1
|
Zhuo G, Lin S, Yuan F, Zheng Q, Guo Y, Wang Z, Hu J, Yao M, Zhong F, Chen S, Chen Y, Chen H. Comprehensive analysis of the expression and prognostic value of ARMCs in pancreatic adenocarcinoma. BMC Cancer 2025; 25:28. [PMID: 39773340 PMCID: PMC11708071 DOI: 10.1186/s12885-024-13365-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) has a very poor prognosis, and there are few treatments for PAAD. Therefore, it is important to find some biomarkers for the diagnosis and treatment of PAAD. Although some members of Armadillo repeat containing proteins (ARMCs) have been implicated in the development of certain cancers, their relationship with PAAD remains unknown. In this study, we aimed to explore the expression and prognostic value of ARMCs in PAAD. METHODS We used the The Cancer Genome Atlas (TCGA) database for survival analysis. Then, Gene Expression Profiling Interactive Analysis (GEPIA), the cBioPortal database, the Human Protein Atlas (HPA), Kaplan-Meier Plotter, LinkedOmics Database, Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG), Cytoscape and Timer were used to analyze the relationship between ARMCs and PAAD. In addition, we established a prognostic model of ARMCs for PAAD. Immunohistochemistry (IHC) was also performed. Then Image-J was used to analyze all images obtained from the experiment, and GraphPad-Prism (9.5.1) was used for statistical analysis to verify the expression of ARMCs in PAAD. RESULTS In the TCGA database, the expressions of ARMC1, 2, 3, 5, 6, 7, 8, 9 and 10 in PAAD tissues were significantly higher than those in normal tissues. And higher expressions of ARMC1 and 10 were associated with lower survival rate of PAAD patients. In addition, ARMC2, 5, 6, and 10 were positively associated with advanced stages of PAAD. ARMCs mutations occur in 11% of PAAD patients, and missense mutations and amplification of ARMCs account for most of them. In addition, ARMC5 and ARMC10 were independent prognostic factors in univariate and multivariate Cox regression analyses. Finally, through our confirmation experiment, it was found that the expression of ARMC1 and 10 in PAAD tissues was significantly increased compared with those in paracancer tissue. CONCLUSION This study suggests that ARMCs may be able to play important roles in PAAD, and they can act as biomarkers, providing valuable clues for the treatment and diagnosis of PAAD.
Collapse
Affiliation(s)
- Guanxiang Zhuo
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
- Department of Hepatobiliary Surgery, Fudan University Shanghai Cancer Center Xiamen Hospital, Xiamen, 350003, Fujian, China
| | - Shengzhai Lin
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
- Fujian Medical University Cancer Center, Fuzhou, 350001, Fujian, China
| | - Fei Yuan
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
- Fujian Medical University Cancer Center, Fuzhou, 350001, Fujian, China
| | - Qiaoling Zheng
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Yinpin Guo
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
- Fujian Medical University Cancer Center, Fuzhou, 350001, Fujian, China
| | - Zuwei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Jianfei Hu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Meihong Yao
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Fuxiu Zhong
- Department of Hepatobiliary Surgery Nursing, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, Fujian, China.
| | - Yanling Chen
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
- Fujian Medical University Cancer Center, Fuzhou, 350001, Fujian, China.
| | - Huixing Chen
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
- Fujian Medical University Cancer Center, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
2
|
Kumar H, Panigrahi M, G Strillacci M, Sonejita Nayak S, Rajawat D, Ghildiyal K, Bhushan B, Dutt T. Detection of genome-wide copy number variation in Murrah buffaloes. Anim Biotechnol 2023; 34:3783-3795. [PMID: 37381739 DOI: 10.1080/10495398.2023.2227670] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Riverine Buffaloes, especially the Murrah breed because of their adaptability to harsh climatic conditions, is farmed in many countries to convert low-quality feed into valuable dairy products and meat. Here, we investigated the copy number variations (CNVs) in 296 Murrah buffalo using the Axiom® Buffalo Genotyping Array 90K (Affymetrix, Santa Clara, CA, USA). The CNVs were detected on the autosomes, using the Copy Number Analysis Module (CNAM) using the univariate analysis. 7937 CNVs were detected in 279 Buffaloes, the average length of the CNVs was 119,048.87 bp that ranged between 7800 and 4,561,030 bp. These CNVs were accounting for 10.33% of the buffalo genome, which was comparable to cattle, sheep, and goat CNV analyses. Further, CNVs were merged and 1541 CNVRs were detected using the Bedtools-mergeBed command. 485 genes were annotated within 196 CNVRs that were identified in at least 10 animals of Murrah population. Out of these, 40 CNVRs contained 59 different genes that were associated with 69 different traits. Overall, the study identified a significant number of CNVs and CNVRs in the Murrah breed of buffalo, with a wide range of lengths and frequencies across the autosomes. The identified CNVRs contained genes associated with important traits related to production and reproduction, making them potentially important targets for future breeding and genetic improvement efforts.
Collapse
Affiliation(s)
- Harshit Kumar
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, India
| | - Manjit Panigrahi
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, India
| | - Maria G Strillacci
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | | | - Divya Rajawat
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, India
| | - Kanika Ghildiyal
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, India
| | - Bharat Bhushan
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, India
| | - Triveni Dutt
- Livestock Production and Management Section, Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
3
|
Ali W, Xiao W, Jacobs D, Kajdacsy-Balla A. Survival and Enrichment Analysis of Epithelial-Mesenchymal Transition Genes in Bladder Urothelial Carcinoma. Genes (Basel) 2023; 14:1899. [PMID: 37895248 PMCID: PMC10606556 DOI: 10.3390/genes14101899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/16/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The escalating prevalence of bladder cancer, particularly urothelial carcinoma, necessitates innovative approaches for prognosis and therapy. This study delves into the significance of genes related to epithelial-mesenchymal transition (EMT), a process inherently linked to carcinogenesis and comparatively better studied in other cancers. We examined 1184 EMT-related gene expression levels in bladder urothelial cancer cases through the TCGA dataset. Genes shown to be differentially expressed in relation to survival underwent further network and enrichment analysis to uncover how they might shape disease outcomes. Our in silico analysis revealed a subset of 32 genes, including those significantly represented in biological pathways such as VEGF signaling and bacterium response. In addition, these genes interact with genes involved in the JAK-STAT signaling pathway. Additionally, some of those 32 genes have been linked to immunomodulators such as chemokines CCL15 and CCL18, as well as to various immune cell infiltrates. Our findings highlight the prognostic utility of various EMT-related genes and identify possible modulators of their effect on survival, allowing for further targeted wet lab research and possible therapeutic intervention.
