Cargando…

HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer

Advanced gallbladder cancer (GBC) is one of the most malignant of all types of biliary tract cancers that is associated with poor prognosis and high mortality. Accumulating evidence suggest that the B7 family of proteins serve an essential role in various types of cancers, including GBC. However, th...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Yizhou, Li, Hanrong, Lv, Chao, Wu, Baokang, Yu, Yang, Zhong, Chongli, Lang, Qi, Liang, Zhiyun, Li, Yang, Shi, Yu, Zhang, Jian, Xu, Feng, Tian, Yu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9374468/
https://www.ncbi.nlm.nih.gov/pubmed/35920182
http://dx.doi.org/10.3892/ijo.2022.5402
_version_ 1784767789621313536
author Zhang, Yizhou
Li, Hanrong
Lv, Chao
Wu, Baokang
Yu, Yang
Zhong, Chongli
Lang, Qi
Liang, Zhiyun
Li, Yang
Shi, Yu
Zhang, Jian
Xu, Feng
Tian, Yu
author_facet Zhang, Yizhou
Li, Hanrong
Lv, Chao
Wu, Baokang
Yu, Yang
Zhong, Chongli
Lang, Qi
Liang, Zhiyun
Li, Yang
Shi, Yu
Zhang, Jian
Xu, Feng
Tian, Yu
author_sort Zhang, Yizhou
collection PubMed
description Advanced gallbladder cancer (GBC) is one of the most malignant of all types of biliary tract cancers that is associated with poor prognosis and high mortality. Accumulating evidence suggest that the B7 family of proteins serve an essential role in various types of cancers, including GBC. However, the potential function and regulatory mechanism of human endogenous retrovirus-H long terminal repeat-associating protein 2 (HHLA2; also known as B7-H7 or B7H5) in GBC remain poorly understood. In the present study, immunohistochemistry was used to examine the expression pattern of HHLA2 in samples from 89 patients with GBC. The possible association between HHLA2 expression and the clinicopathological parameters, including prognosis, were then assessed. Using lentiviruses, overexpression of HHLA2 plasmid or short-hairpin RNA (shRNA) of HHLA2 were transfected into GBC-SD cells to overexpress or knock down HHLA2 expression, respectively. The effects of HHLA2 overexpression and knockdown on the epithelial-mesenchymal transition (EMT) process on GBC-SD cells were measured by the western blotting and immunofluorescence staining of collagen I, N-cadherin, E-cadherin, vimentin and α-SMA. By contrast, changes in cell proliferation were measured using EdU assay. Cell invasion and migration were assessed using Transwell and wound-healing assays, respectively. In addition, a xenograft mouse model was established to evaluate the tumorigenic ability of the GBC cell line in vivo after stable transfection with lentivirus for HHLA2 overexpression or shRNA for HHLA2 knockdown. The regulatory relationships among TGF-β1, long non-coding RNA (lncRNA) H19 (H19) and HHLA2 were then investigated. The mRNA expression of lncRNA H19 were assessed using reverse transcription-quantitative PCR, whereas the expression levels of HHLA2 were detected by western blotting and immunofluorescence staining. HHLA2 expression was found to gradually increase as the stages of the GBC samples become more advanced. In addition, the expression level of HHLA2 was calculated to be positively associated with the Nevin stage, American Joint Committee on Cancer stage, tumor invasion and regional lymph node metastasis but was negatively associated with the overall patient survival (OS). In vitro experiments demonstrated that overexpression of HHLA2 promoted GBC migration, invasion, proliferation and EMT, whereas in vivo experiments found a promoting role of HHLA2 overexpression on GBC tumor growth. After transfection with lentiviruses encoding the overexpression plasmid of lncRNA H19, GBC migration, invasion, proliferation and EMT were increased. By contrast, knocking down HHLA2 expression suppressed TGF-β1- or lncRNA H19 overexpression-induced GBC migration, invasion, proliferation and EMT. In addition, HHLA2 knockdown significantly reduced the sizes of the GBC tumors in vivo. These results suggest that HHLA2 overexpression can promote GBC progression. Conversely, ablation of HHLA2 expression inhibited both TGF-β1- and lncRNA H19-induced GBC progression, suggesting that HHLA2 is a potential therapeutic target for this disease.
