1
|
Mao Y, Hu Y, Meng H, Qin J, An Q, Zhang C, Guo C, Zhao Y, Tan D, Ge X, Shi C. FXYD5 regulates gastric cancer cell metastasis and drug resistance by EMT modulation. Cancer Gene Ther 2025; 32:318-326. [PMID: 39984673 DOI: 10.1038/s41417-025-00878-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/19/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related mortality and the fourth most prevalent malignancy globally. The high prevalence and mortality rates of GC are attributed to various factors, including drug resistance, local recurrence, and distant metastases. There is an urgent need to identify novel therapeutic targets for GC. Patient-derived xenografts (PDX) model offers unique advantages in maintaining the molecular heterogeneity and tumor microenvironment of primary tumors, offering significant advantages for the screening of personalized therapeutic targets. In this study, we established GC PDX models with metastatic potential through orthotopic transplantation and investigated the different gene expressions between primary and metastatic tumors using PCR-array analysis. We found that the metastatic tumors displayed elevated levels of FXYD domain-containing ion transport regulator 5 (FXYD5) compared to the primary tumors. Additionally, reducing FXYD5 expression was found to inhibit the invasion, metastasis, and proliferation of GC cells. Silencing FXYD5 also reversed the resistance of GC cells to doxorubicin and vincristine by modulating the epithelial-mesenchymal transition (EMT) process and the expression of multidrug resistance protein 2. This study indicates that FXYD5 is involved in GC progression and regulates chemotherapy resistance, suggesting its potential as a novel therapeutic target for the clinical treatment of GC.
Collapse
Affiliation(s)
- Yuning Mao
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
- Department of Pathology, Xijing Hospital, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Yaohua Hu
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Han Meng
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Jing Qin
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Qingling An
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Chenbo Guo
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Xu Ge
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Air Force Medical University, 710032, Xi'an, Shaanxi, China.
| |
Collapse
|
2
|
Harvanik P, Šemeláková M, Solárová Z, Solár P. Novel factors of cisplatin resistance in epithelial ovarian tumours. Adv Med Sci 2025; 70:94-102. [PMID: 39880191 DOI: 10.1016/j.advms.2025.01.005] [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: 07/18/2024] [Revised: 10/11/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
Ovarian tumours are these days one of the biggest oncogynecological problems. In addition to surgery, the treatment of ovarian cancer includes also chemotherapy in which platinum preparations are one of the most used chemotherapeutic drugs. The principle of antineoplastic effects of cisplatin (cis-diamminedichloroplatinum(II), CDDP) is its binding to the DNA and the formation of adducts. While DNA adducts induce the process of apoptosis, or inhibit the process of DNA replication, which prevents further division of tumour cells, various molecular mechanisms can reverse this process. On the other hand, with increasing scientific knowledge, it is becoming clearer that chemotherapy resistance is a very complex process. In this regard, factors and the amount of their expression may regulate the effect of resistance to chemotherapy. This review focuses on new molecular mechanisms and factors such as mitochondrial dynamics, epithelial-mesenchymal transition (EMT), cluster of differentiation, exosomes and others, that could be involved in the emergence of CDDP resistance.
Collapse
Affiliation(s)
- Pavol Harvanik
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Martina Šemeláková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Peter Solár
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic.
| |
Collapse
|
3
|
Niinivirta A, Salo T, Åström P, Juurikka K, Risteli M. Prognostic value of dysadherin in cancer: A systematic review and meta-analysis. Front Oncol 2022; 12:945992. [PMID: 36119538 PMCID: PMC9479204 DOI: 10.3389/fonc.2022.945992] [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: 05/17/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Cancer is a leading cause of death worldwide and novel prognostic factors are reported with increasing numbers. Systematic reviews and meta-analyses on cumulative research data are crucial in estimating the true prognostic value of proposed factors. Dysadherin (FXYD Domain Containing Ion Transport Regulator 5; FXYD5) is a cell membrane glycoprotein that modulates Na+, K+-ATPase activity and cell-cell adhesion. It is abundantly expressed in a variety of cancer cells, but only in a limited number of normal cells and its levels are increased in many different tumor types. The expression or level of dysadherin has been suggested as an independent predictor for metastasis and poor prognosis by number of studies, yet we lack a definitive answer. In this study, we systematically evaluated the prognostic value of dysadherin in cancer and summarized the current knowledge on the subject. PubMed, Scopus, Web of Science and relevant clinical trial and preprint databases were searched for relevant publications and PRISMA and REMARK guidelines were applied in the process. After a careful review, a total of 23 original research articles were included. In each study, dysadherin was pointed as a marker for poor prognosis. Meta-analyses revealed 3- and 1.5-fold increases in the risk of death (fixed effects HR 3.08, 95% CI 1.88-5.06, RR 1.47, 95% CI 1.06-2.05 on overall survival, respectively) for patients with high (>50%) tumoral FXYD5 level. In many studies, a connection between dysadherin expression or level and metastatic behavior of the cancer as well as inverse correlation with E-cadherin level were reported. Thus, we conclude that dysadherin might be a useful prognostic biomarker in the assessment of disease survival of patients with solid tumors.