Collapse
Affiliation(s)
- Waleed Ali
- Albert Einstein College of Medicine, New York, NY 10461, USA; (W.X.); (D.J.)
| | - Weirui Xiao
- Albert Einstein College of Medicine, New York, NY 10461, USA; (W.X.); (D.J.)
| | - Daniel Jacobs
- Albert Einstein College of Medicine, New York, NY 10461, USA; (W.X.); (D.J.)
| | - Andre Kajdacsy-Balla
- Professor of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL 60607, USA;
| |
Collapse
|
4
|
van Gen Hassend PM, Pottikkadavath A, Delto C, Kuhn M, Endres M, Schönemann L, Schindelin H. RanBP9 controls the oligomeric state of CTLH complex assemblies. J Biol Chem 2023; 299:102869. [PMID: 36621627 PMCID: PMC9932110 DOI: 10.1016/j.jbc.2023.102869] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023] Open
Abstract
The CTLH (C-terminal to lissencephaly-1 homology motif) complex is a multisubunit RING E3 ligase with poorly defined substrate specificity and flexible subunit composition. Two key subunits, muskelin and Wdr26, specify two alternative CTLH complexes that differ in quaternary structure, thereby allowing the E3 ligase to presumably target different substrates. With the aid of different biophysical and biochemical techniques, we characterized CTLH complex assembly pathways, focusing not only on Wdr26 and muskelin but also on RanBP9, Twa1, and Armc8β subunits, which are critical to establish the scaffold of this E3 ligase. We demonstrate that the ability of muskelin to tetramerize and the assembly of Wdr26 into dimers define mutually exclusive oligomerization modules that compete with nanomolar affinity for RanBP9 binding. The remaining scaffolding subunits, Armc8β and Twa1, strongly interact with each other and with RanBP9, again with nanomolar affinity. Our data demonstrate that RanBP9 organizes subunit assembly and prevents higher order oligomerization of dimeric Wdr26 and the Armc8β-Twa1 heterodimer through its tight binding. Combined, our studies define alternative assembly pathways of the CTLH complex and elucidate the role of RanBP9 in governing differential oligomeric assemblies, thereby advancing our mechanistic understanding of CTLH complex architectures.
Collapse
Affiliation(s)
- Pia Maria van Gen Hassend
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany
| | - Aparna Pottikkadavath
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany
| | - Carolyn Delto
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany
| | - Monika Kuhn
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany
| | - Michelle Endres
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany
| | - Lars Schönemann
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany
| | - Hermann Schindelin
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Institute of Structural Biology, Würzburg, Germany.
| |
Collapse
|
5
|
El-Helbawy NF, El Zowalaty AE. Identification of Age-Associated Transcriptomic Changes Linked to Immunotherapy Response in Primary Melanoma. Curr Issues Mol Biol 2022; 44:4118-4131. [PMID: 36135194 PMCID: PMC9497511 DOI: 10.3390/cimb44090282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Melanoma is a lethal form of skin cancer. Immunotherapeutic agents such as anti-PD-1 (pembrolizumab and nivolumab) and anti-CTLA-4 (ipilimumab) have revolutionized melanoma treatment; however, drug resistance is rapidly acquired. Several studies have reported an increase in melanoma rates in older patients. Thus, the impact of ageing on transcriptional profiles of melanoma and response to immunotherapy is essential to understand. In this study, the bioinformatic analysis of RNA seq data of old and young melanoma patients receiving immunotherapy identifies the significant upregulation of extra-cellular matrix and cellular adhesion genes in young cohorts, while genes involved in cell proliferation, inflammation, non-canonical Wnt signaling and tyrosine kinase receptor ROR2 are significantly upregulated in the old cohort. Several Treg signature genes as well as transcription factors that are associated with dysfunctional T cell tumor infiltration are differentially expressed. The differential expression of several genes involved in oxidative phosphorylation, glycolysis and glutamine metabolism is also observed. Taken together, this study provides novel findings on the impact of ageing on transcriptional changes in melanoma, and novel therapeutic targets for future studies.
Collapse
Affiliation(s)
- Nehal Farid El-Helbawy
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta 31111, Egypt
| | - Ahmed Ezat El Zowalaty
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, 40530 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
6
|
Che N, Zhao N, Zhao X, Su S, Zhang Y, Bai X, Li F, Zhang D, Li Y. The expression and prognostic significance of PIK3CB in lung adenocarcinoma. Ann Diagn Pathol 2022; 60:152001. [PMID: 35780638 DOI: 10.1016/j.anndiagpath.2022.152001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of this study was to explore the expression and prognostic significance of PIK3CB in lung adenocarcinoma (LUAD) and to analyse the possible molecular mechanism that promotes LUAD development. METHODS Differences of PIK3CB expression at transcriptional level between LUAD and normal tissues were analysed with the Timer and UALCAN databases. Then, immunohistochemical staining was performed to investigate PIK3CB expression at the protein level, and relationships between PIK3CB and clinical characteristics were accessed. Univariate and multivariate Cox regression were performed to identify the independent prognostic risk factors for LUAD. Genetic alterations were analysed using the cBioPortal database. The main coexpressed genes and enrichment pathways of PIK3CB were estimated with the LinkedOmics database. RESULTS Compared with normal tissues, PIK3CB was higherly expressed in LUAD at the transcriptional level and protein level, respectively. PIK3CB expression was closely related to prognosis of LUAD patients, and PIK3CB protein expression was associated with lymph node metastasis and pathological differentiation, but not related to sex, age, pleural invasion, vascular invasion, tumour site, tumour size or clinical stage. PIK3CB and tumour size were independent risk factors for LUAD patients. The expression of PIK3CB was negatively correlated with AKT1 and AKT2, but there was no significant correlation with AKT3, and strong positive correlations with ARMC8, DNAJC13 and PIK3R4. The main enrichment pathways of PIK3CB and related genes included adherens junctions and the phosphatidylinositol signalling pathways, ErbB signalling pathways, Hedgehog signalling pathways, and C-type lectin receptor signalling pathways. Therefore, we hypothesized that PIK3CB expression did not promote LUAD development through the classical PI3K/AKT pathway. CONCLUSION High PIK3CB expression was associated with the development of LUAD and worse prognosis. PIK3CB was an independent risk factor for LUAD patients. Therefore, this study provides a reliable reference for the prognostic assessment and targeted therapy for LUAD patients.
Collapse
Affiliation(s)
- Na Che
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shuai Su
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, 300052, China
| | - Yanhui Zhang
- Department of Pathology, Tianjin Medical University Cancer Hospital, Tianjin 300060, China
| | - Xiaoyu Bai
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fan Li
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanlei Li
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Department of Pathology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
7
|
Huang Y, Jiang Z, Gao X, Luo P, Jiang X. ARMC Subfamily: Structures, Functions, Evolutions, Interactions, and Diseases. Front Mol Biosci 2021; 8:791597. [PMID: 34912852 PMCID: PMC8666550 DOI: 10.3389/fmolb.2021.791597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/15/2021] [Indexed: 12/29/2022] Open
Abstract
Armadillo repeat-containing proteins (ARMCs) are widely distributed in eukaryotes and have important influences on cell adhesion, signal transduction, mitochondrial function regulation, tumorigenesis, and other processes. These proteins share a similar domain consisting of tandem repeats approximately 42 amino acids in length, and this domain constitutes a substantial platform for the binding between ARMCs and other proteins. An ARMC subfamily, including ARMC1∼10, ARMC12, and ARMCX1∼6, has received increasing attention. These proteins may have many terminal regions and play a critical role in various diseases. On the one hand, based on their similar central domain of tandem repeats, this ARMC subfamily may function similarly to other ARMCs. On the other hand, the unique domains on their terminals may cause these proteins to have different functions. Here, we focus on the ARMC subfamily (ARMC1∼10, ARMC12, and ARMCX1∼6), which is relatively conserved in vertebrates and highly conserved in mammals, particularly primates. We review the structures, biological functions, evolutions, interactions, and related diseases of the ARMC subfamily, which involve more than 30 diseases and 40 bypasses, including interactions and relationships between more than 100 proteins and signaling molecules. We look forward to obtaining a clearer understanding of the ARMC subfamily to facilitate further in-depth research and treatment of related diseases.
