the molecular mechanisms through which it regulates cancer cell proliferation and migration are largely unknown. Methods We examined the expression of lncRNA AK126698 in 56 non-small cell lung cancer (NSCLC) tissue samples and three NSCLC cell lines using quantitative real-time polymerase chain reaction. Gain and loss of function approaches were used to evaluate the biological function of AK126698 in NSCLC cells. The effects of lncRNA AK126698 on cell proliferation were investigated using cell counting kit-8 and 5-ethynyl-2′-deoxyuridine assays, and apoptosis was measured by flow cytometry. Protein levels of AK126698 targets were evaluated by Western blotting. Results Our results showed that lncRNA AK126698 was significantly downregulated in NSCLC tissues, compared with paired adjacent nontumor tissue samples. Furthermore, lower AK126698 expression was associated with larger tumor size and advanced tumor stage. Ectopic AK126698 expression inhibited cell proliferation and migration and induced apoptosis. Conversely, decreased AK126698 expression promoted cell proliferation and migration and inhibited cell apoptosis. Importantly, we demonstrated that Frizzled-8, a receptor of Wnt/β-catenin pathway, was a target of AK126698. Furthermore, AK126698 could inhibit the activation of Wnt/β-catenin pathway, which was demonstrated by measuring the expression levels of Axin1, β-catenin, c-myc, cyclin D1, and E-cadherin. Conclusion It was found in the study that lncRNA AK126698 inhibits the proliferation and migration of NSCLC cells by targeting Frizzled-8 to suppress the Wnt/β-catenin signaling pathway. It may provide a new target for therapeutic intervention in NSCLC.
Collapse
Affiliation(s)
- Xiao Fu
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hui Li
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Chunxiao Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Tong Li
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yang Wang
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
28
|
The fusion protein SS18-SSX1 employs core Wnt pathway transcription factors to induce a partial Wnt signature in synovial sarcoma. Sci Rep 2016; 6:22113. [PMID: 26905812 PMCID: PMC4764983 DOI: 10.1038/srep22113] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022] Open
Abstract
Expression of the SS18/SYT-SSX fusion protein is believed to underlie the pathogenesis of synovial sarcoma (SS). Recent evidence suggests that deregulation of the Wnt pathway may play an important role in SS but the mechanisms whereby SS18-SSX might affect Wnt signaling remain to be elucidated. Here, we show that SS18/SSX tightly regulates the elevated expression of the key Wnt target AXIN2 in primary SS. SS18-SSX is shown to interact with TCF/LEF, TLE and HDAC but not β-catenin in vivo and to induce Wnt target gene expression by forming a complex containing promoter-bound TCF/LEF and HDAC but lacking β-catenin. Our observations provide a tumor-specific mechanistic basis for Wnt target gene induction in SS that can occur in the absence of Wnt ligand stimulation.
Collapse
|
29
|
Chen H, Shen J, Choy E, Hornicek FJ, Duan Z. Targeting protein kinases to reverse multidrug resistance in sarcoma. Cancer Treat Rev 2015; 43:8-18. [PMID: 26827688 DOI: 10.1016/j.ctrv.2015.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/27/2015] [Accepted: 11/30/2015] [Indexed: 12/28/2022]
Abstract
Sarcomas are a group of cancers that arise from transformed cells of mesenchymal origin. They can be classified into over 50 subtypes, accounting for approximately 1% of adult and 15% of pediatric cancers. Wide surgical resection, radiotherapy, and chemotherapy are the most common treatments for the majority of sarcomas. Among these therapies, chemotherapy can palliate symptoms and prolong life for some sarcoma patients. However, sarcoma cells can have intrinsic or acquired resistance after treatment with chemotherapeutics drugs, leading to the development of multidrug resistance (MDR). MDR attenuates the efficacy of anticancer drugs and results in treatment failure for sarcomas. Therefore, overcoming MDR is an unmet need for sarcoma therapy. Certain protein kinases demonstrate aberrant expression and/or activity in sarcoma cells, which have been found to be involved in the regulation of sarcoma cell progression, such as cell cycle, apoptosis, and survival. Inhibiting these protein kinases may not only decrease the proliferation and growth of sarcoma cells, but also reverse their resistance to chemotherapeutic drugs to subsequently reduce the doses of anticancer drugs and decrease drug side-effects. The discovery of novel strategies targeting protein kinases opens a door to a new area of sarcoma research and provides insight into the mechanisms of MDR in chemotherapy. This review will focus on the recent studies in targeting protein kinase to reverse chemotherapeutic drug resistance in sarcoma.
Collapse
Affiliation(s)
- Hua Chen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, United States; Department of Emergency Surgery, ShenZhen People's Hospital, 2nd Clinical Medical College of Jinan University, No. 1017 Dongmenbei Road, Shenzhen, Guangdong Province 518020, China
| | - Jacson Shen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, United States
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, United States
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, United States
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, United States.