Collapse
Affiliation(s)
- Aino Niinivirta
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Oral and Maxillofacial Diseases, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
- Department of Pathology (HUSLAB), Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
| | - Pirjo Åström
- Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Krista Juurikka
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Maija Risteli
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
4
|
Borzdziłowska P, Bednarek I. Alpha Mangostin and Cisplatin as Modulators of Exosomal Interaction of Ovarian Cancer Cell with Fibroblasts. Int J Mol Sci 2022; 23:8913. [PMID: 36012171 PMCID: PMC9408324 DOI: 10.3390/ijms23168913] [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: 07/14/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
The diversity of exosomes and their role in the microenvironment make them an important point of interest in the development of cancer. In our study, we evaluated the effect of exosomes derived from ovarian cancer cells on gene expression in fibroblasts, including genes involved in metastasis. We also attempted to evaluate the indirect effect of cisplatin and/or α-mangostin on metastasis. In this aspect, we verified the changes induced by the drugs we tested on vesicular transfer associated with the release of exosomes by cells. We isolated exosomes from ovarian cancer cells treated and untreated with drugs, and then normal human fibroblasts were treated with the isolated exosomes. Changes in the expression of genes involved in the metastasis process were then examined. In our study, we observed altered expression of genes involved in various steps of the metastasis process (including genes related to cell adhesion, genes related to the interaction with the extracellular matrix, the cell cycle, cell growth and proliferation, and apoptosis). We have shown that α-mangostin and/or cisplatin, as chemotherapeutic agents, not only directly affect tumor cells but may also indirectly (via exosomes) contribute to delaying metastasis development.
Collapse
Affiliation(s)
- Paulina Borzdziłowska
- Department of Biotechnology and Genetic Engineering, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | | |
Collapse
|
5
|
Themistocleous SC, Yiallouris A, Tsioutis C, Zaravinos A, Johnson EO, Patrikios I. Clinical significance of P-class pumps in cancer. Oncol Lett 2021; 22:658. [PMID: 34386080 PMCID: PMC8298992 DOI: 10.3892/ol.2021.12919] [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: 01/25/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
P-class pumps are specific ion transporters involved in maintaining intracellular/extracellular ion homeostasis, gene transcription, and cell proliferation and migration in all eukaryotic cells. The present review aimed to evaluate the role of P-type pumps [Na+/K+ ATPase (NKA), H+/K+ ATPase (HKA) and Ca2+-ATPase] in cancer cells across three fronts, namely structure, function and genetic expression. It has been shown that administration of specific P-class pumps inhibitors can have different effects by: i) Altering pump function; ii) inhibiting cell proliferation; iii) inducing apoptosis; iv) modifying metabolic pathways; and v) induce sensitivity to chemotherapy and lead to antitumor effects. For example, the NKA β2 subunit can be downregulated by gemcitabine, resulting in increased apoptosis of cancer cells. The sarcoendoplasmic reticulum calcium ATPase can be inhibited by thapsigargin resulting in decreased prostate tumor volume, whereas the HKA α subunit can be affected by proton pump inhibitors in gastric cancer cell lines, inducing apoptosis. In conclusion, the present review highlighted the central role of P-class pumps and their possible use and role as anticancer cellular targets for novel therapeutic chemical agents.
