1
|
Harada K, Sakamoto N, Kitaoka T, Nakamura Y, Kondo R, Morisue R, Hashimoto H, Yamamoto Y, Ukai S, Maruyama R, Sakashita S, Kojima M, Tanabe K, Ohdan H, Shitara K, Kinoshita T, Ishii G, Yasui W, Ochiai A, Ishikawa S. PI3 expression predicts recurrence after chemotherapy with DNA-damaging drugs in gastric cancer. J Pathol 2025; 265:472-485. [PMID: 39980125 PMCID: PMC11880974 DOI: 10.1002/path.6400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025]
Abstract
Despite recent advances in gastric cancer therapy, chemotherapy resistance and lack of methods for selecting combination regimens remain major problems. Organoids, which provide a culture system that more closely resembles tumor cell organization than traditional cell lines, can be established from surgical specimens with a high success rate and are widely used for drug sensitivity assays. In this study, we aimed to identify a novel biomarker for predicting multidrug resistance using gastric cancer organoids (GCOs). We evaluated 5-fluorouracil or oxaliplatin-resistant GCOs to find novel biomarkers that reflect multidrug resistance in gastric cancer. To examine the resistance mechanisms, RNA-sequencing analysis and ex vivo drug sensitivity testing were performed. The association of biomarkers with patient prognosis and chemotherapy efficacy was evaluated using three original cohorts with a total of 230 cases. The results were also validated with two independent public cohorts and single-cell RNA sequence data. Increased expression of peptidase inhibitor 3 (PI3) was detected in all 5-fluorouracil or oxaliplatin-resistant GCOs. Our findings suggest a potential association of PI3 expression with ribosome biosynthesis and RNA metabolism under organoid conditions. We also found that PI3 overexpression promoted 5-fluorouracil/oxaliplatin/cisplatin resistance but not paclitaxel resistance. Immunohistochemical evaluation of PI3 expression revealed that the PI3-positive gastric cancer group had a poorer outcome, especially in terms of time to recurrence. PI3 positivity was also an independent predictor of relapse after chemotherapy with DNA-damaging agents. PI3 promotes DNA-damaging drug resistance through multiple downstream regulations related to RNA and ribosomal metabolism. PI3 may be useful as a biomarker for the therapeutic selection of non-DNA-damaging agents. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kenji Harada
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Naoya Sakamoto
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Takumi Kitaoka
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
- The Department of Pathology, Faculty of MedicineYamagata UniversityYamagataJapan
| | - Yuka Nakamura
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Ryotaro Kondo
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Ryo Morisue
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Hiroko Hashimoto
- Division of Innovative Pathology and Laboratory MedicineExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Yusuke Yamamoto
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Shoichi Ukai
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Ryota Maruyama
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Shingo Sakashita
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Motohiro Kojima
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Kazuaki Tanabe
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Takahiro Kinoshita
- Division of Gastric SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Genichiro Ishii
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
- Division of Innovative Pathology and Laboratory MedicineExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Atsushi Ochiai
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Shumpei Ishikawa
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Preventive Medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
2
|
Zhao A, Zhou S, Yang X, Lu H, Zou D, Zhang X, Liu L. Transcription factor networks and novel immune biomarkers reveal key prognostic and therapeutic insights in ovarian cancer. Discov Oncol 2025; 16:309. [PMID: 40074978 PMCID: PMC11904056 DOI: 10.1007/s12672-025-01788-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/08/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Understanding the tumor microenvironment (TME) is essential for the advancement of immunotherapy for ovarian cancer (OC). Nonetheless, predicting transcription factor (TF) regulation from the TME using single-cell RNA sequencing (scRNA-seq) data is challenging. METHODS The OC scRNA-seq data were analyzed with a specialized scRNA-seq transcriptome analysis application. The OC TME was utilized to instruct the SCENIC procedure for TF regulation. We built a risk model using Lasso regression and identified immunological subgroups using ConsensusClusterPlus. To analyze the percentage of invading immune cells, the algorithms CIBERSORT, ESTIMATE, and xCell were used. We computed the stromal score, immunological score, estimate score, and tumor purity to evaluate the risk model's capacity to predict the tumor immune microenvironment. Additionally, the expression of immunological checkpoints was examined, and for pertinent evaluation, the imvigor 210 dataset of the immunotherapy cohort was used. pRophetic predicted the sensitivity of 138 GDSC database drugs. In addition, we examined the expression of unproven risk model genes using qPCR and immunohistochemistry (JCHAIN, UBD, and RARRES1). Cell proliferation was assessed by colony formation assays. Transwell experiments were used to examine the invasion and migration ability of OC cells. RESULTS Six immunologically malignant cell subpopulations have been identified within the cancer immune microenvironment (referred to as TC0-6). Unique in its immunological profile, TC0 demonstrates the most intimate interactions with immune cells. Following a meta gene screen in the TC0 subpopulation using the top 30 targets of 14 transcription factor (TF) factors, two distinct immunological molecular subtypes-the C1 and C2 subtypes-with notable survival differences were discovered. On the basis of nine genes whose expression differs between the C1 and C2 subtypes, a risk model was constructed. The risk model is an accurate method for forecasting the effectiveness of immunotherapies, clinicopathological characteristics, and survival. JCHAIN and UBD expression in OC tissues was found to be low according to qPCR and IHC analyses, whereas RARRES1 expression was found to be high. The functional experiment results indicated that downregulation of JCHAIN and UBD and overexpression of RARRES1 could suppress the proliferation, migration, and invasion of OC cells in vitro. CONCLUSION Based on TF regulatory networks in the tumor microenvironment, this study developed a 9-gene risk model for the prognosis of ovarian cancer. This model may aid in the future promotion of personalized OC immunotherapy. In addition, JCHAIN, UBD, and RARRES1 were identified as three novel immune-related biomarkers for OC.
Collapse
Affiliation(s)
- Aiqin Zhao
- The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Sufang Zhou
- The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China.
| | - Xiaoyi Yang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Haiying Lu
- The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Dan Zou
- The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Xuan Zhang
- The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Li Liu
- The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China.
| |
Collapse
|
3
|
Zhong L, Chang W, Luo B, Gao W, He H, Fang M, Li H, Wen Z, Chen Y. Development and validation of a disulfidptosis and disulfide metabolism-related risk index for predicting prognosis in lung adenocarcinoma. Cancer Cell Int 2024; 24:2. [PMID: 38167017 PMCID: PMC10763446 DOI: 10.1186/s12935-023-03204-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Disulfidptosis is a recently proposed novel cell death mode in which cells with high SLC7A11 expression induce disulfide stress and cell death in response to glucose deficiency. The purpose of the research was to explore the function of disufidptosis and disulfide metabolism in the progression of lung adenocarcinoma (LUAD). METHODS The RNA-seq data from TCGA were divided into high/low expression group on the base of the median expression of SLC7A11, and the characteristic of differentially expressed disulfide metabolism-related genes. Least absolute shrinkage and selection operator (LASSO) algorithm was conducted the disulfidptosis and disulfide metabolism risk index. The tumor mutation burden (TMB), mechanism, pathways, tumor microenvironment (TME), and immunotherapy response were assessed between different risk groups. The role of TXNRD1 in LUAD was investigated by cytological experiments. RESULTS We established the risk index containing 5 genes. There are significant differences between different risk groups in terms of prognosis, TMB and tumor microenvironment. Additionally, the low-risk group demonstrated a higher rate of response immunotherapy in the prediction of immunotherapy response. Experimental validation suggested that the knockdown of TXNRD1 suppressed cell proliferation, migration, and invasion of LUAD. CONCLUSION Our research highlights the enormous potential of disulfidptosis and disulfide metabolism risk index in predicting the prognosis of LUAD. And TXNRD1 has great clinical translational ability.
