1
|
Zheng Z, Luan N, Tu K, Liu F, Wang J, Sun J. The roles of protocadherin-7 in colorectal cancer cells on cell proliferation and its chemoresistance. Front Pharmacol 2023; 14:1072033. [PMID: 37063257 PMCID: PMC10098175 DOI: 10.3389/fphar.2023.1072033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/09/2023] [Indexed: 04/18/2023] Open
Abstract
Despite the high mutation frequencies of KRAS, NRAS, and BRAF in colorectal cancer (CRC), there are no effective and reliable inhibitors for these biomarkers. Protocadherin-7 (PCDH7) is regarded as a potentially targetable surface molecule in cancer cells and plays an important role in their proliferation, metastasis, and drug resistance. However, the roles and underlying mechanisms of PCDH7 in CRC remain unclear. In the current study, we found that different colorectal cancer cells expressed PCDH7 over a wide range. The levels of PCDH7 expression were positively associated with cell proliferation and drug resistance in CRC cells but negatively correlated with the potential for cell migration and invasion. Our data indicated that PCDH7 mediated the resistance of CRC cells to ABT-263 (a small-molecule Bcl-2 inhibitor that induces apoptosis) by inhibiting cell apoptosis, which was supported by the downregulation of caspase-3, caspase-9, and PARP cleavage. We found that PCDH7 effectively promoted Mcl-1 expression at both mRNA and protein levels. Furthermore, PCDH7 activated the Wnt signaling pathway, which was confirmed by the increase in β-catenin and c-Myc expression. Finally, and notably, S63845, a novel Mcl-1 inhibitor, not only effectively attenuated the inhibitory effect of PCDH7 on cell apoptosis induced by ABT-263 in vitro but also sensitized PCDH7-overexpressed CRC cell-derived xenografts to ABT-263 in vivo. Taken together, although PCDH7 inhibited the migration and invasion of CRC cells, it could facilitate the development of drug resistance in colorectal cancer cells by positively modulating Mcl-1 expression. The application of the Mcl-1 inhibitor S63845 could be a potential strategy for CRC chemotherapy, especially in CRC with high levels of PCDH7.
Collapse
Affiliation(s)
- Zhibao Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Na Luan
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Tu
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Feiyan Liu
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Jianwei Wang
- Department of Colorectal Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Jianguo Sun, ; Jianwei Wang,
| | - Jianguo Sun
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
- *Correspondence: Jianguo Sun, ; Jianwei Wang,
| |
Collapse
|
2
|
Ma C, Li F, He Z, Zhao S. A more novel and powerful prognostic gene signature of lung adenocarcinoma determined from the immune cell infiltration landscape. Front Surg 2022; 9:1015263. [PMID: 36311939 PMCID: PMC9606711 DOI: 10.3389/fsurg.2022.1015263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the leading histological subtype of lung cancer worldwide, causing high mortality each year. The tumor immune cell infiltration (ICI) is closely associated with clinical outcome with LUAD patients. The present study was designed to construct a gene signature based on the ICI of LUAD to predict prognosis. Methods Downloaded the raw data of three cohorts of the TCGA-LUAD, GSE72094, and GSE68465 and treat them as training cohort, validation cohort one, and validation cohort two for this research. Unsupervised clustering detailed grouped LUAD cases of the training cohort based on the ICI profile. The univariate Cox regression and Kaplan-Meier was adopted to identify potential prognostic genes from the differentially expressed genes recognized from the ICI clusters. A risk score-based prognostic signature was subsequently developed using LASSO-penalized Cox regression analysis. The Kaplan-Meier analysis, Cox analysis, ROC, IAUC, and IBS were constructed to assess the ability to predict the prognosis and effects of clinical variables in another two independent validation cohorts. More innovatively, we searched similar papers in the most recent year and made comprehensive comparisons with ours. GSEA was used to discover the related signaling pathway. The immune relevant signature correlation identification and immune infiltrating analysis were used to evaluate the potential role of the signature for immunotherapy and recognize the critical immune cell that can influence the signature's prognosis capability. Results A signature composed of thirteen gene including ABCC2, CCR2, CERS4, CMAHP, DENND1C, ECT2, FKBP4, GJB3, GNG7, KRT6A, PCDH7, PLK1, and VEGFC, was identified as significantly associated with the prognosis in LUAD patients. The thirteen-gene signature exhibited independence in evaluating the prognosis of LUAD patients in our training and validation cohorts. Compared to our predecessors, our model has an advantage in predictive power. Nine well know immunotherapy targets, including TBX2, TNF, CTLA4, HAVCR2, GZMB, CD8A, PRF1, GZMA, and PDCD1 were recognized correlating with our signature. The mast cells were found to play vital parts in backing on the thirteen-gene signature's outcome predictive capacity. Conclusions Collectively, the current study indicated a robust thirteen-gene signature that can accurately predict LUAD prognosis, which is superior to our predecessors in predictive ability. The immune relevant signatures, TBX2, TNF, CTLA4, HAVCR2, GZMB, CD8A, PRF1, GZMA, PDCD1, and mast cells infiltrating were found closely correlate with the thirteen-gene signature's power.