| |
Collapse
|
30
|
|
31
|
Cheng SL, Ramachandran B, Behrmann A, Shao JS, Mead M, Smith C, Krchma K, Bello Arredondo Y, Kovacs A, Kapoor K, Brill LM, Perera R, Williams BO, Towler DA. Vascular smooth muscle LRP6 limits arteriosclerotic calcification in diabetic LDLR-/- mice by restraining noncanonical Wnt signals. Circ Res 2015; 117:142-56. [PMID: 26034040 DOI: 10.1161/circresaha.117.306712] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/28/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Wnt signaling regulates key aspects of diabetic vascular disease. OBJECTIVE We generated SM22-Cre;LRP6(fl/fl);LDLR(-/-) mice to determine contributions of Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6) in the vascular smooth muscle lineage of male low-density lipoprotein receptor-null mice, a background susceptible to diet (high-fat diet)-induced diabetic arteriosclerosis. METHODS AND RESULTS As compared with LRP6(fl/fl);LDLR(-/-) controls, SM22-Cre;LRP6(fl/fl);LDLR(-/-) (LRP6-VKO) siblings exhibited increased aortic calcification on high-fat diet without changes in fasting glucose, lipids, or body composition. Pulse wave velocity (index of arterial stiffness) was also increased. Vascular calcification paralleled enhanced aortic osteochondrogenic programs and circulating osteopontin (OPN), a matricellular regulator of arteriosclerosis. Survey of ligands and Frizzled (Fzd) receptor profiles in LRP6-VKO revealed upregulation of canonical and noncanonical Wnts alongside Fzd10. Fzd10 stimulated noncanonical signaling and OPN promoter activity via an upstream stimulatory factor (USF)-activated cognate inhibited by LRP6. RNA interference revealed that USF1 but not USF2 supports OPN expression in LRP6-VKO vascular smooth muscle lineage, and immunoprecipitation confirmed increased USF1 association with OPN chromatin. ML141, an antagonist of cdc42/Rac1 noncanonical signaling, inhibited USF1 activation, osteochondrogenic programs, alkaline phosphatase, and vascular smooth muscle lineage calcification. Mass spectrometry identified LRP6 binding to protein arginine methyltransferase (PRMT)-1, and nuclear asymmetrical dimethylarginine modification was increased with LRP6-VKO. RNA interference demonstrated that PRMT1 inhibits OPN and TNAP, whereas PRMT4 supports expression. USF1 complexes containing the histone H3 asymmetrically dimethylated on Arg-17 signature of PRMT4 are increased with LRP6-VKO. Jmjd6, a demethylase downregulated with LRP6 deficiency, inhibits OPN and TNAP expression, USF1: histone H3 asymmetrically dimethylated on Arg-17 complex formation, and transactivation. CONCLUSIONS LRP6 restrains vascular smooth muscle lineage noncanonical signals that promote osteochondrogenic differentiation, mediated in part via USF1- and arginine methylation-dependent relays.
Collapse
Affiliation(s)
- Su-Li Cheng
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Bindu Ramachandran
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Abraham Behrmann
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Jian-Su Shao
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Megan Mead
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Carolyn Smith
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Karen Krchma
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Yoanna Bello Arredondo
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Attila Kovacs
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Kapil Kapoor
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Laurence M Brill
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Ranjan Perera
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Bart O Williams
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.)
| | - Dwight A Towler
- From the Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Cardiovascular Pathobiology, Orlando, FL (S.-L.C., B.R., A.B., M.M., C.S., Y.B.A., K.K., L.M.B., R.P., D.A.T.); MD Anderson Cancer Center, Cancer Biology, Houston, TX (J.-S.S.); Washington University, Department of Medicine, St. Louis, MO (K.K., A.K.); and Van Andel Research Institute, Department of Cancer and Cell Biology, Grand Rapids, MI (B.O.W.).
| |
Collapse
|
32
|
Zhao H, Xie C, Lin X, Zhao Y, Han Y, Fan C, Zhang X, Du J, Han Y, Han Q, Wu G, Wang E. Coexpression of IQ-domain GTPase-activating protein 1 (IQGAP1) and Dishevelled (Dvl) is correlated with poor prognosis in non-small cell lung cancer. PLoS One 2014; 9:e113713. [PMID: 25436461 PMCID: PMC4249885 DOI: 10.1371/journal.pone.0113713] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 10/27/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND IQ-domain GTPase-activating protein 1 (IQGAP1) binds to Dishevelled (Dvl) and functions as a modulator of Dvl nuclear localization in Xenopus embryos. However, the relationship between IQGAP1 and Dvl in tumor tissues is unclear. MATERIALS AND METHODS We used immunohistochemistry to assess the expressions of IQGAP1 and Dvl in a cohort of 111 non-small cell lung cancer (NSCLC) patients. Association of their localization expressions with clinicopathological factors was also analyzed. RESULTS The positive rate of IQGAP1 in primary tumors was 48.6% (54/111) for its cytoplamic expression, 9.0% (10/111) for nuclear expression and 31.5% (35/111) for membranous expression; the positive rate of Dvl was 65.8% (73/111) for cytoplamic expression, 9.9% (11/111) for nuclear expression and 10.8% (12/111) for membranous expression. Coexpression rate of IQGAP1 and Dvl was 77.8% (42/54) in the cytoplasm, 80.0% (8/10) in the nucleus and 8.6% (3/35) in the membrane. Coexpression of IQGAP1 and Dvl in the cytoplasm and nucleus were significantly correlated (P<0.05), but not in the membrane (P>0.05). The positive expression rates of cyclin D1 and c-myc were significantly higher in the group of IQGAP1 and Dvl coexpression in the nucleus than that in the cytoplasm. Coexpression rate of IQGAP1 and Dvl in the cytoplasm and nucleus was significantly higher in lymph nodal metastases (63.3%, 19/30) than in primary growths (38.3%, 31/81), correlating with poor prognosis. Five-year survival time after resection in the group with their coexpression in the cytoplasm and nucleus was significantly lower than that with no coexpression (44.705±3.355 vs 58.403±2.543 months, p<0.05). CONCLUSIONS Coexpression of IQGAP1 and Dvl in the cytoplasm and nucleus was correlated with the lymph nodal metastase and poor prognosis of NSCLC, and coexpression in nucleus might play a critical role in the activation of canonical Wnt pathway.