Collapse
Affiliation(s)
- Yutao Huang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Institue of Neurosurgery of People's Liberation Army of China (PLA), PLA's Key Laboratory of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zijian Jiang
- Department of Hepato-biliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiangyu Gao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Institue of Neurosurgery of People's Liberation Army of China (PLA), PLA's Key Laboratory of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Institue of Neurosurgery of People's Liberation Army of China (PLA), PLA's Key Laboratory of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Mohamed WI, Park SL, Rabl J, Leitner A, Boehringer D, Peter M. The human GID complex engages two independent modules for substrate recruitment. EMBO Rep 2021; 22:e52981. [PMID: 34647674 PMCID: PMC8567238 DOI: 10.15252/embr.202152981] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/31/2021] [Accepted: 09/08/2021] [Indexed: 01/06/2023] Open
Abstract
The human GID (hGID) complex is a conserved E3 ubiquitin ligase regulating diverse biological processes, including glucose metabolism and cell cycle progression. However, the biochemical function and substrate recognition of the multi-subunit complex remain poorly understood. Using biochemical assays, cross-linking mass spectrometry, and cryo-electron microscopy, we show that hGID engages two distinct modules for substrate recruitment, dependent on either WDR26 or GID4. WDR26 and RanBP9 cooperate to ubiquitinate HBP1 in vitro, while GID4 is dispensable for this reaction. In contrast, GID4 functions as an adaptor for the substrate ZMYND19, which surprisingly lacks a Pro/N-end degron. GID4 substrate binding and ligase activity is regulated by ARMC8α, while the shorter ARMC8β isoform assembles into a stable hGID complex that is unable to recruit GID4. Cryo-EM reconstructions of these hGID complexes reveal the localization of WDR26 within a ring-like, tetrameric architecture and suggest that GID4 and WDR26/Gid7 utilize different, non-overlapping binding sites. Together, these data advance our mechanistic understanding of how the hGID complex recruits cognate substrates and provides insights into the regulation of its E3 ligase activity.
Collapse
Affiliation(s)
- Weaam I Mohamed
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Sophia L Park
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland.,Life Science Zürich, PhD Program for Molecular Life Sciences, Zürich, Switzerland
| | - Julius Rabl
- Cryo-EM Knowledge Hub (CEMK), Zürich, Switzerland
| | - Alexander Leitner
- Institute of Molecular Systems Biology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | | | - Matthias Peter
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
9
|
Downregulation of ARMC8 promotes tumorigenesis through activating Wnt/β-catenin pathway and EMT in cutaneous squamous cell carcinomas. J Dermatol Sci 2021; 102:184-192. [PMID: 34016486 DOI: 10.1016/j.jdermsci.2021.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Aberrant expression of Armadillo repeat containing 8 (ARMC8) plays crucial roles in tumor growth and metastasis of various cancers. The specific role of ARMC8 in cutaneous squamous cell carcinoma (cSCC) is yet to be elucidated. OBJECTIVE The present study aimed to investigate the molecular mechanisms of ARMC8 and epithelial-mesenchymal transition (EMT) in cSCC development and provide translational insights for future therapeutics. METHODS cSCC tumor specimens were used to determine the ARMC8 by immunohistochemistry. Three cSCC cell lines including HSC-1, HSC-5 and A431 as well as BALB/C mouse tumor model was utilized to study the potential mechanisms in tumorigenesis. RESULTS Our data identified ARMC8 as a direct downstream target of miR-664. We found that ARMC8 was remarkably low expression in cSCC patient specimens and cSCC cell lines. Knockdown of ARMC8 promotes tumorigenic behaviors such as increased cell proliferation, migration and invasion capacities in vitro and enhanced tumorigenicity in xenograft mouse model. Whereas ARMC8 over-expression inhibits tumorigenesis in cSCC. Together, it revealed ARMC8 functions as a tumor suppressor via restraining Wnt/β-catenin pathway and epithelial-mesenchymal transition in cSCC. CONCLUSION Our data verifies that aberrant expression of ARMC8 plays a vital role in carcinogenesis of cSCC. And overexpression of ARMC8 will facilitate future development of cSCC therapeutic interventions.
Collapse
|
10
|
The CTLH Complex in Cancer Cell Plasticity. JOURNAL OF ONCOLOGY 2019; 2019:4216750. [PMID: 31885576 PMCID: PMC6907057 DOI: 10.1155/2019/4216750] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/24/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022]
Abstract
Cancer cell plasticity is the ability of cancer cells to intermittently morph into different fittest phenotypic states. Due to the intrinsic capacity to change their composition and interactions, protein macromolecular complexes are the ideal instruments for transient transformation. This review focuses on a poorly studied mammalian macromolecular complex called the CTLH (carboxy-terminal to LisH) complex. Currently, this macrostructure includes 11 known members (ARMC8, GID4, GID8, MAEA, MKLN1, RMND5A, RMND5B, RANBP9, RANBP10, WDR26, and YPEL5) and it has been shown to have E3-ligase enzymatic activity. CTLH proteins have been linked to all fundamental biological processes including proliferation, survival, programmed cell death, cell adhesion, and migration. At molecular level, the complex seems to interact and intertwine with key signaling pathways such as the PI3-kinase, WNT, TGFβ, and NFκB, which are key to cancer cell plasticity. As a whole, the CTLH complex is overexpressed in the most prevalent types of cancer and may hold the key to unlock many of the biological secrets that allow cancer cells to thrive in harsh conditions and resist antineoplastic therapy.