Collapse
|
3
|
Song C, Pan S, Li D, Hao B, Lu Z, Lai K, Li N, Geng Q. Comprehensive analysis reveals the potential value of inflammatory response genes in the prognosis, immunity, and drug sensitivity of lung adenocarcinoma. BMC Med Genomics 2022; 15:198. [PMID: 36117156 PMCID: PMC9484176 DOI: 10.1186/s12920-022-01340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022] Open
Abstract
Background Although the relationship between inflammatory response and tumor has been gradually recognized, the potential implications of of inflammatory response genes in lung adenocarcinoma (LUAD) remains poorly investigated. Methods RNA sequencing and clinical data were obtained from multiple independent datasets (GSE29013, GSE30219, GSE31210, GSE37745, GSE42127, GSE50081, GSE68465, GSE72094, TCGA and GTEx). Unsupervised clustering analysis was used to identify different tumor subtypes, and LASSO and Cox regression analysis were applied to construct a novel scoring tool. We employed multiple algorithms (ssGSEA, CIBERSORT, MCP counter, and ESTIMATE) to better characterize the LUAD tumor microenvironment (TME) and immune landscapes. GSVA and Metascape analysis were performed to investigate the biological processes and pathway activity. Furthermore, ‘pRRophetic’ R package was used to evaluate the half inhibitory concentration (IC50) of each sample to infer drug sensitivity. Results We identified three distinct tumor subtypes, which were related to different clinical outcomes, biological pathways, and immune characteristics. A scoring tool called inflammatory response gene score (IRGS) was established and well validated in multiple independent cohorts, which could well divide patients into two subgroups with significantly different prognosis. High IRGS patients, characterized by increased genomic variants and mutation burden, presented a worse prognosis, and might show a more favorable response to immunotherapy and chemotherapy. Additionally, based on the cross-talk between TNM stage, IRGS and patients clinical outcomes, we redefined the LUAD stage, which was called ‘IRGS-Stage’. The novel staging system could distinguish patients with different prognosis, with better predictive ability than the conventional TNM staging. Conclusions Inflammatory response genes present important potential value in the prognosis, immunity and drug sensitivity of LUAD. The proposed IRGS and IRGS-Stage may be promising biomarkers for estimating clinical outcomes in LUAD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01340-7.
Collapse
|
4
|
Zhang S, Fu X. The Clinical Significance and Biological Function of PCDH7 in Cervical Cancer. Cancer Manag Res 2021; 13:3841-3847. [PMID: 34012292 PMCID: PMC8126802 DOI: 10.2147/cmar.s298072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/01/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose Cervical cancer is a common cancerous tumor in women that is prone to recurrence and metastasis. Recently, many people have explored the role of protocadherin 7 (PCDH7) in cancer and found that PCDH7 is abnormally expressed in many cancers. The purpose of this study is to explore the expression and mechanism of PCDH7 in cervical cancer and evaluate its clinical prognostic significance. Materials and Methods The expression of PCDH7 in cervical cancer and cells was measured by qRT-PCR. The relationship between PCDH7 expression and the clinical prognosis was calculated using the Kaplan–Meier method and Cox regression analyses. Effects of PCDH7 on cancer cell proliferation, migration, and invasion were studied by MTT assay and transwell assays. Results The expression of PCDH7 in cervical cancer tissues and cell lines was notably downregulated compared with the corresponding control. Low PCDH7 expression was associated with a low survival rate. PCDH7 expression was correlated with lymph node metastasis, cell differentiation, and FIGO staging. PCDH7 can be used as an independent prognostic factor for cervical cancer. Up-regulation of PCDH7 significantly inhibited the proliferation ability, migration potential, and invasion capacity of cancer cells. Conclusion PCDH7 may be used as a prognostic biomarker for cervical cancer patients.