Collapse
Affiliation(s)
- Huanyu Zhao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chengyao Xie
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xuyong Lin
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yue Zhao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yang Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xiupeng Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jiang Du
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yong Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qiang Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Guangping Wu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Enhua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
- * E-mail:
| |
Collapse
|
33
|
BRMS1L suppresses breast cancer metastasis by inducing epigenetic silence of FZD10. Nat Commun 2014; 5:5406. [PMID: 25406648 DOI: 10.1038/ncomms6406] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 09/29/2014] [Indexed: 12/16/2022] Open
Abstract
BRMS1L (breast cancer metastasis suppressor 1 like, BRMS1-like) is a component of Sin3A-histone deacetylase (HDAC) co-repressor complex that suppresses target gene transcription. Here we show that reduced BRMS1L in breast cancer tissues is associated with metastasis and poor patient survival. Functionally, BRMS1L inhibits breast cancer cells migration and invasion by inhibiting epithelial-mesenchymal transition. These effects are mediated by epigenetic silencing of FZD10, a receptor for Wnt signalling, through HDAC1 recruitment and histone H3K9 deacetylation at the promoter. Consequently, BRMS1L-induced FZD10 silencing inhibits aberrant activation of WNT3-FZD10-β-catenin signalling. Furthermore, BRMS1L is a target of miR-106b and miR-106b upregulation leads to BRMS1L reduction in breast cancer cells. RNA interference-mediated silencing of BRMS1L expression promotes metastasis of breast cancer xenografts in immunocompromised mice, whereas ectopic BRMS1L expression inhibits metastasis. Therefore, BRMS1L provides an epigenetic regulation of Wnt signalling in breast cancer cells and acts as a breast cancer metastasis suppressor.
Collapse
|
34
|
Dijksterhuis JP, Petersen J, Schulte G. WNT/Frizzled signalling: receptor-ligand selectivity with focus on FZD-G protein signalling and its physiological relevance: IUPHAR Review 3. Br J Pharmacol 2014; 171:1195-209. [PMID: 24032637 DOI: 10.1111/bph.12364] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/22/2013] [Accepted: 08/25/2013] [Indexed: 12/14/2022] Open
Abstract
The wingless/int1 (WNT)/Frizzled (FZD) signalling pathway controls numerous cellular processes such as proliferation, differentiation, cell-fate decisions, migration and plays a crucial role during embryonic development. Nineteen mammalian WNTs can bind to 10 FZDs thereby activating different downstream pathways such as WNT/β-catenin, WNT/planar cell polarity and WNT/Ca(2+) . However, the mechanisms of signalling specification and the involvement of heterotrimeric G proteins are still unclear. Disturbances in the pathways can lead to various diseases ranging from cancer, inflammatory diseases to metabolic and neurological disorders. Due to the presence of seven-transmembrane segments, evidence for coupling between FZDs and G proteins and substantial structural differences in class A, B or C GPCRs, FZDs were grouped separately in the IUPHAR GPCR database as the class FZD within the superfamily of GPCRs. Recently, important progress has been made pointing to a direct activation of G proteins after WNT stimulation. WNT/FZD and G protein coupling remain to be fully explored, although the basic observation supporting the nature of FZDs as GPCRs is compelling. Because the involvement of different (i) WNTs; (ii) FZDs; and (iii) intracellular binding partners could selectively affect signalling specification, in this review we present the current understanding of receptor/ligand selectivity of FZDs and WNTs. We pinpoint what is known about signalling specification and the physiological relevance of these interactions with special emphasis on FZD-G protein interactions.
Collapse
Affiliation(s)
- J P Dijksterhuis
- Section of Receptor Biology and Signaling, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
35
|
Le PN, McDermott JD, Jimeno A. Targeting the Wnt pathway in human cancers: therapeutic targeting with a focus on OMP-54F28. Pharmacol Ther 2014; 146:1-11. [PMID: 25172549 DOI: 10.1016/j.pharmthera.2014.08.005] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 08/21/2014] [Indexed: 12/15/2022]
Abstract
The Wnt signaling pathways are a group of signal transduction pathways that play an important role in cell fate specification, cell proliferation and cell migration. Aberrant signaling in these pathways has been implicated in the development and progression of multiple cancers by allowing increased proliferation, angiogenesis, survival and metastasis. Activation of the Wnt pathway also contributes to the tumorigenicity of cancer stem cells (CSCs). Therefore, inhibiting this pathway has been a recent focus of cancer research with multiple targetable candidates in development. OMP-54F28 is a fusion protein that combines the cysteine-rich domain of frizzled family receptor 8 (Fzd8) with the immunoglobulin Fc domain that competes with the native Fzd8 receptor for its ligands and antagonizes Wnt signaling. Preclinical models with OMP-54F28 have shown reduced tumor growth and decreased CSC frequency as a single agent and in combination with other chemotherapeutic agents. Due to these findings, a phase 1a study is nearing completion with OMP-54F28 in advanced solid tumors and 3 phase 1b studies have been opened with OMP-54F28 in combination with standard-of-care chemotherapy backbones in ovarian, pancreatic and hepatocellular cancers. This article will review the Wnt signaling pathway, preclinical data on OMP-54F28 and other Wnt pathway inhibitors and ongoing clinical trials.