Collapse
Affiliation(s)
- Leqi Zhong
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wuguang Chang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Bin Luo
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wuyou Gao
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Huanhuan He
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat sen University Cancer Center, Guangzhou, 510060, China
| | - Mouxiang Fang
- Institute of Biophotonics, South China Normal University, Guangzhou, China
| | - Hongmu Li
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Zhesheng Wen
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Youfang Chen
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
4
|
Wu W, Wang J, Hu Z, Zhao Y, Wang X, Bai N, Chen L, Gao P. High WFDC3 gene expression is associated with poor prognosis and reduced immune cells infiltration in pancreatic adenocarcinoma: A study using the TCGA database and bioinformatics analysis. Medicine (Baltimore) 2023; 102:e35595. [PMID: 37861515 PMCID: PMC10589585 DOI: 10.1097/md.0000000000035595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Whey-acidic-protein (WAP) four-disulfide core domain protein 3 (WFDC3) is one of the WAP family proteins. This protein family is associated with the development of solid tumors and affects the tumor immunological microenvironment. However, the prognostic value of WFDC3 in pancreatic adenocarcinoma (PAAD) and its effect on the tumor immune microenvironment is yet to be clarified. The Cancer Genome Atlas database and Genotype-Tissue Expression database were used to analyze the differential expression of WFDC3 between the tumor and adjacent tissues. The clinical significance of WFDC3 was analyzed in The Cancer Genome Atlas and International Cancer Genome Consortium database using WFDC3 transcripts and clinical information. In order to elucidate the underlying mechanisms, gene set enrichment analysis was conducted to determine potential activated pathways. Immune score evaluation and publicly available pharmacogenomics database [the Genomics of Drug Sensitivity in Cancer] were utilized to quantify immune cell infiltration and the effect on chemotherapeutic drug sensitivity. WFDC3 levels were higher in PAAD tissues than in normal pancreatic tissues. High levels of WFDC3 expression progressively increased as PAAD tumor stages progressed. Patients with elevated WFDC3 expression showed a poor prognosis. The gene set enrichment analysis analysis revealed that glutamate, arginine, and proline, and histidine metabolism levels were elevated in patients with a high WFDC3 expression phenotype. B, CD4+ T, and CD8+ T cell infiltration was diminished in PAAD tissues with elevated WFDC3 expression. According to pharmacogenomics, PAAD tissues with high WFDC3 expression are susceptible to gemcitabine. WFDC3 is highly expressed in PAAD, and patients with a high level of WFDC3 expression have a shorter overall survival time, indicating a poorer prognosis. High expression of WFDC3 may lead to the development of PAAD by affecting the amino acid metabolism and the tumor immunological microenvironment. WFDC3 may serve as a potential diagnostic and prognostic biomarker for PAAD patients.
Collapse
Affiliation(s)
- Wei Wu
- Department of General Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Jiayuan Wang
- Department of Medical Oncology, Peking University Shougang Hospital, Beijing, China
| | - Zhiping Hu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Yiguo Zhao
- Department of General Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Xin Wang
- Department of General Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Nan Bai
- Department of General Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Lei Chen
- Department of Hepatobiliary Surgery, Peking University People’s Hospital, Beijing, China
| | - Pengji Gao
- Department of General Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Chen WC, Wu CC, Liu YP, Zhuo GY, Wang YK, Chen YH, Chen CC, Wang YH, Wu MT, Wu IC. Elafin as a Prognostic Marker in Esophageal Squamous Cell Carcinoma: A Pilot Study Using Three-Dimensional Imaging and Genomic Profiling. Cancers (Basel) 2023; 15:3825. [PMID: 37568641 PMCID: PMC10417143 DOI: 10.3390/cancers15153825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Esophageal cancers are globally the sixth deadliest malignancy, with limited curative options. The association of high serum elafin levels, a molecule produced by epithelial cells, with esophageal squamous cell carcinoma (ESCC) risk is established, but its link to poor ESCC prognosis remains unclear. To explore this question, we first used three-dimensional confocal imaging to create a model of the spatial distribution of elafin inside locoregional ESCC tissues. Then, after analyzing data obtained from whole-genome microarrays for ESCC cell lines and their more invasive sublines, we performed in vitro experiments using RNA sequencing to identify possible elafin-related pathways. Three-dimensional tissue imaging showed elafin distributed as an interweaved-like fibrous structure in the stroma of tissue obtained from patients with high serum levels of elafin and poorer prognoses. By contrast, the signal was confined inside or around the tumor nest in patients who had lower serum levels and better survival. The analysis of a TCGA dataset revealed that higher levels of elafin mRNA in stage I-IIIA ESCC patients were associated with shorter survival. The in vitro studies revealed that elafin promoted ESCC cell proliferation, migration, and invasion via the epithelial-mesenchymal transition pathway. Thus, elafin inhibition could potentially be used therapeutically to improve survival in patients with locoregional ESCC.
Collapse
Affiliation(s)
- Wei-Chung Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (M.-T.W.)
| | - Chun-Chieh Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (Y.-K.W.)
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Peng Liu
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Guan-Yu Zhuo
- Institute of New Drug Development, China Medical University, Taichung 404, Taiwan;
| | - Yao-Kuang Wang
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (Y.-K.W.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Yi-Hsun Chen
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Chu-Chih Chen
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli 350, Taiwan; (C.-C.C.); (Y.-H.W.)
| | - Yin-Han Wang
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli 350, Taiwan; (C.-C.C.); (Y.-H.W.)
| | - Ming-Tsang Wu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (M.-T.W.)