Collapse
Affiliation(s)
- Shitong Zhang
- Department of Obstetrics and Gynecology, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Xianhu Fu
- Department of Obstetrics and Gynecology, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| |
Collapse
|
5
|
van der Weyden L, Offord V, Turner G, Swiatkowska A, Speak AO, Adams DJ. Membrane protein regulators of melanoma pulmonary colonisation identified using a CRISPRa screen and spontaneous metastasis assay in mice. G3-GENES GENOMES GENETICS 2021; 11:6272227. [PMID: 33963380 PMCID: PMC8495943 DOI: 10.1093/g3journal/jkab157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/03/2021] [Indexed: 01/30/2023]
Abstract
Metastasis is the spread of cancer cells to a secondary site within the body, and is the leading cause of death for cancer patients. The lung is a common site of metastasis for many cancer types, including melanoma. Identifying the genes involved in aiding metastasis of melanoma cells to the lungs is critical for the development of better treatments. As the accessibility of cell surface proteins makes them attractive therapeutic targets, we performed a CRISPR activation screen using a library of guide RNAs (gRNAs) targeting the transcription start sites of 2195 membrane protein-encoding genes, to identify genes whose upregulated expression aided pulmonary metastasis. Immunodeficient mice were subcutaneously injected in the flank with murine B16-F0 melanoma cells expressing dCas9 and the membrane protein library gRNAs, and their lungs collected after 14–21 days. Analysis was performed to identify the gRNAs that were enriched in the lungs relative to those present in the cells at the time of administration (day 0). We identified six genes whose increased expression promotes lung metastasis. These genes included several with well-characterized pro-metastatic roles (Fut7, Mgat5, and Pcdh7) that have not previously been linked to melanoma progression, genes linked to tumor progression but that have not previously been described as involved in metastasis (Olfr322 and Olfr441), as well as novel genes (Tmem116). Thus, we have identified genes that, when upregulated in melanoma cells, can aid successful metastasis and colonization of the lung, and therefore may represent novel therapeutic targets to inhibit pulmonary metastasis.
Collapse
Affiliation(s)
- Louise van der Weyden
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Victoria Offord
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Gemma Turner
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Agnes Swiatkowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Anneliese O Speak
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - David J Adams
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| |
Collapse
|
6
|
Dutra TTB, Bezerra TMM, Luna ECM, Carvalho FSR, Chaves FN, Barros Silva PGD, Costa FWG, Pereira KMA. Do Protocadherins Show Prognostic Value in the Carcinogenesis of Human Malignant Neoplasms? Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2020; 21:3677-3688. [PMID: 33369468 PMCID: PMC8046292 DOI: 10.31557/apjcp.2020.21.12.3677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Protocadherins (PCDHs) have been reported as tumor suppressor genes, implying that these genes may be involved in tumor suppression in a variety of cancers. However, a thorough understanding of the functions and mechanisms of PCDHs remains limited. Our aim was to investigate the methylation profile of PCDHs in human malignant neoplasms. Methods: This systematic review has been recorded in PROSPERO (#42019117844) and conducted according to PRISMA’s checklist; search was conducted in LILACS, PubMed, Science Direct, Scopus, and Web of Science databases, manually, with search queries and without date or language restrictions. Results: We found 91 articles, of which 26 were used for this meta-analysis and categorized according to the origin of the neoplasia. In total, 3,377 cases were compiled, with PCDH10, PCDH17, and PCDH8 being the most studied; males were 2.22 times more affected than females. Studies have shown significant heterogeneity (p <0.001), with the odds ratio varying between cases and controls [2.20 (95% CI = 1.11– 4.35) to 209.05 (95% CI = 12.64– 2,457.18)], and the value of association between methylation and cancers studied was 26.08 (95% CI = 15.42–44.13). Conclusion: In this systematic review, we have demonstrated using meta-analysis that PCDHs could emerge as potential tumor suppressor genes and that a significant increase in methylation may be useful for early detection of different cancers. This work may help in the identification of new prognostic biomarkers in malignant neoplasms.