Collapse
|
11
|
Wang Z, Zhu X, Zhang T, Yao F. miR-512-5p suppresses the progression of non-small cell lung cancer by targeting β-catenin. Oncol Lett 2019; 19:415-423. [PMID: 31897154 PMCID: PMC6923952 DOI: 10.3892/ol.2019.11102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/27/2019] [Indexed: 01/02/2023] Open
Abstract
The oncogenic protein β-catenin is regulated by microRNAs (miRs) in non-small cell lung cancer (NSCLC). miR-512-5p is downregulated in NSCLC compared with healthy tissues and exhibits a tumour-suppressive effect. To study whether miR-512-5p acts on β-catenin to exert its anticancer effect in NSCLC, miR-512-5p mimic and inhibitor were transfected into NSCLC A549 and H1975 cells. miR-512-5p mimic inhibited the invasion of NSCLC cells and increased apoptosis, which suggested an inhibitory effect of miR-512-5p in NSCLC progression in vitro. By contrast, transfection with the miR-512-5p inhibitor resulted in the opposite effects. A dual-luciferase assay demonstrated that miR-512-5p complementarily bound to the 3′-untranslated region of β-catenin. miR-512-5p mimic suppressed the transcription and translation of β-catenin and reduced the expression of the downstream oncogenes cyclin D1 and matrix metalloproteinases, leading to the inhibition of Wnt/β-catenin signalling and subsequent inhibition of NSCLC tumourigenesis in vitro. In conclusion, miR-512-5p may function as a tumour suppressor in NSCLC by inhibiting the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Zhexin Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiaolei Zhu
- Suzhou Institute of Systems Medicine, Centre of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu 223300, P.R. China
| | - Tuo Zhang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
12
|
Gul IS, Hulpiau P, Sanders E, van Roy F, van Hengel J. Armc8 is an evolutionarily conserved armadillo protein involved in cell-cell adhesion complexes through multiple molecular interactions. Biosci Rep 2019; 39:BSR20180604. [PMID: 30482882 PMCID: PMC6680376 DOI: 10.1042/bsr20180604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 11/08/2018] [Accepted: 11/14/2018] [Indexed: 01/06/2023] Open
Abstract
Armadillo-repeat-containing protein 8 (Armc8) belongs to the family of armadillo-repeat containing proteins, which have been found to be involved in diverse cellular functions including cell-cell contacts and intracellular signaling. By comparative analyses of armadillo repeat protein structures and genomes from various premetazoan and metazoan species, we identified orthologs of human Armc8 and analyzed in detail the evolutionary relationship of Armc8 genes and their encoded proteins. Armc8 is a highly ancestral armadillo protein although not present in yeast. Consequently, Armc8 is not the human ortholog of yeast Gid5/Vid28.Further, we performed a candidate approach to characterize new protein interactors of Armc8. Interactions between Armc8 and specific δ-catenins (plakophilins-1, -2, -3 and p0071) were observed by the yeast two-hybrid approach and confirmed by co-immunoprecipitation and co-localization. We also showed that Armc8 interacts specifically with αE-catenin but neither with αN-catenin nor with αT-catenin. Degradation of αE-catenin has been reported to be important in cancer and to be regulated by Armc8. A similar process may occur with respect to plakophilins in desmosomes. Deregulation of desmosomal proteins has been considered to contribute to tumorigenesis.
Collapse
Affiliation(s)
- Ismail Sahin Gul
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paco Hulpiau
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Howest, University College West Flanders, Bruges, Belgium
| | - Ellen Sanders
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Frans van Roy
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | |
Collapse
|
13
|
Song S, Tan B, Dong X, Yang Q, Chi S, Liu H, Zhang H, Zhang S. Molecular cloning, characterization and expression analysis of ARMC6, ARMC7, ARMC8 from Pacific white shrimp, Litopenaeus vannamei. Gene 2019; 682:50-66. [PMID: 30292870 DOI: 10.1016/j.gene.2018.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/30/2018] [Accepted: 10/04/2018] [Indexed: 10/28/2022]
Abstract
Armadillo repeat-containing proteins (ARMCs) comprise a large family that is widely distributed in eukaryotes and plays prominent roles in cell-cell adhesion, intracellular signaling, and cytoskeletal regulation. In this study, three ARMC genes, termed LvARMC6, LvARMC7 and LvARMC8, were identified and characterized from Litopenaeus vannamei. The complete cDNAs open reading frames (ORF) of LvARMC6, LvARMC7, and LvARMC8 (GenBank accession no. MG735126, MG728109 and KX058562) were 1410 bp, 570 bp and 2046 bp, encoding 469, 189, and 681 amino acids, respectively. Topology analysis indicated that three ARM domains were present in LvARMC6, one in LvARMC7 and six in LvARMC8. The identities of all the three LvARMCs with other species were between 50% and 71%. Phylogenetic analysis illustrated that different subtype of ARMCs formed their own separate branches and LvARMCs were placed in branch of invertebrates respectively with strong bootstrap support. The constitutive expressions of LvARMCs were confirmed by real-time quantitative PCR. LvARMC6, LvARMC7 and LvARMC8 were expressed highest in heart, gills and epithelium, respectively. After challenge with either white spot syndrome virus (WSSV), Vibrio parahemolyticus, or Staphylococcus aureus, all of the LvARMCs demonstrated differential expression profiles in hemocytes, hepatopancreas, intestine and gills. Taken together, our results suggest that LvARMCs may play a role in the innate immune defense against pathogenic viral and bacterial infections of L. vannamei.
Collapse
Affiliation(s)
- Shougang Song
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Beiping Tan
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China
| | - Xiaohui Dong
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China
| | - Qihui Yang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China
| | - Shuyan Chi
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China
| | - Hongyu Liu
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China
| | - Haitao Zhang
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China; The Research Center of Guangdong Evergreen Feed Industry Co., Ltd., Zhanjiang 524022, PR China
| | - Shuang Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, PR China.
| |
Collapse
|
14
|
Yuan J, Jiang L, Guo C. The micro RNA hsa-miR-377-3p inhibits tumor growth in malignant melanoma. RSC Adv 2019; 9:19057-19064. [PMID: 35516861 PMCID: PMC9065064 DOI: 10.1039/c9ra02816a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
Background/Aims: Most recently, micro RNAs (miRNAs/miRs) have been suggested to play a key role in various physiological and pathological processes by regulating the expression of specific genes. The influence of miR-377-3p on multitudinous cancer cells has been investigated; however, its function in melanoma remains undiscovered. Armadillo repeat-containing protein 8 (ARMC8), a target of miR-377-3p, plays essential roles in proliferation, differentiation and apoptosis. Our research aimed to detect the specific roles of miR-377-3p in melanoma. Methods: The MiRNA and mRNA expressions were evaluated by a real-time quantitative polymerase chain reaction in the A375 and HEMa-LP cell lines. We predicted the possible interactions between microRNA and mRNAs by bioinformatics database and constructed them with the Cytoscape software. The proliferation and migration activities were investigated using a cell counting kit-8 (CCK8) and wound-healing assay. Validation of the correlation between miR-377-3p and ARMC8 was implemented by the luciferase reporter assay and PCR. Results: The expression of miR-377-3p was found to be lower in malignant melanoma cells. The upregulation of miR-377-3p inhibited the melanoma cell proliferation, migration, and ARMC8 expression. miR-377-3p was identified to bind to the 3′UTR region of ARMC8 directly; this indicated that miR-377-3p suppressed melanoma cell growth partly mediated via the ARMC8 expression. Conclusion: These findings show that miR-377-3p negatively regulates tumor growth in malignant melanoma, which may thus provide a potential biological target for melanoma treatment and subsequently lead to the development of potential treatments. We have demonstrated that miR-377-3p inhibits melanoma cell growth by binding to the ARMC8 mRNA in the A375 cell line.![]()
Collapse
Affiliation(s)
- Jian Yuan
- TEDA Institute of Biological Sciences and Biotechnology
- Nankai University
- Tianjin 300457
- P. R. China
| | - Lei Jiang
- Department of Medical Technology
- Nanyang Medical College
- Nanyang
- P. R. China
| | - Chaotang Guo
- Department of Bone
- The First People's Hospital of Nanyang
- Nanyang
- P. R. China
| |
Collapse
|
15
|
Chang YC, Fong Y, Tsai EM, Chang YG, Chou HL, Wu CY, Teng YN, Liu TC, Yuan SS, Chiu CC. Exogenous C₈-Ceramide Induces Apoptosis by Overproduction of ROS and the Switch of Superoxide Dismutases SOD1 to SOD2 in Human Lung Cancer Cells. Int J Mol Sci 2018; 19:ijms19103010. [PMID: 30279365 PMCID: PMC6213533 DOI: 10.3390/ijms19103010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 02/07/2023] Open
Abstract
Ceramides, abundant sphingolipids on the cell membrane, can act as signaling molecules to regulate cellular functions including cell viability. Exogenous ceramide has been shown to exert potent anti-proliferative effects against cancer cells, but little is known about how it affects reactive oxygen species (ROS) in lung cancer cells. In this study, we investigated the effect of N-octanoyl-D-erythro-sphingosine (C₈-ceramide) on human non-small-cell lung cancer H1299 cells. Flow cytometry-based assays indicated that C₈-ceramide increased the level of endogenous ROS in H1299 cells. Interestingly, the ratio of superoxide dismutases (SODs) SOD1 and SOD2 seem to be regulated by C₈-ceramide treatment. Furthermore, the accumulation of cell cycle G1 phase and apoptotic populations in C₈-ceramide-treated H1299 cells was observed. The results of the Western blot showed that C₈-ceramide causes a dramatically increased protein level of cyclin D1, a critical regulator of cell cycle G1/S transition. These results suggest that C₈-ceramide acts as a potent chemotherapeutic agent and may increase the endogenous ROS level by regulating the switch of SOD1 and SOD2, causing the anti-proliferation, and consequently triggering the apoptosis of NSCLC H1299 cells. Accordingly, our works may give a promising strategy for lung cancer treatment in the future.