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Chen Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (Y.-K.W.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
6
|
Chen T, Fan X, Li G, Meng Y. Multi-omics data integration reveals the molecular network of dysregulation IQGAP2-mTOR promotes cell proliferation. Hum Cell 2023:10.1007/s13577-023-00912-8. [PMID: 37154877 DOI: 10.1007/s13577-023-00912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023]
Abstract
IQGAP2 as a tumor suppressor gene can influence cell proliferation in multiple tumor cell lines. However, the regulation network of cell proliferation resulting solely from the deficiency of IQGAP2 in cells was still unclear. Here, we integrated transcriptome, proteome, and phosphoproteome analyses to investigate the regulatory network of cell proliferation in IQGAP2 knockdown HaCaT and HEK293 cells. Our findings revealed that the dysregulation of the IQGAP2-mTOR molecular network led to increased cell proliferation. We demonstrated that IQGAP2 knockdown enhanced the phosphorylation levels of AKT and S6K, leading to increased cell proliferation. Additionally, we found that AKT and mTOR inhibitors partially rescued abnormal cell proliferation by reducing hyperphosphorylation. Our data suggest a potential connection between the mTOR signaling pathway and aberrant cell proliferation in IQGAP2 knockdown cells. These findings offer a new therapeutic strategy for patients with IQGAP2 deficiency.
Collapse
Affiliation(s)
- Tao Chen
- Guangzhou KingMed Transformative Medicine Institute Co. Ltd., No. 10 Luoxuan 3Rd Road, Guangzhou International Biotech Island, Guangzhou, China
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Xijie Fan
- Guangzhou KingMed Transformative Medicine Institute Co. Ltd., No. 10 Luoxuan 3Rd Road, Guangzhou International Biotech Island, Guangzhou, China
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Guibin Li
- Guangzhou KingMed Transformative Medicine Institute Co. Ltd., No. 10 Luoxuan 3Rd Road, Guangzhou International Biotech Island, Guangzhou, China
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China
| | - Yuhuan Meng
- Guangzhou KingMed Transformative Medicine Institute Co. Ltd., No. 10 Luoxuan 3Rd Road, Guangzhou International Biotech Island, Guangzhou, China.
- Guangzhou KingMed Diagnostics Group Co. Ltd., Guangzhou, China.
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Arnhold J. Host-Derived Cytotoxic Agents in Chronic Inflammation and Disease Progression. Int J Mol Sci 2023; 24:ijms24033016. [PMID: 36769331 PMCID: PMC9918110 DOI: 10.3390/ijms24033016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
At inflammatory sites, cytotoxic agents are released and generated from invading immune cells and damaged tissue cells. The further fate of the inflammation highly depends on the presence of antagonizing principles that are able to inactivate these host-derived cytotoxic agents. As long as the affected tissues are well equipped with ready-to-use protective mechanisms, no damage by cytotoxic agents occurs and resolution of inflammation is initiated. However, long-lasting and severe immune responses can be associated with the decline, exhaustion, or inactivation of selected antagonizing principles. Hence, cytotoxic agents are only partially inactivated and contribute to damage of yet-unperturbed cells. Consequently, a chronic inflammatory process results. In this vicious circle of permanent cell destruction, not only novel cytotoxic elements but also novel alarmins and antigens are liberated from affected cells. In severe cases, very low protection leads to organ failure, sepsis, and septic shock. In this review, the major classes of host-derived cytotoxic agents (reactive species, oxidized heme proteins and free heme, transition metal ions, serine proteases, matrix metalloproteases, and pro-inflammatory peptides), their corresponding protective principles, and resulting implications on the pathogenesis of diseases are highlighted.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Medical Faculty, Institute of Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
8
|
Lu W, Xie B, Tan G, Dai W, Ren J, Pervaz S, Li K, Li F, Wang Y, Wang M. Elafin is related to immune infiltration and could predict the poor prognosis in ovarian cancer. Front Endocrinol (Lausanne) 2023; 14:1088944. [PMID: 36742380 PMCID: PMC9893492 DOI: 10.3389/fendo.2023.1088944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most lethal gynecologic malignancy, yet the clinical results for OC patients are still variable. Therefore, we examined how elafin expression affects the patients' prognoses and immunotherapy responses in OC, which may facilitate treatment selection and improve prognosis. METHODS The elafin mRNA expression profile was downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus. Elafin's prognostic potential and its relationship with clinical variables were investigated using Kaplan-Meier survival curves, time-dependent receiver operating characteristic curves as well as univariate and multivariate Cox regression models. As validation, protein expression in the tumor and adjacent tissues of OC patients was investigated by using immunohistochemistry (IHC). Comprehensive analyses were then conducted to explore the correlation between immune infiltration and elafin expression. RESULTS A higher mRNA expression of elafin was associated with an unfavorable prognosis in TCGA cohort and was validated in GSE31245 and IHC. Moreover, elafin was indicated as an independent risk factor for OC. A significantly higher protein expression of elafin was detected in the adjacent tissues of OC patients with shorter overall survival (OS). The immune-related pathways were mainly enriched in the high-elafin-mRNA-expression group. However, the mRNA expression of elafin was favorably correlated with indicators of the immune filtration and immunotherapy response, which also proved better immunotherapy outcomes. CONCLUSION The high elafin expression was associated with an unfavorable OS, while it also indicated better immunotherapy responses. Thus, the detection of elafin is beneficial to diagnosis and treatment selection.
Collapse
Affiliation(s)
- Weiyu Lu
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Biao Xie
- Department of Biostatistics, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Guangqing Tan
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Wanying Dai
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Jingyi Ren
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Sadaf Pervaz
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Kun Li
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Meijiao Wang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing, China
- *Correspondence: Meijiao Wang,
| |
Collapse
|
9
|
Deraison C, Bonnart C, Langella P, Roget K, Vergnolle N. Elafin and its precursor trappin-2: What is their therapeutic potential for intestinal diseases? Br J Pharmacol 2023; 180:144-160. [PMID: 36355635 PMCID: PMC10098471 DOI: 10.1111/bph.15985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/22/2022] [Accepted: 05/07/2022] [Indexed: 11/12/2022] Open
Abstract
Elafin and its precursor trappin-2 are known for their contribution to the physiological mucosal shield against luminal microbes. Such a contribution seems to be particularly relevant in the gut, where the exposure of host tissues to heavy loads of microbes is constant and contributes to mucosa-associated pathologies. The expression of trappin-2/elafin has been shown to be differentially regulated in diseases associated with gut inflammation. Accumulating evidence has demonstrated the protective effects of trappin-2/elafin in gut intestinal disorders associated with acute or chronic inflammation, or with gluten sensitization disorders. The protective effects of trappin-2/elafin in the gut are discussed in terms of their pleiotropic modes of action: acting as protease inhibitors, transglutaminase substrates, antimicrobial peptides or as a regulator of pro-inflammatory transcription factors. Further, the question of the therapeutic potential of trappin-2/elafin delivery at the intestinal mucosa surface is raised. Whether trappin-2/elafin mucosal delivery should be considered to ensure intestinal tissue repair is also discussed.