Collapse
Affiliation(s)
- Thaís Torres Barros Dutra
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Thâmara Manoela Marinho Bezerra
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Ealber Carvalho Macêdo Luna
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | | | - Filipe Nobre Chaves
- School of Dentistry, Federal University of Ceara, Campus Sobral, Sobral, Brazil
| | | | - Fábio Wildson Gurgel Costa
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | | |
Collapse
|
7
|
Xu S, Wu X, Tao Z, Li H, Fan C, Chen S, Guo J, Ning Y, Hu X. Effect of aberrantly methylated androgen receptor target gene PCDH7 on the development of androgen-independent prostate cancer cells. Genes Genomics 2019; 42:299-307. [PMID: 31872382 DOI: 10.1007/s13258-019-00903-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/04/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND Androgen-independent prostate cancer (AIPC) is an extremely malignant tumor developed from the androgen dependent (ADPC). However, the mechanism of transition process from ADPC to AIPC remains unknown. OBJECTIVE Here we aimed to identify the androgen receptor (AR) target gene and its roles in AIPC. METHODS Target genes of AR were identified by ChIP-seq in AIPC cells. AR target gene PCDH7 was detected by real time PCR and western blot. Methylation of PCDH7 was measured by bisulfite sequencing and bisulfite amplicon sequencing. Cell growth, invasion and apoptosis were measured by CCK-8, transwell and flow cytometry, respectively. RESULTS AR was significantly enriched in the upstream of PCDH7 gene. The expression of PCDH7 was significantly decreased, while the methylation of PCDH7 was increased in the AIPC cells compared to the ADPC cells. DNA methyltransferase inhibitor significantly suppressed the methylation and increased the mRNA and protein level of PCDH7. Moreover, overexpression of DNMT1 remarkably reduced the mRNA and protein level of PCDH7. DNA methyltransferase inhibitor decreased the cell growth and invasion while promote the cell apoptosis in the AIPC cells. AR significantly target PCDH7, whose hypermethylation may repress cell growth and invasion, and promote apoptosis in AIPC. CONCLUSIONS This study might provide a novel potential target for the treatment of AIPC.
Collapse
Affiliation(s)
- Siqi Xu
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Xiaoyan Wu
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Zhihua Tao
- The Clinical Laboratory of the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hongsheng Li
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Chenliang Fan
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Songjin Chen
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Jianwei Guo
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Yao Ning
- The Clinical Laboratory of the Second Hospital of Jiaxing, Jiaxing, 314000, China
| | - Xuqi Hu
- The Orthopaedics Department of the Second Hospital of Jiaxing, No. 1518, Huancheng North Road, Nanhu District, Jiaxing, 314000, Zhejiang, China.