Collapse
Affiliation(s)
| | - Andreas Yiallouris
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| | - Constantinos Tsioutis
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
- College of Medicine, Member of Qatar University Health, Qatar University, 2713 Doha, Qatar
| | - Elizabeth O. Johnson
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| | - Ioannis Patrikios
- Department of Medicine, School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus
| |
Collapse
|
6
|
Tian M, Wang T, Wang P. Development and Clinical Validation of a Seven-Gene Prognostic Signature Based on Multiple Machine Learning Algorithms in Kidney Cancer. Cell Transplant 2021; 30:963689720969176. [PMID: 33626918 PMCID: PMC7917425 DOI: 10.1177/0963689720969176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
About a third of patients with kidney cancer experience recurrence or cancer-related progression. Clinically, kidney cancer prognoses may be quite different, even in patients with kidney cancer at the same clinical stage. Therefore, there is an urgent need to screen for kidney cancer prognosis biomarkers. Differentially expressed genes (DEGs) were identified using kidney cancer RNA sequencing data from the Gene Expression Omnibus (GEO) database. Biomarkers were screened using random forest (RF) and support vector machine (SVM) models, and a multigene signature was constructed using the least absolute shrinkage and selection operator (LASSO) regression analysis. Univariate and multivariate Cox regression analyses were performed to explore the relationships between clinical features and prognosis. Finally, the reliability and clinical applicability of the model were validated, and relationships with biological pathways were identified. Western blots were also performed to evaluate gene expression. A total of 50 DEGs were obtained by intersecting the RF and SVM models. A seven-gene signature (RNASET2, EZH2, FXYD5, KIF18A, NAT8, CDCA7, and WNT7B) was constructed by LASSO regression. Univariate and multivariate Cox regression analyses showed that the seven-gene signature was an independent prognostic factor for kidney cancer. Finally, a predictive nomogram was established in The Cancer Genome Atlas (TCGA) cohort and validated internally. In tumor tissue, RNASET2 and FXYD5 were highly expressed and NAT8 was lowly expressed at the protein and transcription levels. This model could complement the clinicopathological characteristics of kidney cancer and promote the personalized management of patients with kidney cancer.
Collapse
Affiliation(s)
- Mi Tian
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Wang
- Department of Pathology, Shenyang KingMed Center for Clinical Laboratory Co, Ltd, Shenyang, China
| | - Peng Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Liu YK, Jia YJ, Liu SH, Shi HJ, Ma J. Low expression of FXYD5 reverses the cisplatin resistance of epithelial ovarian cancer cells. Histol Histopathol 2021; 36:535-545. [PMID: 33570156 DOI: 10.14670/hh-18-310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the effect of the downregulation of FXYD domain-containing ion transport regulator 5 (FXYD5) on the cisplatin resistance (CisR) of epithelial ovarian cancer (EOC) cells. METHODS A2780-CisR and SKOV3-CisR cells were obtained through repeated administrations of different cisplatin concentrations, and the half-maximal inhibition concentration (IC50) was calculated by MTT assays. After transfection with FXYD5 siRNA-1 and FXYD5 siRNA-2, the IC50 values of the A2780-CisR and SKOV3-CisR cells were also detected by the MTT method. Cell proliferation, migration, invasion and apoptosis were evaluated through 5-ethynyl-2'-deoxyuridine (EdU) DNA synthesis, wound healing, Transwell invasion and Annexin-V-FITC/PI dual-staining assays, respectively. qRT-PCR and Western blotting were conducted to detect mRNA and protein expression. RESULTS Compared with the sensitive parental cells, the A2780-CisR and SKOV3-CisR cells had increased IC50 and FXYD5 expression. FXYD5 siRNA reduced the IC50 value of cisplatin in the A2780-CisR and SKOV3-CisR cells and decreased the expression of ABCG2 (BCRP) and ABCB1 (MDR1). In addition, FXYD5 inhibition reduced the invasion and migration of the A2780-CisR and SKOV3-CisR cells, with upregulation of E-cadherin and downregulation of Snail and Vimentin. Both FXYD5 siRNA-1 and FXYD5 siRNA-2 inhibited the proliferation and promoted the apoptosis of the A2780-CisR and SKOV3-CisR cells with reduced Ki-67 and increased caspase-3. CONCLUSION FXYD5 downregulation may reduce the invasion, migration and EMT formation of EOC cells to increase their sensitivity to cisplatin chemotherapy by inhibiting cell proliferation and promoting cell apoptosis.