Collapse
Affiliation(s)
- Yuli C Chang
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yao Fong
- Chest Surgery, Chi-Mei Medical Center, Yung Kang City, Tainan 901, Taiwan.
| | - Eing-Mei Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Ya-Gin Chang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Han Lin Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan;.
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan.
| | - Ta-Chih Liu
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Shyng-Shiou Yuan
- Translational Research Center, Cancer Center, Department of Medical Research, Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Chih Chiu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan;.
- Translational Research Center, Cancer Center, Department of Medical Research, Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
16
|
Tangsuwansri C, Saeliw T, Thongkorn S, Chonchaiya W, Suphapeetiporn K, Mutirangura A, Tencomnao T, Hu VW, Sarachana T. Investigation of epigenetic regulatory networks associated with autism spectrum disorder (ASD) by integrated global LINE-1 methylation and gene expression profiling analyses. PLoS One 2018; 13:e0201071. [PMID: 30036398 PMCID: PMC6056057 DOI: 10.1371/journal.pone.0201071] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The exact cause and mechanisms underlying the pathobiology of autism spectrum disorder (ASD) remain unclear. Dysregulation of long interspersed element-1 (LINE-1) has been reported in the brains of ASD-like mutant mice and ASD brain tissues. However, the role and methylation of LINE-1 in individuals with ASD remain unclear. In this study, we aimed to investigate whether LINE-1 insertion is associated with differentially expressed genes (DEGs) and to assess LINE-1 methylation in ASD. METHODS To identify DEGs associated with LINE-1 in ASD, we reanalyzed previously published transcriptome profiles and overlapped them with the list of LINE-1-containing genes from the TranspoGene database. An Ingenuity Pathway Analysis (IPA) of DEGs associated with LINE-1 insertion was conducted. DNA methylation of LINE-1 was assessed via combined bisulfite restriction analysis (COBRA) of lymphoblastoid cell lines from ASD individuals and unaffected individuals, and the methylation levels were correlated with the expression levels of LINE-1 and two LINE-1-inserted DEGs, C1orf27 and ARMC8. RESULTS We found that LINE-1 insertion was significantly associated with DEGs in ASD. The IPA showed that LINE-1-inserted DEGs were associated with ASD-related mechanisms, including sex hormone receptor signaling and axon guidance signaling. Moreover, we observed that the LINE-1 methylation level was significantly reduced in lymphoblastoid cell lines from ASD individuals with severe language impairment and was inversely correlated with the transcript level. The methylation level of LINE-1 was also correlated with the expression of the LINE-1-inserted DEG C1orf27 but not ARMC8. CONCLUSIONS In ASD individuals with severe language impairment, LINE-1 methylation was reduced and correlated with the expression levels of LINE-1 and the LINE-1-inserted DEG C1orf27. Our findings highlight the association of LINE-1 with DEGs in ASD blood samples and warrant further investigation. The molecular mechanisms of LINE-1 and the effects of its methylation in ASD pathobiology deserve further study.
Collapse
Affiliation(s)
- Chayanin Tangsuwansri
- M.Sc. Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Thanit Saeliw
- M.Sc. Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Surangrat Thongkorn
- M.Sc. Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Weerasak Chonchaiya
- Division of Growth and Development and Maximizing Thai Children’s Developmental Potential Research Unit, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genetics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Medical Genetics, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Apiwat Mutirangura
- Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Valerie Wailin Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Tewarit Sarachana
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Ding L, Yao W, Lu J, Gong J, Zhang X. Upregulation of circ_001569 predicts poor prognosis and promotes cell proliferation in non-small cell lung cancer by regulating the Wnt/β-catenin pathway. Oncol Lett 2018; 16:453-458. [PMID: 29928432 DOI: 10.3892/ol.2018.8673] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 01/05/2018] [Indexed: 12/30/2022] Open
Abstract
Circular RNAs (circRNAs) are a large class of RNAs that have previously been identified to be involved in certain diseases, including the development of cancer. However, the role of circ_001569 in non-small cell lung cancer (NSCLC) remains unknown. In the present study, it was demonstrated that expression levels of circ_001569 were significantly increased in NSCLC tissues compared with in adjacent normal tissues. Increased circ_001569 expression was closely associated with tumor differentiation, lymph node metastasis and Tumor-Node-Metastasis classification in NSCLC. Patients that exhibited higher circ_001569 expression demonstrated a poorer survival outcome compared with patients with lower circ_001569 expression. Functional assay results indicated that the knockdown of circ_001569 inhibited the cell proliferation ability of NSCLC in vitro. In addition, it was identified that circ_001569 knockdown reduced the mRNA and protein expression levels of Wnt/β-catenin pathway-associated genes proto-oncogene protein Wnt1, transcription factor 4 and β-catenin in NSCLC cells. Therefore, the results indicated that circ_001569 promoted cell proliferation by regulating the Wnt/β-catenin pathway in NSCLC, and circ_001569 may be a potential target of NSCLC treatment.