Collapse
Affiliation(s)
- Phuong N Le
- University of Colorado School of Medicine, Division of Medical Oncology, United States
| | - Jessica D McDermott
- University of Colorado School of Medicine, Division of Medical Oncology, United States
| | - Antonio Jimeno
- University of Colorado School of Medicine, Division of Medical Oncology, United States.
| |
Collapse
|
36
|
Takahashi R, Nagayama S, Furu M, Kajita Y, Jin Y, Kato T, Imoto S, Sakai Y, Toguchida J. AFAP1L1, a novel associating partner with vinculin, modulates cellular morphology and motility, and promotes the progression of colorectal cancers. Cancer Med 2014; 3:759-74. [PMID: 24723436 PMCID: PMC4303145 DOI: 10.1002/cam4.237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/17/2014] [Accepted: 03/03/2014] [Indexed: 12/18/2022] Open
Abstract
We have previously identified actin filament-associated protein 1-like 1 (AFAP1L1) as a metastasis-predicting marker for spindle cell sarcomas by gene expression profiling, and demonstrated that AFAP1L1 is involved in the cell invasion process by in vitro analyses. However, its precise molecular function has not been fully elucidated, and it remains unknown whether AFAP1L1 could be a prognostic marker and/or therapeutic target of other malignancies. In this study, we found a marked elevation of AFAP1L1 gene expression in colorectal cancer (CRC) tissues as compared to the adjacent normal mucosa. Multivariate analysis revealed that AFAP1L1 was an independent and significant factor for the recurrence of rectal cancers. Moreover, the addition of the AFAP1L1 expression level to the lymph node metastasis status provided more predictive information regarding postoperative recurrence in rectal cancers. AFAP1L1-transduced CRC cells exhibited a rounded shape, increased cell motility on planar substrates, and resistance to anoikis in vitro. AFAP1L1 localized to the ringed structure of the invadopodia, together with vinculin, and AFAP1L1 was identified as a novel associating partner of vinculin by immunoprecipitation assay. AFAP1L1-transduced cells showed accelerated tumor growth in vivo, presumably reflecting the anoikis resistance of these AFAP1L1-expressing cells. Furthermore, the local administration of a siRNA against AFAP1L1 significantly suppressed the in vivo tumor growth of xenografts, suggesting that AFAP1L1 might be a candidate therapeutic target for CRCs. These results suggest that AFAP1L1 plays a role in the progression of CRCs by modulating cell shape and motility and by inhibiting anoikis, presumably through interactions with vinculin-including protein complexes.
Collapse
Affiliation(s)
- Ryo Takahashi
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Velho S, Pinto A, Licastro D, Oliveira MJ, Sousa F, Stupka E, Seruca R. Dissecting the signaling pathways associated with the oncogenic activity of MLK3 P252H mutation. BMC Cancer 2014; 14:182. [PMID: 24628919 PMCID: PMC3995575 DOI: 10.1186/1471-2407-14-182] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 02/25/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MLK3 gene mutations were described to occur in about 20% of microsatellite unstable gastrointestinal cancers and to harbor oncogenic activity. In particular, mutation P252H, located in the kinase domain, was found to have a strong transforming potential, and to promote the growth of highly invasive tumors when subcutaneously injected in nude mice. Nevertheless, the molecular mechanism underlying the oncogenic activity of P252H mutant remained elusive. METHODS In this work, we performed Illumina Whole Genome arrays on three biological replicas of human HEK293 cells stably transfected with the wild-type MLK3, the P252H mutation and with the empty vector (Mock) in order to identify the putative signaling pathways associated with P252H mutation. RESULTS Our microarray results showed that mutant MLK3 deregulates several important colorectal cancer- associated signaling pathways such as WNT, MAPK, NOTCH, TGF-beta and p53, helping to narrow down the number of potential MLK3 targets responsible for its oncogenic effects. A more detailed analysis of the alterations affecting the WNT signaling pathway revealed a down-regulation of molecules involved in the canonical pathway, such as DVL2, LEF1, CCND1 and c-Myc, and an up-regulation of DKK, a well-known negative regulator of canonical WNT signaling, in MLK3 mutant cells. Additionally, FZD6 and FZD10 genes, known to act as negative regulators of the canonical WNT signaling cascade and as positive regulators of the planar cell polarity (PCP) pathway, a non-canonic WNT pathway, were found to be up-regulated in P252H cells. CONCLUSION The results provide an overall view of the expression profile associated with mutant MLK3, and they support the functional role of mutant MLK3 by showing a deregulation of several signaling pathways known to play important roles in the development and progression of colorectal cancer. The results also suggest that mutant MLK3 may be a novel modulator of WNT signaling, and pinpoint the activation of PCP pathway as a possible mechanism underlying the invasive potential of MLK3 mutant cells.
Collapse
Affiliation(s)
- Sérgia Velho
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto,Portugal
| | - Ana Pinto
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto,Portugal
- New Therapies Group, INEB-Institute for Biomedical Engineering, Porto, Portugal
| | | | - Maria José Oliveira
- New Therapies Group, INEB-Institute for Biomedical Engineering, Porto, Portugal
| | - Filipa Sousa
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto,Portugal
| | - Elia Stupka
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Milan, Italy
| | - Raquel Seruca
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto,Portugal
- Medical Faculty of the University of Porto, Porto, Portugal
| |
Collapse
|
38
|
Overexpression of Pygopus-2 is required for canonical Wnt activation in human lung cancer. Oncol Lett 2013; 7:233-238. [PMID: 24348855 PMCID: PMC3861596 DOI: 10.3892/ol.2013.1691] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 11/11/2013] [Indexed: 02/02/2023] Open
Abstract
Lung cancer is the most common cause of cancer-related mortality worldwide. It is necessary to improve the understanding of the molecular mechanisms involved in lung cancer in order to develop more effective therapeutics for the treatment of this fatal disease. The canonical Wnt signaling pathway has been known to be important in a number of cancer types, including lung cancer. Pygopus (Pygo) is a recently identified downstream component of the Wnt signaling pathway required for β-catenin/T-cell factor (TCF)-dependent transcription. However, the role of Pygo in lung cancer remains to be elucidated. The present study showed that Pygo2 is overexpressed in human lung cancer tissue samples and cell lines. Expression levels of Pygo2 were found to correlate with cytosolic β-catenin protein levels in the samples examined. Co-immunofluorescent staining showed that Pygo2 protein accumulated in the nuclei and colocalized with nuclear β-catenin in lung cancer cell lines expressing Pygo2. To investigate the functional importance of the Pygo2 overexpression in lung cancer, short hairpin RNA (shRNA) was used to knockdown Pygo2 mRNA in lung cancer cells expressing the gene. Pygo2 shRNA was observed to inhibit cell proliferation and decrease β-catenin/TCF-dependent transcriptional activity in vitro. Furthermore, Pygo2 shRNA significantly suppressed lung cancer xenograft models in vivo (P<0.05). These results suggested that Pygo2 is a putative therapeutic target for human lung cancer and overexpression of Pygo2 may be important for aberrant Wnt activation in lung cancer.