Collapse
Affiliation(s)
- Ya-Kun Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Ya-Jing Jia
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shi-Hao Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hong-Jie Shi
- Department of Gynecology, People's Hospital of Tang County, Baoding, Hebei, China
| | - Jing Ma
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Wang X, Lin FK, Li JR, Wang HS. A Comprehensive Risk Assessment Model for Ovarian Cancer Patients with Phospho-STAT3 and IL-31 as Immune Infiltration Relevant Genes. Onco Targets Ther 2020; 13:5617-5628. [PMID: 32606776 PMCID: PMC7305843 DOI: 10.2147/ott.s254494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/22/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Ovarian carcinoma is a malignant tumor with a high mortality rate and a lack of effective treatment options for patients at advanced stages. For improving outcomes and helping patients with poor prognosis, choose a suitable therapy and an excellent risk assessment model and new treatment options are needed. Materials and Methods Ovarian cancer gene expression profile of GSE32062 was downloaded from the NCBI GEO database for screening differentially expressed genes (DEGs) between well and poor prognosis groups using limma package in R (version 3.4.1). Prognosis-related genes and clinical prognostic factors were obtained from univariate and multivariate Cox regression analyses, and a comprehensive risk assessment model was constructed using a Pathway Dysregulation Score (PDS) matrix, Cox-Proportional Hazards (Cox-PH) regression, as well as L1-least absolute shrinkage and selection operator (L1-LASSO) penalization. Then, significant DEGs were converted to pathways and optimal prognosis-related pathways were screened. Finally, risk prediction models based on pathways, genes involved in pathways, and comprehensive clinical risk factors with pathways were built. Their prognostic functions were assessed in verification sets. Besides, genes involved in immune-pathways were checked for immune infiltration using immunohistochemistry. Results A superior risk assessment model involving 9 optimal combinations of pathways and one clinical factor was constructed. The pathway-based model was found to be superior to the gene-based model. Phospho-STAT3 (from JAK-STAT signaling pathway) and IL-31 (from DEGs) were found to be related to immune infiltration. Conclusion We have generated a comprehensive risk assessment model consisting of a clinical risk factor and pathways that showed a possible bright foreground. The set of significant pathways might play as a better prognosis model which is more accurate and applicable than the DEG set. Besides, p-STAT3 and IL-31 showing correlation to immune infiltration of ovarian cancer tissues may be potential therapeutic targets for treating ovarian cancers.
Collapse
Affiliation(s)
- Xue Wang
- Department of Obstetrics & Gynecology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China.,Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Fei-Kai Lin
- Department of Obstetrics & Gynecology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China.,Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Jia-Rui Li
- Department of Obstetrics & Gynecology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Hu-Sheng Wang
- Department of Obstetrics & Gynecology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
| |
Collapse
|
9
|
Chen X, Ding L, Kong D, Zhao X, Liao L, Zhang Y, Li F, Liu R. FXYD6 overexpression in HBV-related hepatocellular carcinoma with cirrhosis. Open Life Sci 2020; 15:259-266. [PMID: 33817214 PMCID: PMC7874599 DOI: 10.1515/biol-2020-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 11/22/2022] Open
Abstract
Objective The aim of this study was to investigate the expression of FXYD domain-containing
ion transport regulator 6 (FXYD6) mRNA and protein in hepatitis B virus
(HBV)-related hepatocellular carcinoma (HCC) tissues with cirrhosis, the
corresponding paracancerous tissues and the normal liver tissues, and to explore
the clinical significance of FXYD6 expression in HBV-related HCC with
cirrhosis. Methods The FXYD6 mRNA and protein were examined by semi-quantitative reverse
transcription polymerase chain reaction and immunohistochemistry,
respectively. Results The FXYD6 mRNA in HBV-related HCC tissues was significantly higher than that in
the cirrhosis tissues or that in the normal liver tissues. The positive expression
rate of FXYD6 protein was statistically higher in HBV-related HCC tissues than
that in HBV-related cirrhosis or that in normal liver tissues. There was no
significant correlation between the expression of FXYD6 protein and gender, age,
histological differentiation, tumor diameter, tumor number, integrity of tumor
capsule or not and alpha fetoprotein (AFP) concentration in serum, but the protein
expression was associated with microvascular invasion, pathological stage, and
early recurrence after operation within 1 year. Conclusion FXYD6 might be involved in hepatocyte carcinogenesis and tumor progression in
HBV-related HCC with cirrhosis and indicated a poor prognosis.