Collapse
Affiliation(s)
- Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Chrystelle Bonnart
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Philippe Langella
- Université Paris-Saclay, AgroParisTech, Micalis Institute, INRAE, Jouy-en-Josas, France
| | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Krawiec P, Pac-Kożuchowska E. Clinical Significance of Serum Elafin in Children with Inflammatory Bowel Disease. Biomedicines 2022; 10:3267. [PMID: 36552023 PMCID: PMC9775364 DOI: 10.3390/biomedicines10123267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The role of elafin in the pathophysiology of inflammatory bowel disease (IBD) has not been not elucidated. We aimed to evaluate serum elafin in children with IBD and assess its relationship with disease activity. METHODS We enrolled children with IBD in the study group and children with functional abdominal pain in the control group. We evaluated serum elafin using enzyme-linked immunosorbent assay kits. RESULTS In children with IBD, serum elafin (mean ± SD: 4.192 ± 1.424 ng/mL) was significantly elevated compared with controls (mean ± SD: 3.029 ± 1.366 ng/mL) (p = 0.0005). Elafin was significantly increased in children in the active phase of IBD (mean ± SD: 4.424 ± 1.449 ng/mL) compared with the control group (p = 0.0003). In IBD remission, only children with ulcerative colitis (mean ± SD: 4.054 ± 1.536 ng/mL) had elevated elafin compared with controls (p = 0.004). ROC analysis revealed that the area under the curve (AUC) of serum elafin was 0.809 while discriminating patients with ulcerative colitis from the control group, and the AUC was 0.664 while differentiating patients with Crohn's disease from the control group. CONCLUSIONS Serum elafin was found to be elevated in our cohort of children with IBD, depending on disease activity. Serum elafin was increased in the active phases of both ulcerative colitis and Crohn's disease, but only in the remission of ulcerative colitis. Elafin appears to be a potential candidate for a biomarker of ulcerative colitis.
Collapse
Affiliation(s)
- Paulina Krawiec
- Department of Pediatrics and Gastroenterology, Medical University of Lublin, Al. Racławickie 1, 20-059 Lublin, Poland
| | | |
Collapse
|
11
|
Zhao C, Yan S, Song Y, Xia X. Roles of Antimicrobial Peptides in Gynecological Cancers. Int J Mol Sci 2022; 23:ijms231710104. [PMID: 36077500 PMCID: PMC9456504 DOI: 10.3390/ijms231710104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 12/29/2022] Open
Abstract
Antimicrobial peptides (AMPs) are essential components of the mucosal barrier of the female reproductive tract (FRT) and are involved in many important physiological processes, including shaping the microbiota and maintaining normal reproduction and pregnancy. Gynecological cancers seriously threaten women's health and bring a heavy burden to society so that new strategies are needed to deal with these diseases. Recent studies have suggested that AMPs also have a complex yet intriguing relationship with gynecological cancers. The expression level of AMPs changes during tumor progression and they may act as promising biomarkers in cancer detection and prognosis prediction. Although AMPs have long been considered as host protective, they actually play a "double-edged sword" role in gynecological cancers, either tumorigenic or antitumor, depending on factors such as AMP and cancer types, as well as AMP concentrations. Moreover, AMPs are associated with chemoresistance and regulation of AMPs' expression may alter sensitivity of cancer cells to chemotherapy. However, more work is needed, especially on the identification of molecular mechanisms of AMPs in the FRT, as well as the clinical application of these AMPs in detection, diagnosis and treatment of gynecological malignancies.
Collapse
|
12
|
Tan M, Wang S, Li F, Xu H, Gao J, Zhu L. A methylation-driven genes prognostic signature and the immune microenvironment in epithelial ovarian cancer. Carcinogenesis 2022; 43:635-646. [PMID: 35639961 DOI: 10.1093/carcin/bgac048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant gene methylation has been implicated in the development and progression of tumors. In this study, we aimed to identity methylation driven genes involved in epithelial ovarian cancer (EOC) to establish a prognostic signature for patients with EOC. We identified and verified 6 MDGs that are closely related to the prognosis of ovarian cancer. A prognostic risk score model and nomogram for predicting the prognosis of ovarian cancer were constructed based on the six MDGs. It can also effectively reflect the immune environment and immunotherapy response of ovarian cancer. These MDGs have great significance to the implementation of individualized treatment and disease monitoring of ovarian cancer patients.
Collapse
Affiliation(s)
- Mingzi Tan
- Department of Gynecology, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, P R China.,Department of Gynecology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, P R China
| | - Shengtan Wang
- Department of Gynecology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570011, P.R. China
| | - Feifei Li
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Haoya Xu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, P.R. China
| | - Jian Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, P.R. China
| | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, P.R. China
| |
Collapse
|
13
|
A bioinformatic analysis of WFDC2 (HE4) expression in high grade serous ovarian cancer reveals tumor-specific changes in metabolic and extracellular matrix gene expression. Med Oncol 2022; 39:71. [PMID: 35568777 PMCID: PMC9107348 DOI: 10.1007/s12032-022-01665-4] [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: 10/27/2021] [Accepted: 01/22/2022] [Indexed: 10/31/2022]
Abstract
Human epididymis protein-4 (HE4/WFDC2) has been well-studied as an ovarian cancer clinical biomarker. To improve our understanding of its functional role in high grade serous ovarian cancer, we determined transcriptomic differences between ovarian tumors with high- versus low-WFDC2 mRNA levels in The Cancer Genome Atlas dataset. High-WFDC2 transcript levels were significantly associated with reduced survival in stage III/IV serous ovarian cancer patients. Differential expression and correlation analyses revealed secretory leukocyte peptidase inhibitor (SLPI/WFDC4) as the gene most positively correlated with WFDC2, while A kinase anchor protein-12 was most negatively correlated. WFDC2 and SLPI were strongly correlated across many cancers. Gene ontology analysis revealed enrichment of oxidative phosphorylation in differentially expressed genes associated with high-WFDC2 levels, while extracellular matrix organization was enriched among genes associated with low-WFDC2 levels. Immune cell subsets found to be positively correlated with WFDC2 levels were B cells and plasmacytoid dendritic cells, while neutrophils and endothelial cells were negatively correlated with WFDC2. Results were compared with DepMap cell culture gene expression data. Gene ontology analysis of k-means clustering revealed that genes associated with low-WFDC2 were also enriched in extracellular matrix and adhesion categories, while high-WFDC2 genes were enriched in epithelial cell proliferation and peptidase activity. These results support previous findings regarding the effect of HE4/WFDC2 on ovarian cancer pathogenesis in cell lines and mouse models, while adding another layer of complexity to its potential functions in ovarian tumor tissue. Further experimental explorations of these findings in the context of the tumor microenvironment are merited.
Collapse
|
14
|
Kim YJ, Zhao Y, Myung JK, Yi JM, Kim MJ, Lee SJ. Suppression of breast cancer progression by FBXL16 via oxygen-independent regulation of HIF1α stability. Cell Rep 2021; 37:109996. [PMID: 34818544 DOI: 10.1016/j.celrep.2021.109996] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/06/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancers (TNBCs) are characterized by high rates of recurrence and poor clinical outcomes. Deregulated E3 ligases are involved in breast cancer pathogenesis and progression, but the underlying mechanisms are unclear. Here, we find that F-box and leucine-rich repeat protein 16 (FBXL16) acts as a tumor suppressor in TNBCs. FBXL16 directly binds to HIF1α and induces its ubiquitination and degradation, regardless of the tumor microenvironment, resulting in blockade of the HIF1α-mediated epithelial-mesenchymal transition (EMT) and angiogenesis features of breast cancer. In TNBCs, FBXL16 expression is downregulated by the p38/miR-135b-3p axis, and loss of FBXL16 expression restores HIF1α-mediated metastatic features of breast cancer. Low expression of FBXL16 is associated with high-grade and lymph node-positive tumors and poor overall survival of breast cancer. Taken together, these findings demonstrate that modulation of FBXL16 expression may offer a favorable strategy for treatment of patients with metastatic breast cancer, including TNBCs.