Collapse
|
39
|
Muñoz-Soriano V, Belacortu Y, Paricio N. Planar cell polarity signaling in collective cell movements during morphogenesis and disease. Curr Genomics 2013; 13:609-22. [PMID: 23730201 PMCID: PMC3492801 DOI: 10.2174/138920212803759721] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/14/2012] [Accepted: 09/17/2012] [Indexed: 01/01/2023] Open
Abstract
Collective and directed cell movements are crucial for diverse developmental processes in the animal kingdom, but they are also involved in wound repair and disease. During these processes groups of cells are oriented within the tissue plane, which is referred to as planar cell polarity (PCP). This requires a tight regulation that is in part conducted by the PCP pathway. Although this pathway was initially characterized in flies, subsequent studies in vertebrates revealed a set of conserved core factors but also effector molecules and signal modulators, which build the fundamental PCP machinery. The PCP pathway in Drosophila regulates several developmental processes involving collective cell movements such as border cell migration during oogenesis, ommatidial rotation during eye development, and embryonic dorsal closure. During vertebrate embryogenesis, PCP signaling also controls collective and directed cell movements including convergent extension during gastrulation, neural tube closure, neural crest cell migration, or heart morphogenesis. Similarly, PCP signaling is linked to processes such as wound repair, and cancer invasion and metastasis in adults. As a consequence, disruption of PCP signaling leads to pathological conditions. In this review, we will summarize recent findings about the role of PCP signaling in collective cell movements in flies and vertebrates. In addition, we will focus on how studies in Drosophila have been relevant to our understanding of the PCP molecular machinery and will describe several developmental defects and human disorders in which PCP signaling is compromised. Therefore, new discoveries about the contribution of this pathway to collective cell movements could provide new potential diagnostic and therapeutic targets for these disorders.
Collapse
Affiliation(s)
- Verónica Muñoz-Soriano
- Departamento de Genética, Facultad de CC Biológicas, Universidad de Valencia, Burjassot 46100, Valencia, Spain
| | | | | |
Collapse
|
40
|
Yin S, Xu L, Bonfil RD, Banerjee S, Sarkar FH, Sethi S, Reddy KB. Tumor-initiating cells and FZD8 play a major role in drug resistance in triple-negative breast cancer. Mol Cancer Ther 2013; 12:491-8. [PMID: 23445611 DOI: 10.1158/1535-7163.mct-12-1090] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Triple-negative breast cancer (TNBC) studies have shown that neoadjuvant chemotherapy before surgery was effective in the minority of women, whereas the majority who had residual tumor had a relatively poor outcome. To identify the mechanism by which residual cancer cells survive chemotherapy, we initially conducted gene expression profiling using the CRL2335 TNBC cell line derived from a squamous breast carcinoma before and after treatment with cisplatin plus TRAIL. We found a significant increase in the expression of FZD8, one of Wnt receptors, and its downstream targets LEF1 and TCF in residual CRL2335 tumor cells after treatment with cisplatin plus TRAIL. Increased FZD8 levels were further confirmed in other TNBC cell lines. Inhibition of FZD8 by siRNA in CRL2335 cells in the presence of cisplatin plus TRAIL reduced β-catenin and survivin levels and increased apoptosis compared with scrambled siRNA-treated cells. In vivo data show that cisplatin plus TRAIL treatment significantly reduces tumor volume in NOD/SCID mice. However, we found that cisplatin plus TRAIL treatment predominantly eliminated non-tumor-initiating cells, as shown by whole-body fluorescent imaging of mice injected with mammosphere-forming CRL2335 cells stably transfected with DsRed. This led to TIC enrichment in residual tumors, as confirmed by immunostaining for TIC markers. Moreover, an increase in FZD8 expression was observed in residual tumors treated with cisplatin and TRAIL. Taken together, our findings suggest that FZD8-mediated Wnt signaling may play a major role in mediating resistance to chemotherapy, making it a potential target to enhance chemotherapeutic efficacy in patients with TNBCs.