Collapse
Affiliation(s)
- Lingchi Ding
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Weidong Yao
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Junguo Lu
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Jun Gong
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Xiaodong Zhang
- Department of Oncology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| |
Collapse
|
18
|
Xu J, Lv W, Hu Y, Wang L, Wang Y, Cao J, Hu J. Wnt3a Expression Is Associated with Epithelial-Mesenchymal Transition and Impacts Prognosis of Lung Adenocarcinoma Patients. J Cancer 2017; 8:2523-2531. [PMID: 28900490 PMCID: PMC5595082 DOI: 10.7150/jca.18560] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 05/08/2017] [Indexed: 12/29/2022] Open
Abstract
Background: Epithelial-mesenchymal transition (EMT) plays an important role in the invasion and migration during cancer metastasis. Wnt3a is one of the ligands in canonical Wnt/β-catenin signaling pathway, which contributes to the carcinogenesis and progression of lung cancer cell lines. The aim of this study was to evaluate the association between Wnt3a and EMT-related proteins (E-cadherin and N-cadherin), and to further investigate its impact on prognosis of lung adenocarcinoma patients. Methods: A total of 147 lung adenocarcinoma patients were included and their clinicopathological characteristics were collected in this retrospective study. The expression levels of Wnt3a, E-cadherin and N-cadherin in post-surgery cancerous and adjacent normal tissues were assessed by immunohistochemistry. The association between Wnt3a and EMT-related proteins and their prognostic values were systematically evaluated. HCC827 and PC9 cell lines were treated with Wnt3a to detect the expression of EMT-related and Wnt/β-catenin signaling-associated proteins, as well as the in vitro migration and invasion abilities. Results: High Wnt3a expression level was significantly associated with low E-cadherin (P<0.001) and high N-cadherin (P<0.001) expression levels in lung adenocarcinoma tissues. Besides, high Wnt3a level predicted poorer lung adenocarcinoma survival by univariate Cox analysis (P=0.001), while the multivariate result was not significant (P=0.355). Subgroup analysis suggested that the prognostic value of Wnt3a expression level was significant in stage T1-T2 (log rank P=0.003) and stage N0 (log rank P=0.031) patients. The multivariate Cox analysis suggested N-cadherin was an independent prognostic factor for lung adenocarcinoma patients (P=0.012). After including these markers into a nomogram, the Harrell's C-index of the nomogram was 0.755. The decision-curve analysis of our nomogram performed net benefit at the threshold probability from 21.6% to 82.0%, and the current model had a better prognostic value than TNM-classification with a lower Akaike information criterion (AIC) value of 166.54. In vitro experiments suggested that Wnt3a could regulate EMT-related proteins and promotes in vitro invasion and migration abilities. Conclusions: Wnt3a could regulate EMT-related proteins and promote the migration and invasion process of lung adenocarcinoma. Although its value as an independent prognostic factor was limited, the combined model suggested good prognostic performance for lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Jinming Xu
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wang Lv
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yeji Hu
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Luming Wang
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yiqing Wang
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jinlin Cao
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Hu
- Department of Thoracic Surgery, The first Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
19
|
Armc5 deletion causes developmental defects and compromises T-cell immune responses. Nat Commun 2017; 8:13834. [PMID: 28169274 PMCID: PMC5309699 DOI: 10.1038/ncomms13834] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 11/04/2016] [Indexed: 01/10/2023] Open
Abstract
Armadillo repeat containing 5 (ARMC5) is a cytosolic protein with no enzymatic activities. Little is known about its function and mechanisms of action, except that gene mutations are associated with risks of primary macronodular adrenal gland hyperplasia. Here we map Armc5 expression by in situ hybridization, and generate Armc5 knockout mice, which are small in body size. Armc5 knockout mice have compromised T-cell proliferation and differentiation into Th1 and Th17 cells, increased T-cell apoptosis, reduced severity of experimental autoimmune encephalitis, and defective immune responses to lymphocytic choriomeningitis virus infection. These mice also develop adrenal gland hyperplasia in old age. Yeast 2-hybrid assays identify 16 ARMC5-binding partners. Together these data indicate that ARMC5 is crucial in fetal development, T-cell function and adrenal gland growth homeostasis, and that the functions of ARMC5 probably depend on interaction with multiple signalling pathways. Mutations in ARMC5 are associated with risk of primary macronodular adrenal gland hyperplasia. Here the authors show that mice lacking Armc5 have adrenal gland hyperplasia and defective T-cell proliferation, differentiation, survival and in vivo T-cell-mediated immune responses.
Collapse
|
20
|
Gul IS, Hulpiau P, Saeys Y, van Roy F. Metazoan evolution of the armadillo repeat superfamily. Cell Mol Life Sci 2017; 74:525-541. [PMID: 27497926 PMCID: PMC11107757 DOI: 10.1007/s00018-016-2319-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/05/2016] [Accepted: 07/25/2016] [Indexed: 02/08/2023]
Abstract
The superfamily of armadillo repeat proteins is a fascinating archetype of modular-binding proteins involved in various fundamental cellular processes, including cell-cell adhesion, cytoskeletal organization, nuclear import, and molecular signaling. Despite their diverse functions, they all share tandem armadillo (ARM) repeats, which stack together to form a conserved three-dimensional structure. This superhelical armadillo structure enables them to interact with distinct partners by wrapping around them. Despite the important functional roles of this superfamily, a comprehensive analysis of the composition, classification, and phylogeny of this protein superfamily has not been reported. Furthermore, relatively little is known about a subset of ARM proteins, and some of the current annotations of armadillo repeats are incomplete or incorrect, often due to high similarity with HEAT repeats. We identified the entire armadillo repeat superfamily repertoire in the human genome, annotated each armadillo repeat, and performed an extensive evolutionary analysis of the armadillo repeat proteins in both metazoan and premetazoan species. Phylogenetic analyses of the superfamily classified them into several discrete branches with members showing significant sequence homology, and often also related functions. Interestingly, the phylogenetic structure of the superfamily revealed that about 30 % of the members predate metazoans and represent an ancient subset, which is gradually evolving to acquire complex and highly diverse functions.
Collapse
Affiliation(s)
- Ismail Sahin Gul
- Inflammation Research Center (IRC), VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, FSVM Building, Technologiepark 927, 9052, Ghent, Belgium
| | - Paco Hulpiau
- Inflammation Research Center (IRC), VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, FSVM Building, Technologiepark 927, 9052, Ghent, Belgium
| | - Yvan Saeys
- Inflammation Research Center (IRC), VIB, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Frans van Roy
- Inflammation Research Center (IRC), VIB, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, FSVM Building, Technologiepark 927, 9052, Ghent, Belgium.
| |
Collapse
|
21
|
Liang X, Men QL, Li YW, Li HC, Chong T, Li ZL. Silencing of Armadillo Repeat-Containing Protein 8 (ARMc8) Inhibits TGF-β-Induced EMT in Bladder Carcinoma UMUC3 Cells. Oncol Res 2017; 25:99-105. [PMID: 28081738 PMCID: PMC7840676 DOI: 10.3727/096504016x14719078133609] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Armadillo repeat-containing protein 8 (ARMc8) is a key factor in regulating cell migration, proliferation, tissue maintenance, and tumorigenesis. However, its role in bladder cancer remains unknown. Thus, in this study we sought to investigate the effect of ARMc8 on the epithelial-to-mesenchymal transition (EMT) progress in bladder cancer cells induced by transforming growth factor-β1 (TGF-β1). Our results found that ARMc8 was highly expressed in bladder cancer cell lines. ARMc8 silencing inhibited the TGF-β1-induced migration and invasion and suppressed the EMT progress in bladder cancer cells. Furthermore, ARMc8 silencing inhibited the TGF-β1-induced expression of β-catenin, cyclin D1, and c-myc in bladder cancer cells. In conclusion, the present study demonstrates a novel function for ARMc8, which acts as a mediator for TGF-β1-induced cell migration/invasion through modulation of the Wnt/β-catenin signaling pathway in bladder cancer cells. This study suggests that ARMc8 may be a potential therapeutic target for the development of therapies for bladder cancer.