Collapse
Affiliation(s)
- Yeon-Ju Kim
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Yi Zhao
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Jae Kyung Myung
- Department of Pathology, Hanyang University Medical Center, 222-1, Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Joo Mi Yi
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, South Korea
| | - Min-Jung Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
15
|
Zhang H, Wu Y, Li H, Sun L, Meng X. Model constructions of chemosensitivity and prognosis of high grade serous ovarian cancer based on evaluation of immune microenvironment and immune response. Cancer Cell Int 2021; 21:593. [PMID: 34736480 PMCID: PMC8567582 DOI: 10.1186/s12935-021-02295-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prognosis of high grade serous ovarian cancer (HGSOC) patients is closely related to the immune microenvironment and immune response. Based on this, the purpose of this study was to construct a model to predict chemosensitivity and prognosis, and provide novel biomarkers for immunotherapy and prognosis evaluation of HGSOC. METHODS GSE40595 (38 samples), GSE18520 (63 samples), GSE26712 (195 samples), TCGA (321 samples) and GTEx (88 samples) were integrated to screen differential expressed genes (DEGs) of HGSOC. The prognosis related DEGs (DEPGs) were screened through overall survival analysis. The DEGs-encoded protein-protein interaction network was constructed and hub genes of DEPGs (DEPHGs) were generated by STRING. Immune characteristics of the samples were judged by ssGSEA, ESTIMATE and CYBERSORT. TIMER was used to analyze the relationship between DEPHGs and tumor-infiltrating immunocytes, as well as the immune checkpoint genes, finally immune-related DEPHGs (IDEPHGs) were determined, and whose expression in 12 pairs of HGSOC tissues and tumor-adjacent tissues were analyzed by histological verification. Furthermore, the chemosensitivity genes in IDEPHGs were screened according to GSE15622 (n = 65). Finally, two prediction models of paclitaxel sensitivity score (PTX score) and carboplatin sensitivity score (CBP score) were constructed by lasso algorithm. The area under curve was calculated to estimate the accuracy of candidate gene models in evaluating chemotherapy sensitivity. RESULTS 491 DEGs were screened and 37 DEGs were identified as DEPGs, and 11 DEPHGs were further identified. Among them, CXCL13, IDO1, PI3, SPP1 and TRIM22 were screened as IDEPHGs and verified in the human tissues. Further analysis showed that IDO1, PI3 and TRIM22 could independently affect the chemotherapy sensitivity of HGSOC patients. The PTX score was significantly better than TRIM22, PI3, SPP1, IDO1 and CXCL13 in predicting paclitaxel sensitivity, so was CBP score in predicting carboplatin sensitivity. What's more, both of the HGSOC patients with high PTX score or high CBP score had longer survival time. CONCLUSIONS Five IDEPHGs identified through comprehensive bioinformatics analysis were closely related with the prognosis, immune microenvironment and chemotherapy sensitivity of HGSOC. Two prediction models based on IDEPHGs might have potential application of chemotherapy sensitivity and prognosis for patients with HGSOC.
Collapse
Affiliation(s)
- Han Zhang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yijun Wu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Hao Li
- Department of Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiangkai Meng
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
16
|
Regner MJ, Wisniewska K, Garcia-Recio S, Thennavan A, Mendez-Giraldez R, Malladi VS, Hawkins G, Parker JS, Perou CM, Bae-Jump VL, Franco HL. A multi-omic single-cell landscape of human gynecologic malignancies. Mol Cell 2021; 81:4924-4941.e10. [PMID: 34739872 PMCID: PMC8642316 DOI: 10.1016/j.molcel.2021.10.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/05/2021] [Accepted: 10/13/2021] [Indexed: 01/05/2023]
Abstract
Deconvolution of regulatory mechanisms that drive transcriptional programs in cancer cells is key to understanding tumor biology. Herein, we present matched transcriptome (scRNA-seq) and chromatin accessibility (scATAC-seq) profiles at single-cell resolution from human ovarian and endometrial tumors processed immediately following surgical resection. This dataset reveals the complex cellular heterogeneity of these tumors and enabled us to quantitatively link variation in chromatin accessibility to gene expression. We show that malignant cells acquire previously unannotated regulatory elements to drive hallmark cancer pathways. Moreover, malignant cells from within the same patients show substantial variation in chromatin accessibility linked to transcriptional output, highlighting the importance of intratumoral heterogeneity. Finally, we infer the malignant cell type-specific activity of transcription factors. By defining the regulatory logic of cancer cells, this work reveals an important reliance on oncogenic regulatory elements and highlights the ability of matched scRNA-seq/scATAC-seq to uncover clinically relevant mechanisms of tumorigenesis in gynecologic cancers.
Collapse
Affiliation(s)
- Matthew J. Regner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,These authors contributed equally
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,These authors contributed equally
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Aatish Thennavan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Oral and Craniofacial Biomedicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Raul Mendez-Giraldez
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Venkat S. Malladi
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Gabrielle Hawkins
- Division of Gynecology Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Victoria L. Bae-Jump
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Division of Gynecology Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hector L. Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Lead contact.,Correspondence:
| |
Collapse
|
17
|
K-RAS Acts as a Critical Regulator of CD44 to Promote the Invasiveness and Stemness of GBM in Response to Ionizing Radiation. Int J Mol Sci 2021; 22:ijms222010923. [PMID: 34681583 PMCID: PMC8539357 DOI: 10.3390/ijms222010923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/21/2022] Open
Abstract
Radiation therapy is a current standard-of-care treatment and is used widely for GBM patients. However, radiation therapy still remains a significant barrier to getting a successful outcome due to the therapeutic resistance and tumor recurrence. Understanding the underlying mechanisms of this resistance and recurrence would provide an efficient approach for improving the therapy for GBM treatment. Here, we identified a regulatory mechanism of CD44 which induces infiltration and mesenchymal shift of GBM. Ionizing radiation (IR)-induced K-RAS/ERK signaling activation elevates CD44 expression through downregulation of miR-202 and miR-185 expression. High expression of CD44 promotes SRC activation to induce cancer stemness and EMT features of GBM cells. In this study, we demonstrate that the K-RAS/ERK/CD44 axis is a key mechanism in regulating mesenchymal shift of GBM cells after irradiation. These findings suggest that blocking the K-RAS activation or CD44 expression could provide an efficient way for GBM treatment.