| |
Collapse
|
8
|
Okuno K, Akiyama Y, Shimada S, Nakagawa M, Tanioka T, Inokuchi M, Yamaoka S, Kojima K, Tanaka S. Asymmetric dimethylation at histone H3 arginine 2 by PRMT6 in gastric cancer progression. Carcinogenesis 2019; 40:15-26. [PMID: 30508037 DOI: 10.1093/carcin/bgy147] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/21/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Histone modification plays important molecular roles in development and progression of cancers. Dysregulation of histone H3 arginine (R) methylation is still unknown in primary cancer, including gastric cancer (GC). Although PRMT6 contributes to asymmetric dimethylation at H3R2 (H3R2me2as) in cancer cells, its molecular functions are poorly understood in GC. In this study, we assessed H3R2me2as and PRMT6 expression levels in 133 primary GC tissues by immunohistochemistry. Increased H3R2me2as was found in 68 GC (51.1%) cases and independently related to poor prognosis. PRMT6 was overexpressed in 70 GC (52.6%) and strongly correlated with the global H3R2me2as levels (P < 0.001). By analyzing biological functions of PRMT6 in GC cell lines by lentivirus-based systems, PRMT6 overexpression enhanced global H3R2me2as and invasiveness in vitro, while PRMT6 knockout (PRMT6-KO) suppressed these effects and tumorigenicity in vivo. ChIP and microarray assays demonstrated that PRMT6-KO GC cells decreased the enrichments of H3R2me2as at the promoter regions of PCDH7, SCD and IGFBP5, resulting in upregulation of their gene expression. PRMT6 was recruited to the promoter regions of PCDH7 and SCD in the PRMT6-overexpressed cells. Knockdown of tumor suppressor PCDH7 in the PRMT6-KO GC cells elevated cell migration and invasion. PRMT6 expression inversely correlated with PCDH7 expression in primary GC (P = 0.021). Collectively, our findings strongly indicate that H3R2me2as is a strong prognostic indicator of GC patients, and PRMT6-overexpressing GC cells may acquire invasiveness through direct transcriptional inhibition of PCDH7 by increasing H3R2me2as level. Thus, inhibition of the PRMT6-H3R2me2as pathway could be a promising new therapeutic strategy in GC.
Collapse
Affiliation(s)
- Keisuke Okuno
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Gastric Surgery, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Minimally Invasive Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masatoshi Nakagawa
- Department of Gastric Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshiro Tanioka
- Department of Gastric Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mikito Inokuchi
- Department of Gastric Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuyuki Kojima
- Department of Gastric Surgery, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Minimally Invasive Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
9
|
Shishodia G, Koul S, Koul HK. Protocadherin 7 is overexpressed in castration resistant prostate cancer and promotes aberrant MEK and AKT signaling. Prostate 2019; 79:1739-1751. [PMID: 31449679 DOI: 10.1002/pros.23898] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/29/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND Castrate resistant prostate cancer (CRPC) accounts for almost all prostate cancer (PCa) deaths. Aberrant activation of ERK/MEK and PI3K/AKT signaling pathways plays an important role in subsets of patients with CRPC. The role of protocadherin 7 (PCDH7) in modulating these signaling pathways is investigated for the first time in PCa in the present investigation. METHODS PCDH7 expression was analyzed in CRPC/neuroendocrine prostate cancer (NEPC) dataset. Protein expression was assessed by Western blotting and immunohistochemistry, and messenger RNA (mRNA) by quantitative real-time polymerase chain reaction. Small hairpin ribonucleic acid was used to knockdown PCDH7. Colony formation, cell migration, and invasion studies were done using standard protocols. RESULTS PCDH7 amplification/mRNA upregulation was observed in 41% of patients in CRPC/NEPC dataset. PCDH7 was also overexpressed in CRPC cells. Increased PCDH protein expression was observed during tumor progression in PCa tissues and in TRAMP mice. Epidermal growth factor treatment resulted in aberrant activation of ERK/AKT. Knockdown of PCDH7 decreased ERK, AKT, and RB phosphorylation and reduced colony formation, decreased cell invasion, and cell migration. CONCLUSIONS These data show for the first time that PCDH7 is overexpressed in a large number of patients with CRPC and suggest that PCDH7 may be an attractive target in subsets of patients with CRPC for whom there is no cure to-date.