Collapse
Affiliation(s)
- Xuan Liang
- *Department of Oncology, The First Affiliated Hospital, Xi’an Jiaotong University Medical College, Xi’an, Shaanxi, P.R. China
| | - Qun-Li Men
- †Department of Urology, The Central Hospital of Baoji, Baoji, Shaanxi, P.R. China
| | - Yong-wei Li
- ‡Department of Urology, The Central Hospital of Weinan, Weinan, Shaanxi, P.R. China
| | - He-Cheng Li
- §Department of Urology, The Second Affiliated Hospital, Xi’an Jiaotong University Medical College, Xi’an, Shaanxi, P.R. China
| | - Tie Chong
- §Department of Urology, The Second Affiliated Hospital, Xi’an Jiaotong University Medical College, Xi’an, Shaanxi, P.R. China
| | - Zhao-lun Li
- §Department of Urology, The Second Affiliated Hospital, Xi’an Jiaotong University Medical College, Xi’an, Shaanxi, P.R. China
| |
Collapse
|
22
|
Yang J, Chen J, He J, Li J, Shi J, Cho WC, Liu X. Wnt signaling as potential therapeutic target in lung cancer. Expert Opin Ther Targets 2016; 20:999-1015. [PMID: 26882052 DOI: 10.1517/14728222.2016.1154945] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/12/2016] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Wingless-type (Wnt) signaling is tightly regulated at multiple cellular levels and is dysregulated in lung cancer. Therefore, it offers therapeutic targets for developing novel agents for lung cancer treatment. AREAS COVERED In this article, we discuss the role of the Wnt signaling pathway in lung cancer, highlighting the aberrant activation of Wnt in lung cancer stem cells and its implication in resistance to radiotherapy, chemotherapy and targeted therapy. We also expound the regulatory roles of microRNAs in Wnt signaling, as well as the potential of the Wnt pathway to provide biomarkers and therapeutic targets in lung cancer. The potential use of small molecule and biological inhibitors targeting the Wnt pathway for lung cancer therapy and prevention is also discussed. EXPERT OPINION Wnt signaling plays an important role in the development and metastasis of lung cancer; the pathway provides targets to develop agents towards for cancer prevention and therapy. A number of clinical trials have shown the effectiveness of Wnt pathway inhibitors in epithelial tumors. However, the side effects should be considered. Nevertheless, the results from clinical studies suggest that inhibitors targeting the Wnt signaling show promise against lung cancer.
Collapse
Affiliation(s)
- Jiali Yang
- a Ningxia Key laboratory of Clinical and Pathogenic Microbiology , Center of Laboratory Medicine of General Hospital at Ningxia Medical University , Yinchuan , Ningxia 750004 , China
| | - Juan Chen
- b Department of Pulmonary and Critical Care Medicine , General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| | - Jinxi He
- c Department of Thoracic Surgery , General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| | - Jing Li
- c Department of Thoracic Surgery , General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| | - Juan Shi
- a Ningxia Key laboratory of Clinical and Pathogenic Microbiology , Center of Laboratory Medicine of General Hospital at Ningxia Medical University , Yinchuan , Ningxia 750004 , China
| | - William C Cho
- d Department of Clinical Oncology , Queen Elizabeth Hospital , Kowloon , Hong Kong
| | - Xiaoming Liu
- a Ningxia Key laboratory of Clinical and Pathogenic Microbiology , Center of Laboratory Medicine of General Hospital at Ningxia Medical University , Yinchuan , Ningxia 750004 , China
- e Human Stem Cell Institute, General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| |
Collapse
|
23
|
Zhou D, Zhang W, Wang Y, Chen L, Luan J. ARMc8: a potential diagnostic and therapeutic target for cancers. Hum Pathol 2016; 54:201. [PMID: 27036312 DOI: 10.1016/j.humpath.2016.02.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
Affiliation(s)
- Dexi Zhou
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China 241000; Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China 241000
| | - Wen Zhang
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China 241000; Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China 241000
| | - Yaqin Wang
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China 241000; Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China 241000
| | - Lu Chen
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China 241000; Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China 241000
| | - Jiajie Luan
- Laboratory of Clinical Pharmacy of Wannan Medical College, Wuhu, Anhui Province, China 241000; Department of Pharmacy in Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China 241000.
| |
Collapse
|
24
|
Cui Z, Zhou L, Song Y, Liu C, Zhu G, Wu X, Yan Y, Xia X, Duan C, Zhou Y, Huang Y, Zhang D. Up-Regulation of Corticocerebral NKD2 in Lipopolysaccharide-Induced Neuroinflammation. Cell Mol Neurobiol 2016; 36:47-55. [PMID: 26084600 PMCID: PMC11482430 DOI: 10.1007/s10571-015-0219-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/01/2015] [Indexed: 12/17/2022]
Abstract
Naked2 (NKD2), one member of Naked family, has been shown to negatively regulate Wnt/β-catenin signaling pathway. It has been recognized that NKD2 plays a vital role in cell homeostasis and prevention of tumorigenesis. However, NKD2 expression and its functional role in the brain in neuroinflammatory processes remain unclear. In our study, we investigated NKD2 distribution and role in lipopolysaccharide (LPS)-induced neuroinflammation rat model. The data indicated that NKD2 was up-regulated in LPS-injected brain, and the cellular localization of NKD2 was predominantly in cerebral cortical neurons. Furthermore, we treated primary neurons with conditioned media (CM) collected from LPS-stimulated mixed glial cultures (MGC). We detected that the up-regulation of NKD2 might be associated with the subsequent apoptosis in neurons. We also found knockdown NKD2 partially depressed the increase of cleaved caspase-3 and increased the reduction of β-catenin stimulated by MGC-CM. Taken together, these results suggested that NKD2 might be involved in neuronal apoptosis via the Wnt/β-catenin pathway during neuroinflammation in CNS. Our findings might provide a new therapeutic target for the prevention of neuroinflammation-involved neurological disorders.