Collapse
|
18
|
Luna C, Parker M, Challa P, Gonzalez P. Long-Term Decrease of Intraocular Pressure in Rats by Viral Delivery of miR-146a. Transl Vis Sci Technol 2021; 10:14. [PMID: 34254987 PMCID: PMC8287046 DOI: 10.1167/tvst.10.8.14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Purpose To evaluate the effects of miR-146a in trabecular meshwork (TM) cells and on intraocular pressure (IOP) in vivo via viral delivery of miR-146a to the anterior chamber of rat eyes. Methods Human TM cells were transfected with miR-146 mimic or inhibitor. Some cells from each group were then subjected to cyclic mechanical stress (CMS). Other cells from each group had no force applied. Gene expression was then analyzed by quantitative polymerase chain reaction (qPCR). Replication-deficient adenovirus and lentivirus expressing miR-146a were inoculated into the anterior segment of Brown Norway rat eyes. IOP was monitored by rebound tonometry, visual acuity was evaluated by optokinetic tracking (OKT), and inflammation markers in the anterior segment were examined by slit-lamp, qPCR, and semi-thin sections. Results miR-146 affected the expression of genes potentially involved in outflow homeostasis at basal levels and under CMS. Both lentiviral and adenoviral vectors expressing miR-146a resulted in sustained decreases in IOP ranging from 2.6 to 4.4 mmHg. Long term follow-up of rats injected with lentiviral vectors showed a sustained effect on IOP of 4.4 ± 2.9 mmHg that lasted until rats were sacrificed more than 8 months later. Eyes showed no signs of inflammation, loss of visual acuity, or other visible abnormalities. Conclusions Intracameral delivery of miR-146a can provide a long-term decrease of IOP in rats without signs of inflammation or other visible adverse effects. Transitional Relevance The IOP-lowering effects of miR-146 observed in rats provides a necessary step toward the development of an effective gene therapy for glaucoma in humans.
Collapse
|
19
|
Zhang C, Hu H, Wang X, Zhu Y, Jiang M. WFDC Protein: A Promising Diagnosis Biomarker of Ovarian Cancer. J Cancer 2021; 12:5404-5412. [PMID: 34405003 PMCID: PMC8364637 DOI: 10.7150/jca.57880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
An initial diagnosis of cancer is usually based on symptoms, abnormal physical examination and imaging tests. Ovarian cancer is difficult to be diagnosed timely due to the nonspecific symptoms, thus resulting in the high-risk mortality. Despite of the various diagnostic methods, there is still no reliable diagnostic test. Clinically, carbohydrate antigen 125(CA125) is widely recognized as a diagnosis biomarker of ovary cancer. However, CA125 is not sensitive to detect the ovary cancer at the early stage. It is essential to explore other potential biomarkers. Human epididymis protein 4 (HE4) in the whey/four-disulfide core (WFDC) proteins family shows satisfactory sensitivity in the early diagnosis of ovary cancer. In this present review, we summarized the important effects of WFDC family proteins on the proliferation, apoptosis and migration of ovary cancer and intended to provide more evidence to explore the possibility of WFDC protein as a diagnosis biomarker.
Collapse
Affiliation(s)
- Chen Zhang
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haoyue Hu
- Lung Cancer Center, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoyan Wang
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yajuan Zhu
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ming Jiang
- West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
20
|
Wu IC, Wang YK, Chen YH, Wu CC, Wu MC, Chen WC, Wang WL, Lin HS, Chen CC, Chou SH, Liu YP, Wu MT. High Serum Elafin Prediction of Poor Prognosis of Locoregional Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13123082. [PMID: 34205756 PMCID: PMC8233752 DOI: 10.3390/cancers13123082] [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: 05/14/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Conventional serum markers such as carcinoembryonic antigen (CEA), squamous cell carcinoma antigen (SCC), and tissue polypeptide antigen (TPA) have a low sensitivity in predicting the prognosis of locoregional esophageal squamous cell carcinoma cell (ESCC). In our clinical study, we found high serum elafin to be an independent outcome predictor for stage I-IIIA ESCC, considering T, N, overall stage, and treatment. In vitro experiments showed that adding recombinant elafin drove ESCC cell proliferation, migration and invasion, while shRNA attenuated elafin levels, abrogating those effects. Our results suggested serum elafin might be a noninvasive biomarker to predict the outcome of locoregional ESCC and could potentially be used as a therapeutic target. Abstract Esophageal squamous cell carcinoma (ESCC) is a highly aggressive tumor known to have locally advanced and metastatic features which cause a dismal prognosis. We sought to determine whether elafin, a non-invasive and secretory small-molecule marker, could be used to predict prognosis in locoregional ESCC patients in human and in vitro studies. In our human study, 119 subjects were identified as having incident and pathologically-proved ESCC with stage I-IIIA tumors from southern Taiwan between 2000 and 2016. We measured their serum elafin levels at baseline and followed them until the date of cancer death or until January 2020, the end of this study. Those with high serum elafin levels were found to have a 1.99-fold risk (95% confidence interval: 1.17–3.38) shorter survival than those who did not. In our in vitro experiments, elevated elafin levels were found to drive ESCC cell proliferation, migration and invasion, while attenuation of elafin level by shRNA abrogated those effects. We concluded that elafin promotes ESCC motility and invasion and leads to a worse clinical prognosis in ESCC patients without distant metastasis.
Collapse
Affiliation(s)
- I-Chen Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (I.-C.W.); (Y.-K.W.); (Y.-H.C.)
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
| | - Yao-Kuang Wang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (I.-C.W.); (Y.-K.W.); (Y.-H.C.)
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
| | - Yi-Hsun Chen
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (I.-C.W.); (Y.-K.W.); (Y.-H.C.)
| | - Chun-Chieh Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Meng-Chieh Wu
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 807, Taiwan;
| | - Wei-Chung Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wen-Lun Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-DA Hospital/I-Shou University, Kaohsiung 824, Taiwan;
| | - Hung-Shun Lin
- Department of Laboratory Medicine & Department of Research, Education & Training, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Public Health, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chou-Cheng Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
| | - Shah-Hwa Chou
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
- Division of Chest Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Peng Liu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (Y.-P.L.); (M.-T.W.); Tel.: +886-7-3121101 (ext. 5092-424) (Y.-P.L.); +886-7-3121101 (ext. 2315) (M.-T.W.)
| | - Ming-Tsang Wu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Public Health, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Rapid Screening Research Center for Toxicology and Biomedicine, National Sun Yat-Sen University, Kaohsiung 807, Taiwan
- Correspondence: (Y.-P.L.); (M.-T.W.); Tel.: +886-7-3121101 (ext. 5092-424) (Y.-P.L.); +886-7-3121101 (ext. 2315) (M.-T.W.)
| |
Collapse
|
21
|
Abstract
Biomarkers factor into the diagnosis and treatment of almost every patient with cancer. The innovation in proteomics follows improvement of mass spectrometry techniques and data processing strategy. Recently, proteomics and typical biological studies have been the answer for clinical applications. The clinical proteomics techniques are now actively adapted to protein identification in large patient cohort, biomarker development for more sensitive and specific screening based on quantitative data. And, it is important for clinical, translational researchers to be acutely aware of the issues surrounding appropriate biomarker development, in order to facilitate entry of clinically useful biomarkers into the clinic. Here, we discuss in detail include the case research for clinical proteomics. Furthermore, we give an overview on the current developments and novel findings in proteomics-based cancer biomarker research.