Collapse
Affiliation(s)
- Shuping Yin
- Department of Pathology, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Caverzasio J, Biver E, Thouverey C. Predominant role of PDGF receptor transactivation in Wnt3a-induced osteoblastic cell proliferation. J Bone Miner Res 2013; 28:260-70. [PMID: 22927028 DOI: 10.1002/jbmr.1748] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 08/02/2012] [Accepted: 08/20/2012] [Indexed: 12/17/2022]
Abstract
Previous studies have shown that Wnt3a enhances the proliferation and inhibits the osteogenic differentiation of human mesenchymal stem cells (hMSCs). In this study, we investigated the signaling pathways involved in Wnt3a-induced osteoblastic cell proliferation. Experiments with DKK1, a natural antagonist of Lrp5/6, indicated that Wnt/β-catenin did not play a major role in Wnt3a-induced osteoblastic cell proliferation. The use of selective inhibitors of known mitogenic pathways implicates Src family kinases (SFKs) and a protein kinase C (PKC) in this cellular response. Time-dependent analysis of signaling molecules activated by Wnt3a in MC3T3-E1 cells revealed parallel activation of the canonical pathway and of several tyrosine kinases, including SFKs and PDGF receptors (PDGF-Rs). Functional analysis with specific inhibitors suggested a major role of PDGF-Rs in mediating Wnt3a-induced cell proliferation. Further investigation with an si-RNA approach confirmed a predominant role of this receptor in this cellular response. The use of soluble decoy PDGF-Rs that can sequester extracellular PDGFs excluding that part of the increased PDGF receptor phosphorylation by Wnt3a was the result of autocrine production of PDGFs. A selective SFK inhibitor blunted the enhanced PDGF-R phosphorylation and cell proliferation induced by Wnt3a. Studies of initial events involved in the regulation of this pathway suggest a role of dishevelled. In conclusion, data presented in this study indicate that cell proliferation induced by Wnt3a in osteoblastic cells is mediated by a dishevelled-dependent and β-catenin-independent pathway, which involves the transactivation of PDGF receptors.
Collapse
Affiliation(s)
- Joseph Caverzasio
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland.
| | | | | |
Collapse
|
42
|
Nikaido M, Law EWP, Kelsh RN. A systematic survey of expression and function of zebrafish frizzled genes. PLoS One 2013; 8:e54833. [PMID: 23349976 PMCID: PMC3551900 DOI: 10.1371/journal.pone.0054833] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 12/19/2012] [Indexed: 01/07/2023] Open
Abstract
Wnt signaling is crucial for the regulation of numerous processes in development. Consistent with this, the gene families for both the ligands (Wnts) and receptors (Frizzleds) are very large. Surprisingly, while we have a reasonable understanding of the Wnt ligands likely to mediate specific Wnt-dependent processes, the corresponding receptors usually remain to be elucidated. Taking advantage of the zebrafish model's excellent genomic and genetic properties, we undertook a comprehensive analysis of the expression patterns of frizzled (fzd) genes in zebrafish. To explore their functions, we focused on testing their requirement in several developmental events known to be regulated by Wnt signaling, convergent extension movements of gastrulation, neural crest induction, and melanocyte specification. We found fourteen distinct fzd genes in the zebrafish genome. Systematic analysis of their expression patterns between 1-somite and 30 hours post-fertilization revealed complex, dynamic and overlapping expression patterns. This analysis demonstrated that only fzd3a, fzd9b, and fzd10 are expressed in the dorsal neural tube at stages corresponding to the timing of melanocyte specification. Surprisingly, however, morpholino knockdown of these, alone or in combination, gave no indication of reduction of melanocytes, suggesting the important involvement of untested fzds or another type of Wnt receptor in this process. Likewise, we found only fzd7b and fzd10 expressed at the border of the neural plate at stages appropriate for neural crest induction. However, neural crest markers were not reduced by knockdown of these receptors. Instead, these morpholino knockdown studies showed that fzd7a and fzd7b work co-operatively to regulate convergent extension movement during gastrulation. Furthermore, we show that the two fzd7 genes function together with fzd10 to regulate epiboly movements and mesoderm differentiation.
Collapse
Affiliation(s)
- Masataka Nikaido
- Department of Biology and Biochemistry, University of Bath, Claverton Down, United Kingdom
| | - Edward W. P. Law
- Department of Biology and Biochemistry, University of Bath, Claverton Down, United Kingdom
| | - Robert N. Kelsh
- Department of Biology and Biochemistry, University of Bath, Claverton Down, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Disheveled proteins promote cell growth and tumorigenicity in ALK-positive anaplastic large cell lymphoma. Cell Signal 2012; 25:295-307. [PMID: 23022960 DOI: 10.1016/j.cellsig.2012.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 09/12/2012] [Accepted: 09/24/2012] [Indexed: 01/21/2023]
Abstract
Our previous oligonucleotide array studies revealed that ALK-positive anaplastic large cell lymphoma (ALK(+)ALCL) express high levels of the disheveled proteins (Dvls), a family of proteins that is integral to the Wnt signaling pathways. In this study, we assessed whether the Dvls are important in the pathogenesis of ALK(+)ALCL. By Western blotting, Dvl-2 and Dvl-3 were found to be highly expressed in ALK(+)ALCL cell lines and patient samples. The higher molecular weight forms, consistent with phosphorylated/active Dvl proteins, were observed in these lysates. siRNA knock-down of Dvls did not affect the Wnt canonical pathway, as assessed by the β-catenin protein levels and nuclear localization. In contrast, the same treatment led to changes in the transcriptional activity of NFAT and the phosphorylation status of Src, both of which are known to be regulated by the Wnt non-canonical signaling pathways in other cell types. Coupled with these biochemical changes, there was a significant decrease in cell growth and soft agar colony formation. NPM-ALK, the oncogenic tyrosine kinase characteristic of ALK(+)ALCL, was found to bind to the Dvls and enhance their tyrosine phosphorylation. In conclusion, our data suggest that the Dvls contribute to the pathogenesis of ALK(+)ALCL via signaling in the Wnt non-canonical pathways. To our knowledge, this is the first report demonstrating a physical and functional interaction between the Dvls and an oncogenic tyrosine kinase.