Collapse
Affiliation(s)
- Zhiming Cui
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, 226002, Jiangsu, People's Republic of China
| | - Li Zhou
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yan Song
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Chun Liu
- Laboratory Animal Center, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Guanghui Zhu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, 226002, Jiangsu, People's Republic of China
| | - Xinmin Wu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, 226002, Jiangsu, People's Republic of China
| | - Yaohua Yan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, 226002, Jiangsu, People's Republic of China
| | - Xiaopeng Xia
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, 226002, Jiangsu, People's Republic of China
| | - Chengwei Duan
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Ying Zhou
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuejiao Huang
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, 226361, Jiangsu, People's Republic of China
| | - Dongmei Zhang
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
25
|
Jiang F, Shi Y, Lu H, Li G. Armadillo Repeat-Containing Protein 8 (ARMC8) Silencing Inhibits Proliferation and Invasion in Osteosarcoma Cells. Oncol Res 2016; 24:381-389. [PMID: 27712595 PMCID: PMC7838741 DOI: 10.3727/096504016x14685034103392] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Armadillo repeat-containing protein 8 (ARMC8) plays an important role in regulating cell migration, proliferation, tissue maintenance, signal transduction, and tumorigenesis. However, the expression pattern and role of ARMC8 in osteosarcoma are still unclear. In this study, our aims were to examine the effects of ARMC8 on osteosarcoma and to explore its underlying mechanism. Our results demonstrated that ARMC8 was overexpressed in osteosarcoma cell lines. Knockdown of ARMC8 significantly inhibited osteosarcoma cell proliferation in vitro and markedly inhibited xenograft tumor growth in vivo. ARMC8 silencing also suppressed the epithelial-mesenchymal transition (EMT) phenotype, as well as inhibited the migration and invasion of osteosarcoma cells. Furthermore, knockdown of ARMC8 obviously inhibited the expression of β-catenin, c-Myc, and cyclin D1 in MG-63 cells. In conclusion, this report demonstrates that ARMC8 silencing inhibits proliferation and invasion of osteosarcoma cells. Therefore, ARMC8 may play an important role in the development and progression of human osteosarcoma and may represent a novel therapeutic target in the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Feng Jiang
- *Department of Orthopedics, Huaihe Hospital of Henan University, Kaifeng, Henan Province, P.R. China
| | - Yan Shi
- †Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan Province, P.R. China
| | - Hong Lu
- †Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan Province, P.R. China
| | - Guojun Li
- *Department of Orthopedics, Huaihe Hospital of Henan University, Kaifeng, Henan Province, P.R. China
| |
Collapse
|
26
|
Jiang G, Zhang Y, Zhang X, Fan C, Wang L, Xu H, Yu J, Wang E. ARMc8 indicates aggressive colon cancers and promotes invasiveness and migration of colon cancer cells. Tumour Biol 2015; 36:9005-13. [PMID: 26081621 DOI: 10.1007/s13277-015-3664-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/10/2015] [Indexed: 10/23/2022] Open
Abstract
Recent studies have implicated ARMc8 in promoting tumor formation in non-small cell lung cancer and breast cancer; however, so far, no studies have revealed the expression pattern or cellular function of ARMc8 in colon cancer. In this study, we used immunohistochemical staining to measure ARMc8 expression in 206 cases of colon cancer and matched adjacent normal colon tissue. Clinically important behaviors of cells, including invasiveness and migration, were evaluated after upregulation of ARMc8 expression in HT29 cells through gene transfection or downregulation of expression in LoVo cells using RNAi. We found that ARMc8 was primarily located in the membrane and cytoplasm of tumor cells, and its expression level was significantly higher in colon cancer in comparison to that in the adjacent normal colon tissues (p < 0.001). ARMc8 expression was closely related to TNM stage (p = 0.006), lymph node metastasis (p = 0.001), and poor prognosis (p = 0.002) of colon cancer. The invasiveness and migration capacity of HT29 cells transfected with ARMc8 were significantly greater than those of control cells (p < 0.001), while ARMc8 siRNA treatment significantly reduced cell invasion and migration in LoVo cells (p < 0.001). Furthermore, we demonstrated that ARMc8 could upregulate the expression of MMP7 and snail and downregulate the expression of p120ctn and α-catenin. Therefore, ARMc8 probably enhanced invasiveness and metastatic capacity by affecting these tumor-associated factors, thereby playing a role in enhancing the tumorigenicity of colon cancer cells. ARMc8 is likely to become a potential therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Guiyang Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yong Zhang
- 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
| | - Chuifeng Fan
- 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
| | - Hongtao Xu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Juanhan Yu
- 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
|
27
|
Jiang G, Yang D, Wang L, Zhang X, Xu H, Miao Y, Wang E, Zhang Y. A novel biomarker ARMc8 promotes the malignant progression of ovarian cancer. Hum Pathol 2015; 46:1471-9. [PMID: 26232863 DOI: 10.1016/j.humpath.2015.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/26/2015] [Accepted: 06/03/2015] [Indexed: 10/23/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy worldwide, and the survival rates have remained low in spite of medical advancements. More research is dedicated to the identification of novel biomarkers for this deadly disease. The association between ARMc8 and ovarian cancer remained unraveled. In this study, immunohistochemical staining was used to examine ARMc8 expression in 247 cases of ovarian cancer, 19 cases of borderline ovarian tumors, 41 cases of benign ovarian tumors, and 9 cases of normal ovarian tissues. It was shown that ARMc8 was predominantly located in the cytoplasm of tumor cells, and its expression was up-regulated in the ovarian cancer (61.9%) and the borderline ovarian tumor tissues (57.9%), in comparison with the benign ovarian tumors (12.2%; P < .05) and the normal ovarian tissues (11.1%; P < .05). In ovarian cancer, ARMc8 expression was closely related to International Federation of Gynecology and Obstetrics stages (P = .002), histology grade (P < .001), lymph node metastasis (P = .008), and poor prognosis (P < .001). Univariate and multivariate Cox analyses revealed that ARMc8 expression was an independent prognostic factor for ovarian cancer (P = .039 and P = .005). In addition, ARMc8 could promote the invasion and migration of ovarian cancer cells. Overexpressing ARMc8 enhanced the invasion and metastasis capacity of ARMc8-low Cavo-3 cells (P < .001), whereas interfering ARMc8 significantly reduced cell invasion and metastasis in ARMc8-high SK-OV-3 cells (P < .001). Furthermore, ARMc8 could up-regulate matrix metalloproteinase-7 and snail and down-regulate α-catenin, p120ctn, and E-cadherin. Collectively, ARMc8 may enhance the invasion and metastasis of ovarian cancer cells and likely to become a potential therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Guiyang Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001
| | - Dalei Yang
- Center for Assisted Reproduction, Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, China, 110004
| | - Liang Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001
| | - Xiupeng Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001
| | - Hongtao Xu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001
| | - Yuan Miao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001
| | - Enhua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001
| | - Yong Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China, 110001.
| |
Collapse
|
28
|
Li C, Song G, Zhang S, Wang E, Cui Z. Wnt3a increases the metastatic potential of non-small cell lung cancer cells in vitro in part via its upregulation of Notch3. Oncol Rep 2014; 33:1207-14. [PMID: 25572698 DOI: 10.3892/or.2014.3700] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 12/11/2014] [Indexed: 11/06/2022] Open
Abstract
Metastasis is the leading cause of death in lung cancer. Understanding the mechanisms underlying the process of metastasis is crucial for identifying novel anti-metastatic therapies. Studies indicate that the highly conserved developmental pathways, such as the Wnt and Notch signaling pathways, play important roles in the non-small cell lung cancer (NSCLC) tumorigenesis. However, the roles of both pathways in NSCLC metastasis are unclear. The present study aimed to investigate whether Wnt3a and Notch3, key components of the Wnt and Notch signaling pathways, respectively, regulate the metastatic abilities of NSCLC cells and whether there is some relationship during these regulatory events. Here, we observed that Wnt3a treatment upregulated, not only the protein expression of Notch3, but also the mRNA expression of Notch3 and its downstream genes, HES1 and HEYL. In addition, Wnt3a promoted cell invasion and anchorage-independent growth. Meanwhile, Wnt3a treatment caused epithelial‑mesenchymal transition (EMT)-like morphological changes and F-actin reorganization. The western blotting data showed that Wnt3a treatment decreased the expression of E-cadherin and increased the expression of N-cadherin and vimentin. Compared with Wnt3a treatment, Notch3 shRNA transfection had opposite effects. Furthermore, Notch3 shRNA weakened the effects of Wnt3a treatment on the in vitro cell invasion and EMT. Overall, these observations suggest that Wnt3a and Notch3 may promote the metastasis of NSCLC and Notch3 upregulation is required for the Wnt3a mediated increased metastatic abilities of NSCLC.
Collapse
Affiliation(s)
- Chunyan Li
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Gongru Song
- Department of Biotechnology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Siyang Zhang
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Enhua Wang
- Department of Pathology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zeshi Cui
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|