Collapse
|
22
|
Wang C, Liao Y, He W, Zhang H, Zuo D, Liu W, Yang Z, Qiu J, Yuan Y, Li K, Zhang Y, Wang Y, Shi Y, Qiu Y, Gao S, Yuan Y, Li B. Elafin promotes tumour metastasis and attenuates the anti-metastatic effects of erlotinib via binding to EGFR in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:113. [PMID: 33771199 PMCID: PMC7995733 DOI: 10.1186/s13046-021-01904-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Background Elafin is a serine protease inhibitor critical for host defence. We previously reported that Elafin was associated with the recurrence of early-stage hepatocellular carcinoma (HCC) after surgery. However, the exact role of Elafin in HCC remains obscure. Methods HCC tissue microarrays were used to investigate the correlation between Elafin expression and the prognosis of HCC patients. In vitro migration, invasion and wound healing assays and in vivo lung metastasis models were used to determine the role of Elafin in HCC metastasis. Mass spectrometry, co-immunoprecipitation, western blotting, and immunofluorescence staining assays were performed to uncover the mechanism of Elafin in HCC. Dual-luciferase reporter and chromatin immunoprecipitation assays were employed to observe the transcriptional regulation of Elafin. Results Elafin expression was frequently increased in HCC tissues compared to normal tissues, and high Elafin expression in HCC tissues was correlated with aggressive tumour phenotypes and a poor prognosis in HCC patients. Elafin dramatically enhanced the metastasis of HCC cells both in vitro and in vivo by interacting with EGFR and activating EGFR/AKT signalling. Moreover, Elafin attenuated the suppressive effects of erlotinib on HCC metastasis. Besides, Elafin was transcriptionally regulated by Sp1 in HCC cells. Clinically, Elafin expression was positively correlated with Sp1, Vimentin, and EGFR signalling in both our HCC tissue microarrays and TCGA database analysis. Conclusions Upregulation of Elafin by Sp1 enhanced HCC metastasis via EGFR/AKT pathway, and overexpression of Elafin attenuated the anti-metastatic effects of erlotinib, suggesting a valuable prognostic biomarker and therapeutic target for HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01904-y.
Collapse
Affiliation(s)
- Chenwei Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yadi Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Wei He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Hong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Dinglan Zuo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Wenwu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Zhiwen Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yichuan Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Kai Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yuanping Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yongjin Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yunxing Shi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yuxiong Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Song Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China. .,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| | - Binkui Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China. .,Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
23
|
Longatto-Filho A, Fregnani JH, Mafra da Costa A, de Araujo-Souza PS, Scapulatempo-Neto C, Herbster S, Boccardo E, Termini L. Evaluation of Elafin Immunohistochemical Expression as Marker of Cervical Cancer Severity. Acta Cytol 2020; 65:165-174. [PMID: 33271565 DOI: 10.1159/000512010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The main risk factor for the development of cervical cancer (CC) is persistent infection by human papillomavirus (HPV) oncogenic types. In order to persist, HPV exhibits a plethora of immune evasion mechanisms. PI3/Elafin (Peptidase Inhibitor 3) is an endogenous serine protease inhibitor involved in epithelial protection against pathogens. PI3/Elafin's role in CC is still poorly understood. MATERIALS AND METHODS In the present study, we addressed PI3/Elafin protein detection in 123 CC samples by immunohistochemistry and mRNA expression in several datasets available at Gene Expression Omnibus and The Cancer Genome Atlas platforms. RESULTS We observed that PI3/Elafin is consistently downregulated in CC samples when compared to normal tissue. Most of PI3/Elafin-positive samples exhibited this protein at the plasma membrane. Besides, high PI3/Elafin expression at the cellular membrane was more frequent in in situ stages I + II than in invasive cervical tumor stages III + IV. This indicates that PI3/Elafin expression is gradually lost during the CC progression. Of note, advanced stages of CC were more frequently associated with a more intense PI3/Elafin reaction in the nuclei and cytoplasm. CONCLUSION Our results suggest that PI3/Elafin levels and subcellular localization may be used as a biomarker for CC severity.
Collapse
Affiliation(s)
- Adhemar Longatto-Filho
- Laboratory of Medical Investigation (LIM) 14, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, ICVS/3B's, Braga, Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Pio XII Foundation, Barretos, Brazil
| | | | - Allini Mafra da Costa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Pio XII Foundation, Barretos, Brazil
- Cancer Surveillance Section, International Agency for Research on Cancer, Lyon, France
| | - Patricia Savio de Araujo-Souza
- Laboratory of Immunogenetics and Histocompatibility, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
| | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Pio XII Foundation, Barretos, Brazil
- Pathology and Molecular Diagnostics (GeneOne), DASA Laboratories, São Paulo, São Paulo, Brazil
| | - Suellen Herbster
- Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Enrique Boccardo
- Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Lara Termini
- Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, São Paulo, Brazil,
| |
Collapse
|
24
|
Zheng M, Mullikin H, Hester A, Czogalla B, Heidegger H, Vilsmaier T, Vattai A, Chelariu-Raicu A, Jeschke U, Trillsch F, Mahner S, Kaltofen T. Development and Validation of a Novel 11-Gene Prognostic Model for Serous Ovarian Carcinomas Based on Lipid Metabolism Expression Profile. Int J Mol Sci 2020; 21:E9169. [PMID: 33271935 PMCID: PMC7731240 DOI: 10.3390/ijms21239169] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/06/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Biomarkers might play a significant role in predicting the clinical outcomes of patients with ovarian cancer. By analyzing lipid metabolism genes, future perspectives may be uncovered; (2) Methods: RNA-seq data for serous ovarian cancer were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases. The non-negative matrix factorization package in programming language R was used to classify molecular subtypes of lipid metabolism genes and the limma package in R was performed for functional enrichment analysis. Through lasso regression, we constructed a multi-gene prognosis model; (3) Results: Two molecular subtypes were obtained and an 11-gene signature was constructed (PI3, RGS, ADORA3, CH25H, CCDC80, PTGER3, MATK, KLRB1, CCL19, CXCL9 and CXCL10). Our prognostic model shows a good independent prognostic ability in ovarian cancer. In a nomogram, the predictive efficiency was notably superior to that of traditional clinical features. Related to known models in ovarian cancer with a comparable amount of genes, ours has the highest concordance index; (4) Conclusions: We propose an 11-gene signature prognosis prediction model based on lipid metabolism genes in serous ovarian cancer.