Collapse
Affiliation(s)
- Xiongfei Chen
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Lishuang Ding
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Deshuai Kong
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Xiulei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Lili Liao
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Yaomin Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Fengshan Li
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| | - Ruhai Liu
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, 16 Xinhuaxi Road, Yunhe District, Cangzhou, 061001, P. R. China
| |
Collapse
|
10
|
Wu Z, Liu R, Xiong L, Miao X, Li D, Zou Q, Yuan Y, Yang Z. Prognostic and Clinicopathological Significance of EphB3 and Dysadherin Expression in Extrahepatic Cholangiocarcinoma. Cancer Manag Res 2020; 12:221-232. [PMID: 32021438 PMCID: PMC6959498 DOI: 10.2147/cmar.s232278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Aim EphB3 and dysadherin are involved in tumorigenesis and progression of many neoplasms. However, the roles of EphB3 and dysadherin in extrahepatic cholangiocarcinoma (ECC) remain to be revealed. In this study, we aimed to evaluate the expression of EphB3 and dysadherin, and investigate their clinicopathological significance in ECC. Methods We examined EphB3 and dysadherin expression in 100 ECC, 30 peritumoral tissues, 10 adenoma and 15 normal biliary tract tissues using EnVision immunohistochemistry. The relationship between EphB3 or dysadherin expression and clinicopathological features was evaluated using the χ2 test or Fisher’s exact test. The overall survival of ECC patients was analyzed using Kaplan-Meier univariate survival analysis and Log rank tests. Results We found that EphB3 expression was significantly down-regulated and dysadherin expression was significantly up-regulated in ECC tissues compared with normal tissues (P < 0.01). EphB3 expression was negatively correlated with dysadherin expression in ECC (P < 0.01). The positive rate of EphB3 expression and negative rate of dysadherin expression was significantly higher in patients with well-differentiated type, no lymph node metastasis, no surrounding tissues and organs invasion, early TNM stages (I + II) and radical resection (P < 0.01). The survival of ECC patients with positive EphB3 or negative dysadherin expression was significantly longer than patients with negative EphB3 or positive dysadherin expression (P < 0.01). Cox multivariate analysis demonstrated that negative EphB3 or positive dysadherin expression were independent poor prognostic factors in ECC patients. The ROC curves suggested that EphB3 and dysadherin combined diagnostic efficacy (AUC=0.688, 95%CI: 0.603-0.772) was significantly higher EphB3 diagnostic efficacy (AUC=0.654, 95%CI: 0.564-0.743) or dysadherin diagnostic efficacy (AUC=0.648, 95%CI: 0.558-0.737) alone. Conclusion EphB3 and dysadherin are involved in the carcinogenesis and progression of ECC, and ECC patients with negative EphB3 or positive dysadherin expression have a poor prognosis.
Collapse
Affiliation(s)
- Zhengchun Wu
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Rushi Liu
- Laboratory of Medical Molecular and Immunological Diagnostics, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Xiongying Miao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Daiqiang Li
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Yuan Yuan
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Zhulin Yang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| |
Collapse
|
11
|
Bai Y, Li LD, Li J, Chen RF, Yu HL, Sun HF, Wang JY, Lu X. A FXYD5/TGF‑β/SMAD positive feedback loop drives epithelial‑to‑mesenchymal transition and promotes tumor growth and metastasis in ovarian cancer. Int J Oncol 2019; 56:301-314. [PMID: 31746425 DOI: 10.3892/ijo.2019.4911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/02/2019] [Indexed: 11/06/2022] Open
Abstract
Epithelial ovarian cancer is aggressive and lacks effective prognostic indicators or therapeutic targets. In the present study, using immunohistochemistry and bioinformatics analysis on ovarian cancer tissue data from The Obstetrics and Gynecology Hospital of Fudan University and The Cancer Genome Atlas database, it was identified that FXYD domain‑containing ion transport regulator 5 (FXYD5) expression was upregulated in the SKOV3‑IP cell line compared with its parental cell line, SKOV3, and in ovarian cancer tissues compared with in normal tissues. In addition, FXYD5 upregulation was predictive of poor patient survival. Furthermore, through various in vitro (Transwell assay, clonogenic assay and western blot analysis) and in vivo (nude mouse model) experiments, it was demonstrated that FXYD5 promoted the metastasis of ovarian cancer cells. Mechanistically, RNA sequencing, western blot analysis, a luciferase reporter assay and chromatin immunoprecipitation were performed to reveal that FXYD5 dispersed the SMAD7‑SMAD specific E3 ubiquitin protein ligase 2‑TGF‑β receptor 1 (TβR1) complex, deubiquitinated and stabilized TβR1, and subsequently enhanced transforming growth factor‑β (TGF‑β) signaling and sustained TGF‑β‑driven epithelial‑mesenchymal transition (EMT). The TGF‑β‑activated SMAD3/SMAD4 complex was in turn directly recruited to the FXYD5 promoter region, interacted with specific SMAD‑binding elements, and then promoted FXYD5 transcription. In brief, FXYD5 positively regulated TGF‑β/SMADs signaling activities, which in turn induced FXYD5 expression, creating a positive feedback loop to drive EMT in the process of ovarian cancer progression. Collectively, the findings of the present study suggested a mechanism through which FXYD5 serves a critical role in the constitutive activation of the TGF‑β/SMADs signaling pathways in ovarian cancer, and provided a promising therapeutic target for human ovarian cancer.