Collapse
Affiliation(s)
- Gauri Shishodia
- Department of Biochemistry and Molecular Biology, LSU Health Sciences Center, Shreveport, Louisiana
- Feist Weiller Cancer Center, Shreveport, Louisiana
| | - Sweaty Koul
- Feist Weiller Cancer Center, Shreveport, Louisiana
- Department of Urology, LSU Health Sciences Center, Shreveport, Louisiana
| | - Hari K Koul
- Department of Biochemistry and Molecular Biology, LSU Health Sciences Center, Shreveport, Louisiana
- Feist Weiller Cancer Center, Shreveport, Louisiana
- Overton Brooks Veterans Administrative Medical Center, Shreveport, Louisiana
| |
Collapse
|
10
|
Colás-Algora N, Millán J. How many cadherins do human endothelial cells express? Cell Mol Life Sci 2019; 76:1299-1317. [PMID: 30552441 PMCID: PMC11105309 DOI: 10.1007/s00018-018-2991-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/16/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
The vasculature is the paradigm of a compartment generated by parallel cellular barriers that aims to transport oxygen, nutrients and immune cells in complex organisms. Vascular barrier dysfunction leads to fatal acute and chronic inflammatory diseases. The endothelial barrier lines the inner side of vessels and is the main regulator of vascular permeability. Cadherins comprise a superfamily of 114 calcium-dependent adhesion proteins that contain conserved cadherin motifs and form cell-cell junctions in metazoans. In mature human endothelial cells, only VE (vascular endothelial)-cadherin and N (neural)-cadherin have been investigated in detail. Although both cadherins are essential for regulating endothelial permeability, no comprehensive expression studies to identify which other family members could play a relevant role in endothelial cells has so far been performed. Here, we have reviewed gene and protein expression databases to analyze cadherin expression in mature human endothelium and found that at least 24 cadherin superfamily members are significantly expressed. Based on data obtained from other cell types, organisms and experimental models, we discuss their potential functions, many of them unrelated to the formation of endothelial cell-cell junctions. The expression of this new set of endothelial cadherins highlights the important but still poorly defined roles of planar cell polarity, the Hippo pathway and mitochondria metabolism in human vascular homeostasis.
Collapse
Affiliation(s)
- Natalia Colás-Algora
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
11
|
Zhou X, Padanad MS, Evers BM, Smith B, Novaresi N, Suresh S, Richardson JA, Stein E, Zhu J, Hammer RE, O'Donnell KA. Modulation of Mutant KrasG12D -Driven Lung Tumorigenesis In Vivo by Gain or Loss of PCDH7 Function. Mol Cancer Res 2018; 17:594-603. [PMID: 30409919 DOI: 10.1158/1541-7786.mcr-18-0739] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/26/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022]
Abstract
PROTOCADHERIN 7 (PCDH7), a transmembrane receptor and member of the Cadherin superfamily, is frequently overexpressed in lung adenocarcinoma and is associated with poor clinical outcome. Although PCDH7 was recently shown to promote transformation and facilitate brain metastasis in lung and breast cancers, decreased PCDH7 expression has also been documented in colorectal, gastric, and invasive bladder cancers. These data suggest context-dependent functions for PCDH7 in distinct tumor types. Given that PCDH7 is a potentially targetable molecule on the surface of cancer cells, further investigation of its role in tumorigenesis in vivo is needed to evaluate the therapeutic potential of its inhibition. Here, we report the analysis of novel PCDH7 gain- and loss-of-function mouse models and provide compelling evidence that this cell-surface protein acts as a potent lung cancer driver. Employing a Cre-inducible transgenic allele, we demonstrated that enforced PCDH7 expression significantly accelerates KrasG12D -driven lung tumorigenesis and potentiates MAPK pathway activation. Furthermore, we performed in vivo somatic genome editing with CRISPR/Cas9 in KrasLSL-G12D ; Tp53fl/fl (KP) mice to assess the consequences of PCDH7 loss of function. Inactivation of PCDH7 in KP mice significantly reduced lung tumor development, prolonged survival, and diminished phospho-activation of ERK1/2. Together, these findings establish a critical oncogenic function for PCDH7 in vivo and highlight the therapeutic potential of PCDH7 inhibition for lung cancer. Moreover, given recent reports of elevated or reduced PCDH7 in distinct tumor types, the new inducible transgenic model described here provides a robust experimental system for broadly elucidating the effects of PCDH7 overexpression in vivo. IMPLICATIONS: In this study, we establish a critical oncogenic function for PCDH7 in vivo using novel mouse models and CRISPR/Cas9 genome editing, and we validate the therapeutic potential of PCDH7 inhibition for lung cancer.