Collapse
|
44
|
Ueno K, Hirata H, Hinoda Y, Dahiya R. Frizzled homolog proteins, microRNAs and Wnt signaling in cancer. Int J Cancer 2012; 132:1731-40. [PMID: 22833265 DOI: 10.1002/ijc.27746] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 07/10/2012] [Indexed: 01/12/2023]
Abstract
Wnt signaling pathways play important roles in tumorigenesis and are initiated by binding of Wnt to various receptors including frizzleds (FZDs). FZDs are one of several families of receptors comprised of FZD/LRP/ROR2/RYK in the Wnt signaling pathway. Expression of some FZD receptors are up regulated, thereby activating the Wnt signaling pathway and is correlated with cancer malignancy and patient outcomes (recurrence and survival) in many cancers. The FZD family contains ten genes in humans and their function has not been completely examined including the regulatory mechanisms of FZD genes in cancer. Knockdown of FZDs may suppress the Wnt signaling pathway resulting in decreased cell growth, invasion, motility and metastasis of cancer cells. Recently a number of microRNAs (miRNAs) have been identified and reported to be important in several cancers. MiRNAs regulate target gene expression at both the transcription and translation levels. The study of miRNA is a newly emerging field and promises to be helpful in understanding the pathogenesis of FZDs in cancer. In addition, miRNAs may be useful in regulating FZDs in cancer cells. Therefore, the aim of this review is to discuss current knowledge of the functional mechanisms of FZDs in cancer, including regulation by miRNAs and the potential for possible use of miRNAs and FZDs in future clinical applications.
Collapse
Affiliation(s)
- Koji Ueno
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California at San Francisco, San Francisco, CA 94121, USA
| | | | | | | |
Collapse
|
45
|
Caron A, Xu X, Lin X. Wnt/β-catenin signaling directly regulates Foxj1 expression and ciliogenesis in zebrafish Kupffer's vesicle. Development 2011; 139:514-24. [PMID: 22190638 DOI: 10.1242/dev.071746] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cilia are essential for normal development. The composition and assembly of cilia has been well characterized, but the signaling and transcriptional pathways that govern ciliogenesis remain poorly studied. Here, we report that Wnt/β-catenin signaling directly regulates ciliogenic transcription factor foxj1a expression and ciliogenesis in zebrafish Kupffer's vesicle (KV). We show that Wnt signaling acts temporally and KV cell-autonomously to control left-right (LR) axis determination and ciliogenesis. Specifically, reduction of Wnt signaling leads to a disruption of LR patterning, shorter and fewer cilia, a loss of cilia motility and a downregulation of foxj1a expression. However, these phenotypes can be rescued by KV-targeted overexpression of foxj1a. In comparison to the FGF pathway that has been previously implicated in the control of ciliogenesis, our epistatic studies suggest a more downstream function of Wnt signaling in the regulation of foxj1a expression and ciliogenesis in KV. Importantly, enhancer analysis reveals that KV-specific expression of foxj1a requires the presence of putative Lef1/Tcf binding sites, indicating that Wnt signaling activates foxj1a transcription directly. We also find that impaired Wnt signaling leads to kidney cysts and otolith disorganization, which can be attributed to a loss of foxj1 expression and disrupted ciliogenesis in the developing pronephric ducts and otic vesicles. Together, our data reveal a novel role of Wnt/β-catenin signaling upstream of ciliogenesis, which might be a general developmental mechanism beyond KV. Moreover, our results also prompt a hypothesis that certain developmental effects of the Wnt/β-catenin pathway are due to the activation of Foxj1 and cilia formation.
Collapse
Affiliation(s)
- Alissa Caron
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | |
Collapse
|
46
|
Wang HQ, Xu ML, Ma J, Zhang Y, Xie CH. Frizzled-8 as a putative therapeutic target in human lung cancer. Biochem Biophys Res Commun 2011; 417:62-6. [PMID: 22138402 DOI: 10.1016/j.bbrc.2011.11.055] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 11/11/2011] [Indexed: 10/15/2022]
Abstract
Lung cancer is the leading cause of cancer related deaths worldwide. It is necessary to better understand the molecular mechanisms involved in lung cancer in order to develop more effective therapeutics for the treatment of this disease. Recent reports have shown that Wnt signaling pathway is important in a number of cancer types including lung cancer. However, the role of Frizzled-8 (Fzd-8), one of the Frizzled family of receptors for the Wnt ligands, in lung cancer still remains to be elucidated. Here in this study we showed that Fzd-8 was over-expressed in human lung cancer tissue samples and cell lines. To investigate the functional importance of the Fzd-8 over-expression in lung cancer, we used shRNA to knock down Fzd-8 mRNA in lung cancer cells expressing the gene. We observed that Fzd-8 shRNA inhibited cell proliferation along with decreased activity of Wnt pathway in vitro, and also significantly suppressed A549 xenograft model in vivo (p<0.05). Furthermore, we found that knocking down Fzd-8 by shRNA sensitized the lung cancer cells to chemotherapy Taxotere. These data suggest that Fzd-8 is a putative therapeutic target for human lung cancer and over-expression of Fzd-8 may be important for aberrant Wnt activation in lung cancer.