Collapse
Affiliation(s)
- Yang Bai
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| | - Liang-Dong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Jun Li
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| | - Rui-Fang Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| | - Hai-Lin Yu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| | - He-Fen Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200030, P.R. China
| | - Jie-Yu Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| | - Xin Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P.R. China
| |
Collapse
|
12
|
FXYD5 (Dysadherin) upregulation predicts shorter survival and reveals platinum resistance in high-grade serous ovarian cancer patients. Br J Cancer 2019; 121:584-592. [PMID: 31434988 PMCID: PMC6889357 DOI: 10.1038/s41416-019-0553-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/26/2019] [Accepted: 08/01/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND High-grade serous ovarian carcinoma (HGSOC) is generally associated with a very dismal prognosis. Nevertheless, patients with similar clinicopathological characteristics can have markedly different clinical outcomes. Our aim was the identification of novel molecular determinants influencing survival. METHODS Gene expression profiles of extreme HGSOC survivors (training set) were obtained by microarray. Differentially expressed genes (DEGs) and enriched signalling pathways were determined. A prognostic signature was generated and validated on curatedOvarianData database through a meta-analysis approach. The best prognostic biomarker from the signature was confirmed by RT-qPCR and by immunohistochemistry on an independent validation set. Cox regression model was chosen for survival analysis. RESULTS Eighty DEGs and the extracellular matrix-receptor (ECM-receptor) interaction pathway were associated to extreme survival. A 10-gene prognostic signature able to correctly classify patients with 98% of accuracy was identified. By an 'in-silico' meta-analysis, overexpression of FXYD domain-containing ion transport regulator 5 (FXYD5), also known as dysadherin, was confirmed in HGSOC short-term survivors compared to long-term ones. Its prognostic and predictive power was then successfully validated, both at mRNA and protein level, first on training than on validation sample set. CONCLUSION We demonstrated the possible involvement of FXYD5 and ECM-receptor interaction signal pathway in HCSOC survival and prognosis.
Collapse
|
13
|
Vigano S, Alatzoglou D, Irving M, Ménétrier-Caux C, Caux C, Romero P, Coukos G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front Immunol 2019; 10:925. [PMID: 31244820 PMCID: PMC6562565 DOI: 10.3389/fimmu.2019.00925] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
T cells play a critical role in cancer control, but a range of potent immunosuppressive mechanisms can be upregulated in the tumor microenvironment (TME) to abrogate their activity. While various immunotherapies (IMTs) aiming at re-invigorating the T-cell-mediated anti-tumor response, such as immune checkpoint blockade (ICB), and the adoptive cell transfer (ACT) of natural or gene-engineered ex vivo expanded tumor-specific T cells, have led to unprecedented clinical responses, only a small proportion of cancer patients benefit from these treatments. Important research efforts are thus underway to identify biomarkers of response, as well as to develop personalized combinatorial approaches that can target other inhibitory mechanisms at play in the TME. In recent years, adenosinergic signaling has emerged as a powerful immuno-metabolic checkpoint in tumors. Like several other barriers in the TME, such as the PD-1/PDL-1 axis, CTLA-4, and indoleamine 2,3-dioxygenase (IDO-1), adenosine plays important physiologic roles, but has been co-opted by tumors to promote their growth and impair immunity. Several agents counteracting the adenosine axis have been developed, and pre-clinical studies have demonstrated important anti-tumor activity, alone and in combination with other IMTs including ICB and ACT. Here we review the regulation of adenosine levels and mechanisms by which it promotes tumor growth and broadly suppresses protective immunity, with extra focus on the attenuation of T cell function. Finally, we present an overview of promising pre-clinical and clinical approaches being explored for blocking the adenosine axis for enhanced control of solid tumors.