Collapse
Affiliation(s)
- Xiaorong Zhou
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas.,Department of Immunology, Nantong University School of Medicine, Nantong, China
| | - Mahesh S Padanad
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Bret M Evers
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Bethany Smith
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Nicole Novaresi
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Shruthy Suresh
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - James A Richardson
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas.,Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Emily Stein
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Jingfei Zhu
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas
| | - Robert E Hammer
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
| | - Kathryn A O'Donnell
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, Texas. .,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas.,Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
12
|
Wu DQ, Yang ZF, Wang KJ, Feng XY, Lv ZJ, Li Y, Jian ZX. AQP8 inhibits colorectal cancer growth and metastasis by down-regulating PI3K/AKT signaling and PCDH7 expression. Am J Cancer Res 2018; 8:266-279. [PMID: 29511597 PMCID: PMC5835694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/29/2017] [Indexed: 06/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant cancers and the leading cause of cancer-related deaths in worldwide. Although the monoclonal antibody therapy is prescribed for CRC, the metastasis resistant to therapy is the major cause of death of patients with CRC, which indicating the urgent demands for new therapeutic targets discovery. Aquaporin 8 (AQP8) has been identified alter expressed in several cancers including breast cancer, lung cancer and prostatic carcinoma. Our study demonstrated the functional significance of AQP8 in CRC cells growth and metastasis. Over-expression of AQP8 remarkably decreased growth, aggressiveness and colony formation in the CRC SW480 and HT-29 cells. Mechanistically, AQP8 over-expression inhibited tumorigenic phenotype by inactivating PI3K/AKT signaling and inhibiting PCDH7 expression. Furthermore, in vivo studies using nude mice xenograft and metastasis model identified the pivotal role of AQP8 in CRC cells growth and metastasis. Taken together, the present study verifies the vital role of the endogenous AQP8 in colorectal cancer progression.
Collapse
Affiliation(s)
- De Qing Wu
- Southen Medical UniversityNo. 1838 Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, China
- Guangdong General Hospital, Guangdong Academy of Medical ScienceNo. 106, Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Zi Feng Yang
- Guangdong General Hospital, Guangdong Academy of Medical ScienceNo. 106, Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Ke Jian Wang
- The Third Afiiliated Hospital of Guangzhou Medical UniversityNo. 63 Dorbao Road, Liwan District, Guangzhou, Guangdong, China
| | - Xing Yu Feng
- Guangdong General Hospital, Guangdong Academy of Medical ScienceNo. 106, Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Ze Jian Lv
- Guangdong General Hospital, Guangdong Academy of Medical ScienceNo. 106, Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Yong Li
- Guangdong General Hospital, Guangdong Academy of Medical ScienceNo. 106, Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Zhi Xiang Jian
- Southen Medical UniversityNo. 1838 Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, China
- Guangdong General Hospital, Guangdong Academy of Medical ScienceNo. 106, Zhongshan 2nd Road, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Li D, Wang X, Mei H, Fang E, Ye L, Song H, Yang F, Li H, Huang K, Zheng L, Tong Q. Long Noncoding RNA pancEts-1 Promotes Neuroblastoma Progression through hnRNPK-Mediated β-Catenin Stabilization. Cancer Res 2018; 78:1169-1183. [PMID: 29311158 DOI: 10.1158/0008-5472.can-17-2295] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/02/2017] [Accepted: 01/02/2018] [Indexed: 11/16/2022]
Abstract
Long noncoding RNAs (lncRNA) play essential roles in tumor progression. However, the functions of lncRNAs in the tumorigenesis and aggressiveness of neuroblastoma still remain to be determined. Here, we report the identification of lncRNA pancEts-1 as a novel driver of neuroblastoma progression by using a public microarray dataset. LncRNA pancEts-1 promoted the growth, invasion, and metastasis of neuroblastoma cells in vitro and in vivo Mechanistically, pancEts-1 bound to hnRNPK to facilitate its physical interaction with β-catenin, whereas hnRNPK stabilized the β-catenin by inhibiting proteasome-mediated degradation, resulting in transcriptional alteration of target genes associated with neuroblastoma progression. Both pancEts-1 and hnRNPK were upregulated in clinical neuroblastoma tissues, and were associated with unfavorable outcome of patients. Overall, our results define an oncogenic role of pancEts-1 in neuroblastoma progression through hnRNPK-mediated β-catenin stabilization, with potential implications for the clinical therapeutics of neuroblastoma.Significance: These findings reveal the oncogenic functions of a long noncoding RNA in neuroblastoma progression, offering a potential target for clinical therapeutics. Cancer Res; 78(5); 1169-83. ©2018 AACR.