format Online
Article
Text
id pubmed-9374468
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-93744682022-08-14 HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer Zhang, Yizhou Li, Hanrong Lv, Chao Wu, Baokang Yu, Yang Zhong, Chongli Lang, Qi Liang, Zhiyun Li, Yang Shi, Yu Zhang, Jian Xu, Feng Tian, Yu Int J Oncol Articles Advanced gallbladder cancer (GBC) is one of the most malignant of all types of biliary tract cancers that is associated with poor prognosis and high mortality. Accumulating evidence suggest that the B7 family of proteins serve an essential role in various types of cancers, including GBC. However, the potential function and regulatory mechanism of human endogenous retrovirus-H long terminal repeat-associating protein 2 (HHLA2; also known as B7-H7 or B7H5) in GBC remain poorly understood. In the present study, immunohistochemistry was used to examine the expression pattern of HHLA2 in samples from 89 patients with GBC. The possible association between HHLA2 expression and the clinicopathological parameters, including prognosis, were then assessed. Using lentiviruses, overexpression of HHLA2 plasmid or short-hairpin RNA (shRNA) of HHLA2 were transfected into GBC-SD cells to overexpress or knock down HHLA2 expression, respectively. The effects of HHLA2 overexpression and knockdown on the epithelial-mesenchymal transition (EMT) process on GBC-SD cells were measured by the western blotting and immunofluorescence staining of collagen I, N-cadherin, E-cadherin, vimentin and α-SMA. By contrast, changes in cell proliferation were measured using EdU assay. Cell invasion and migration were assessed using Transwell and wound-healing assays, respectively. In addition, a xenograft mouse model was established to evaluate the tumorigenic ability of the GBC cell line in vivo after stable transfection with lentivirus for HHLA2 overexpression or shRNA for HHLA2 knockdown. The regulatory relationships among TGF-β1, long non-coding RNA (lncRNA) H19 (H19) and HHLA2 were then investigated. The mRNA expression of lncRNA H19 were assessed using reverse transcription-quantitative PCR, whereas the expression levels of HHLA2 were detected by western blotting and immunofluorescence staining. HHLA2 expression was found to gradually increase as the stages of the GBC samples become more advanced. In addition, the expression level of HHLA2 was calculated to be positively associated with the Nevin stage, American Joint Committee on Cancer stage, tumor invasion and regional lymph node metastasis but was negatively associated with the overall patient survival (OS). In vitro experiments demonstrated that overexpression of HHLA2 promoted GBC migration, invasion, proliferation and EMT, whereas in vivo experiments found a promoting role of HHLA2 overexpression on GBC tumor growth. After transfection with lentiviruses encoding the overexpression plasmid of lncRNA H19, GBC migration, invasion, proliferation and EMT were increased. By contrast, knocking down HHLA2 expression suppressed TGF-β1- or lncRNA H19 overexpression-induced GBC migration, invasion, proliferation and EMT. In addition, HHLA2 knockdown significantly reduced the sizes of the GBC tumors in vivo. These results suggest that HHLA2 overexpression can promote GBC progression. Conversely, ablation of HHLA2 expression inhibited both TGF-β1- and lncRNA H19-induced GBC progression, suggesting that HHLA2 is a potential therapeutic target for this disease. D.A. Spandidos 2022-08-01 /pmc/articles/PMC9374468/ /pubmed/35920182 http://dx.doi.org/10.3892/ijo.2022.5402 Text en Copyright: © Zhang et al. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Zhang, Yizhou
Li, Hanrong
Lv, Chao
Wu, Baokang
Yu, Yang
Zhong, Chongli
Lang, Qi
Liang, Zhiyun
Li, Yang
Shi, Yu
Zhang, Jian
Xu, Feng
Tian, Yu
HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer
title HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer
title_full HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer
title_fullStr HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer
title_full_unstemmed HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer
title_short HHLA2 promotes tumor progression by long non-coding RNA H19 in human gallbladder cancer
title_sort hhla2 promotes tumor progression by long non-coding rna h19 in human gallbladder cancer
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9374468/
https://www.ncbi.nlm.nih.gov/pubmed/35920182
http://dx.doi.org/10.3892/ijo.2022.5402
work_keys_str_mv AT zhangyizhou hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT lihanrong hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT lvchao hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT wubaokang hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT yuyang hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT zhongchongli hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT langqi hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT liangzhiyun hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT liyang hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT shiyu hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT zhangjian hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT xufeng hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer
AT tianyu hhla2promotestumorprogressionbylongnoncodingrnah19inhumangallbladdercancer