Collapse
Affiliation(s)
- Hua-qing Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, China
| | | | | | | | | |
Collapse
|
47
|
McQueen P, Ghaffar S, Guo Y, Rubin EM, Zi X, Hoang BH. The Wnt signaling pathway: implications for therapy in osteosarcoma. Expert Rev Anticancer Ther 2011; 11:1223-32. [PMID: 21916576 DOI: 10.1586/era.11.94] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Osteosarcoma is the most common primary bone malignancy, with a high propensity for local invasion, early metastasis and relapse. While the molecular mechanisms behind osteosarcoma development and metastasis have not yet been fully elucidated, research has highlighted an important role for Wnt signaling. Several Wnt ligands, receptors and coreceptors are highly expressed in osteosarcoma cell lines, while Wnt inhibitors are downregulated. As a result, research has begun to identify mechanisms with which to inhibit Wnt signaling. The use of Wnt pathway inhibitors and the targeting of c-Met, a Wnt regulated proto-oncogene, may be two possible mechanisms for treatment of osteosarcoma. In addition, as the Wnt signaling pathway is a regulator of stem cells, reagents that function as Wnt inhibitors are currently under investigation as inhibitors of cancer stem cell proliferation. Research involving the Wnt signaling pathway and cancer stem cells holds promise for novel treatment options in the future.
Collapse
Affiliation(s)
- Peter McQueen
- Department of Orthopaedic Surgery, University of California at Irvine, Orange, CA 92868, USA
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
G protein-coupled receptors (GPCRs) belong to a superfamily of cell surface signalling proteins that have a pivotal role in many physiological functions and in multiple diseases, including the development of cancer and cancer metastasis. Current drugs that target GPCRs - many of which have excellent therapeutic benefits - are directed towards only a few GPCR members. Therefore, huge efforts are currently underway to develop new GPCR-based drugs, particularly for cancer. We review recent findings that present unexpected opportunities to interfere with major tumorigenic signals by manipulating GPCR-mediated pathways. We also discuss current data regarding novel GPCR targets that may provide promising opportunities for drug discovery in cancer prevention and treatment.
Collapse
|
49
|
Hirata H, Hinoda Y, Majid S, Chen Y, Zaman MS, Ueno K, Nakajima K, Tabatabai ZL, Ishii N, Dahiya R. DICKKOPF-4 activates the noncanonical c-Jun-NH2 kinase signaling pathway while inhibiting the Wnt-canonical pathway in human renal cell carcinoma. Cancer 2010; 117:1649-60. [PMID: 21472712 DOI: 10.1002/cncr.25666] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 08/04/2010] [Accepted: 08/06/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND To the authors' knowledge, the functional significance of the Wnt antagonist dickkopf homolog 4 (DKK4) has not been investigated previously in renal cancer. METHODS The authors initially observed that the expression of DKK4 was significantly higher in renal cancer tissues compared with adjacent normal kidney tissues. To assess the function of DKK4, stable DKK4-transfected cells were established, and functional analyses were performed, including a T-cell factor/lymphoid enhancer factor (TCF/LEF) reporter assay and tests for cell viability, colony formation, apoptosis, cell cycle, invasive capability, wound-healing capability, and in vivo tumor growth. RESULTS The relative TCF/LEF activity was significantly lower in DKK4-transfected cells compared with empty vector, and nuclear β-catenin expression was decreased in DKK4 transfectants. In addition, expression levels of the β-catenin downstream effector proteins cyclin D1 and c-Myc were decreased in DKK4 transfectants. However, greater invasiveness and migration were observed in stably transfected DKK4 cells. Increased growth of DKK4-transfected tumors also was observed in nude mice. Members of the Wnt noncanonical/c-Jun-NH2 kinase (JNK) signaling pathway also were effected, such as c-Jun, which had significantly increased expression and phosphorylation in DKK4-stable transfectants, and matrix metalloproteinase-2, which had significantly increased expression in DKK4-stable transfectants. CONCLUSIONS This is the first study to indicate that DKK4 expression is increased in renal cancer tissues and that DKK4 activates the noncanonical JNK signaling pathway while inhibiting the Wnt-canonical pathway.
Collapse
Affiliation(s)
- Hiroshi Hirata
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California at San Francisco, San Francisco, California 94121, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Su HY, Lai HC, Lin YW, Liu CY, Chen CK, Chou YC, Lin SP, Lin WC, Lee HY, Yu MH. Epigenetic silencing of SFRP5 is related to malignant phenotype and chemoresistance of ovarian cancer through Wnt signaling pathway. Int J Cancer 2010; 127:555-67. [PMID: 19957335 DOI: 10.1002/ijc.25083] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Oncogenic activation of the Wnt signaling pathway is common in cancers, but mutation of beta-catenin in ovarian cancer is rare. In addition to genetic events, epigenetic modification of secreted frizzled-related protein (SFRP) family has been shown to be important in regulating Wnt signaling. Although high degree of homology is observed in the same family, different SFRPs may have opposing effects on the same process. We reported recently that a Wnt antagonist, SFRP5, is downregulated frequently through promoter hypermethylation and that this hypermethylation is associated with overall survival in ovarian cancer. The aim of this study was to analyze the function of SFRP5 in ovarian cancer. Functional assays including measuring cell proliferation, invasion, colony formation and xenograft were performed using ovarian cancer cell lines with overexpression of SFRP5 or a short hairpin RNA silencing. The methylation status of SFRP5 in relation to cisplatin resistance in ovarian cancer patients was analyzed. Restoration of the expression of SFRP5 attenuated Wnt signaling in ovarian cancer cells and suppressed cancer cell growth, invasion of cells and tumorigenicity in mice. These effects were independent of the canonical pathway. The expression of SFRP5 inhibited epithelial-mesenchymal transition (EMT). The restoration of SFRP5 downregulated AKT2 and sensitized ovarian cancer cells to chemotherapy. These effects are consistent with the poor response to platinum-based chemotherapy in patients with methylation of SFRP5. Our data suggested that epigenetic silencing of SFRP5 leads to oncogenic activation of the Wnt pathway and contributes to ovarian cancer progression and chemoresistance through the TWIST-mediated EMT and AKT2 signaling.
Collapse
Affiliation(s)
- Her-Young Su
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|