Collapse
Affiliation(s)
- Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Xiaojing Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Hong Mei
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Erhu Fang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Lin Ye
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Huajie Song
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Feng Yang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Huanhuan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Kai Huang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Liduan Zheng
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China.
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China.
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| |
Collapse
|
14
|
Chen HF, Ma RR, He JY, Zhang H, Liu XL, Guo XY, Gao P. Protocadherin 7 inhibits cell migration and invasion through E-cadherin in gastric cancer. Tumour Biol 2017; 39:1010428317697551. [PMID: 28381163 DOI: 10.1177/1010428317697551] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The protocadherin 7 is a member of the protocadherin family that expressed aberrantly in many types of human cancers. However, its expression, function, and underlying mechanisms are little known in gastric cancer. In this study, we detected protocadherin 7 expression in gastric cancer tissues and non-tumorous gastric mucosa tissues by real-time quantitative polymerase chain reaction and immunohistochemistry. The association of protocadherin 7 expression with the clinicopathological characteristics and the prognosis was subsequently analyzed. MTS ((3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)) and transwell assays were performed to assess the effect of protocadherin 7 on proliferation, migration, and invasion in gastric cancer cell lines. Moreover, real-time quantitative polymerase chain reaction and western blot were used to detect the expression of epithelial-mesenchymal transition markers. Protocadherin 7 expression was decreased gradiently from normal tissue to gastric cancer, especially in gastric cancer tissue with lymph node metastasis. Low expression of protocadherin 7 was significantly associated with Lauren's classification ( p = 0.0005), lymph node metastases ( p = 0.0002), and tumor node metastasis stage ( p = 0.0221), as well as poor prognosis ( p < 0.05). Furthermore, down-regulation of protocadherin 7 in gastric cancer cell lines significantly increased their migration and invasion abilities (both p < 0.05), while it had no influence on the gastric cancer cell proliferation ( p > 0.05). Additionally, our results demonstrated that E-cadherin expression was down-regulated in gastric cancer cells with protocadherin 7 depletion. Our data indicated that protocadherin 7 may play important roles in the invasion and metastasis of gastric cancer, and protocadherin 7 could suppress cell migration and invasion through E-cadherin inhibition. Protocadherin 7 can serve as a novel biomarker for diagnostic and prognosis in patients with gastric cancer.
Collapse
Affiliation(s)
- Hong-Fang Chen
- 1 Department of Pathology, Qilu Hospital, Shandong University, Jinan, China.,2 Department of Pathology, Yidu Central Hospital of Weifang, Weifang, China
| | - Ran-Ran Ma
- 1 Department of Pathology, Qilu Hospital, Shandong University, Jinan, China.,3 Department of Pathology, School of Medicine, Shandong University, Jinan, China
| | - Jun-Yi He
- 1 Department of Pathology, Qilu Hospital, Shandong University, Jinan, China.,3 Department of Pathology, School of Medicine, Shandong University, Jinan, China
| | - Hui Zhang
- 1 Department of Pathology, Qilu Hospital, Shandong University, Jinan, China.,3 Department of Pathology, School of Medicine, Shandong University, Jinan, China
| | - Xiao-Ling Liu
- 2 Department of Pathology, Yidu Central Hospital of Weifang, Weifang, China
| | - Xiang-Yu Guo
- 1 Department of Pathology, Qilu Hospital, Shandong University, Jinan, China.,3 Department of Pathology, School of Medicine, Shandong University, Jinan, China
| | - Peng Gao
- 1 Department of Pathology, Qilu Hospital, Shandong University, Jinan, China.,3 Department of Pathology, School of Medicine, Shandong University, Jinan, China
| |
Collapse
|