Collapse
Affiliation(s)
- Selena Vigano
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dimitrios Alatzoglou
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Ménétrier-Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Christophe Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
14
|
Shorthouse D, Riedel A, Kerr E, Pedro L, Bihary D, Samarajiwa S, Martins CP, Shields J, Hall BA. Exploring the role of stromal osmoregulation in cancer and disease using executable modelling. Nat Commun 2018; 9:3011. [PMID: 30069015 PMCID: PMC6070494 DOI: 10.1038/s41467-018-05414-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/29/2018] [Indexed: 02/07/2023] Open
Abstract
Osmotic regulation is a vital homoeostatic process in all cells and tissues. Cells initially respond to osmotic stresses by activating transmembrane transport proteins to move osmotically active ions. Disruption of ion and water transport is frequently observed in cellular transformations such as cancer. We report that genes involved in membrane transport are significantly deregulated in many cancers, and that their expression can distinguish cancer cells from normal cells with a high degree of accuracy. We present an executable model of osmotic regulation and membrane transport in mammalian cells, providing a mechanistic explanation for phenotype change in varied disease states, and accurately predicting behaviour from single cell expression data. We also predict key proteins involved in cellular transformation, SLC4A3 (AE3), and SLC9A1 (NHE1). Furthermore, we predict and verify a synergistic drug combination in vitro, of sodium and chloride channel inhibitors, which target the osmoregulatory network to reduce cancer-associated phenotypes in fibroblasts.
Collapse
Affiliation(s)
- David Shorthouse
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Angela Riedel
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Emma Kerr
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Luisa Pedro
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Dóra Bihary
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Shamith Samarajiwa
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Carla P Martins
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Jacqueline Shields
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK.
| | - Benjamin A Hall
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK.
| |
Collapse
|
15
|
Mittal D, Sinha D, Barkauskas D, Young A, Kalimutho M, Stannard K, Caramia F, Haibe-Kains B, Stagg J, Khanna KK, Loi S, Smyth MJ. Adenosine 2B Receptor Expression on Cancer Cells Promotes Metastasis. Cancer Res 2016; 76:4372-82. [PMID: 27221704 DOI: 10.1158/0008-5472.can-16-0544] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/10/2016] [Indexed: 11/16/2022]
Abstract
Adenosine plays an important role in inflammation and tumor development, progression, and responses to therapy. We show that an adenosine 2B receptor inhibitor (A2BRi) decreases both experimental and spontaneous metastasis and combines with chemotherapy or immune checkpoint inhibitors in mouse models of melanoma and triple-negative breast cancer (TNBC) metastasis. Decreased metastasis upon A2BR inhibition is independent of host A2BR and lymphocytes and myeloid cells. Knockdown of A2BR on mouse and human cancer cells reduces their metastasis in vivo and decreases their viability and colony-forming ability, while transiently delaying cell-cycle arrest in vitro The prometastatic activity of adenosine is partly tumor A2BR dependent and independent of host A2BR expression. In humans, TNBC cell lines express higher A2BR than luminal and Her2(+) breast cancer cell lines, and high expression of A2BR is associated with worse prognosis in TNBC. Collectively, high A2BR on mouse and human tumors promotes cancer metastasis and is an ideal candidate for therapeutic intervention. Cancer Res; 76(15); 4372-82. ©2016 AACR.
Collapse
Affiliation(s)
- Deepak Mittal
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Debottam Sinha
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Deborah Barkauskas
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Arabella Young
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Murugan Kalimutho
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kimberley Stannard
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Franco Caramia
- Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, Victoria, Australia
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John Stagg
- Institut du Cancer de Montréal, Centre de Recherche du Centre Hospitalier del 'Université de Montréal, Canada
| | - Kum Kum Khanna
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Sherene Loi
- Peter MacCallum Cancer Centre, University of Melbourne, East Melbourne, Victoria, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Medicine, The University of Queensland, Herston, Queensland, Australia.
| |
Collapse
|