Collapse
Affiliation(s)
- Mingjun Zheng
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Heather Mullikin
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Anna Hester
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Helene Heidegger
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Theresa Vilsmaier
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Aurelia Vattai
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Till Kaltofen
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| |
Collapse
|
25
|
Zhao Y, Kaushik N, Kang JH, Kaushik NK, Son SH, Uddin N, Kim MJ, Kim CG, Lee SJ. A Feedback Loop Comprising EGF/TGFα Sustains TFCP2-Mediated Breast Cancer Progression. Cancer Res 2020; 80:2217-2229. [PMID: 32193292 DOI: 10.1158/0008-5472.can-19-2908] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/04/2020] [Accepted: 03/12/2020] [Indexed: 11/16/2022]
Abstract
Stemness and epithelial-mesenchymal transition (EMT) are two fundamental characteristics of metastasis that are controlled by diverse regulatory factors, including transcription factors. Compared with other subtypes of breast cancer, basal-type or triple-negative breast cancer (TNBC) has high frequencies of tumor relapse. However, the role of alpha-globin transcription factor CP2 (TFCP2) has not been reported as an oncogenic driver in those breast cancers. Here, we show that TFCP2 is a potent factor essential for EMT, stemness, and metastasis in breast cancer. TFCP2 directly bound promoters of EGF and TGFα to regulate their expression and stimulate autocrine signaling via EGFR. These findings indicate that TFCP2 is a new antimetastatic target and reveal a novel regulatory mechanism in which a positive feedback loop comprising EGF/TGFα and AKT can control malignant breast cancer progression. SIGNIFICANCE: TFCP2 is a new antimetastatic target that controls TNBC progression via a positive feedback loop between EGF/TGFα and the AKT signaling axis.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Neha Kaushik
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Jae-Hyeok Kang
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul, South Korea
| | - Seung Han Son
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Nizam Uddin
- Center for Cell Analysis & Modeling, University of Connecticut Health Center, Farmington, Connecticut
| | - Min-Jung Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Chul Geun Kim
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea.
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea.
| |
Collapse
|
26
|
Kengkarn S, Petmitr S, Boonyuen U, Reamtong O, Poomsawat S, Sanguansin S. Identification of Novel Candidate Biomarkers for Oral Squamous Cell Carcinoma Based on Whole Gene Expression Profiling. Pathol Oncol Res 2020; 26:2315-2325. [PMID: 32468250 DOI: 10.1007/s12253-020-00828-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/23/2020] [Accepted: 05/20/2020] [Indexed: 12/25/2022]
Abstract
This study aimed to determine the whole gene expression profiles and to ascertain potential biomarkers for 22 oral squamous cell carcinoma (OSCC) among Thai patients using the Illumina Human HT-12, V4.0 Expression BeadChip array. Result indicated 2,724 differential expressed genes composed of 1,560 up-regulated and 1,164 down-regulated genes (unpaired t-test, p-value <0.05; fold change ≥2.0 and ≤2.0). The top 9 up-regulated genes were validated in 39 OSCC cases using TaqMan real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assay. Among these, the up-regulation of peptidase inhibitor 3 (PI3) and keratin 17 (KRT17) genes was harbored in all 39 OSCC patients (100%). Likewise, statistical analysis indicated that gene expression in 8 selective genes including keratin 16 (KRT16), keratin 14 (KRT14), keratinocyte differentiation-associated protein (KRTDAP), keratin 6B (KRT6B), PI3, S100 calcium binding protein A7 (S100A7), stratifin (SFN) and keratin 5 (KRT5) was significantly associated with well differentiated OSCC (p-value <0.05). Moreover, high level of KRT17 protein was significantly associated with well differentiated OSCC compared to moderately OSCC (p-value = 0.041). Notably, using nested-PCR analysis indicated all OSCC cases in this study were HPV-free. Especially, KRTDAP, PI3, SFN mRNA expression were first reported among patients with OSCC. Conclusion, the whole transcript expression study and TaqMan real-time qRT-PCR assay were relevant regarding the increase in gene expression in OSCC. In addition, the up-regulation of PI3 and KRT17 might constitute potential candidate molecular biomarkers to diagnose patients with OSCC.
Collapse
Affiliation(s)
- Sudaporn Kengkarn
- Department of Molecular Tropical Medicine & Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Songsak Petmitr
- Department of Molecular Tropical Medicine & Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine & Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine & Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sopee Poomsawat
- Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Sirima Sanguansin
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
27
|
Rahman MM, Lai YC, Husna AA, Chen HW, Tanaka Y, Kawaguchi H, Hatai H, Miyoshi N, Nakagawa T, Fukushima R, Miura N. Transcriptome analysis of dog oral melanoma and its oncogenic analogy with human melanoma. Oncol Rep 2019; 43:16-30. [PMID: 31661138 PMCID: PMC6908934 DOI: 10.3892/or.2019.7391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022] Open
Abstract
Dogs have been considered as an excellent immunocompetent model for human melanoma due to the same tumor location and the common clinical and pathological features with human melanoma. However, the differences in the melanoma transcriptome between the two species have not been yet fully determined. Considering the role of oncogenes in melanoma development, in this study, we first characterized the transcriptome in canine oral melanoma and then compared the transcriptome with that of human melanoma. The global transcriptome from 8 canine oral melanoma samples and 3 healthy oral tissues were compared by RNA-Seq followed by RT-qPCR validation. The results revealed 2,555 annotated differentially expressed genes, as well as 364 novel differentially expressed genes. Dog chromosomes 1 and 9 were enriched with downregulated and upregulated genes, respectively. Along with 10 significant transcription site binding motifs; the NF-κB and ATF1 binding motifs were the most significant and 4 significant unknown motifs were indentified among the upregulated differentially expressed genes. Moreover, it was found that canine oral melanoma shared >80% significant oncogenes (upregulated genes) with human melanoma, and JAK-STAT was the most common significant pathway between the species. The results identified a 429 gene signature in melanoma, which was up-regulated in both species; these genes may be good candidates for therapeutic development. Furthermore, this study demonstrates that as regards oncogene expression, human melanoma contains an oncogene group that bears similarities with dog oral melanoma, which supports the use of dogs as a model for the development of novel therapeutics and experimental trials before human application.
Collapse
Affiliation(s)
- Md Mahfuzur Rahman
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Yu-Chang Lai
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Al Asmaul Husna
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Hui-Wen Chen
- Joint Graduate School of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Yuiko Tanaka
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113‑8657, Japan
| | - Hiroaki Kawaguchi
- Hygiene and Health Promotion Medicine, Kagoshima University Graduate School of Medicine and Dental Science, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Hitoshi Hatai
- Department of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Noriaki Miyoshi
- Department of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| | - Takayuki Nakagawa
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113‑8657, Japan
| | - Ryuji Fukushima
- Animal Medical Center, Tokyo University of Agriculture and Technology, Tokyo 183‑8538, Japan
| | - Naoki Miura
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890‑0065, Japan
| |
Collapse
|
28
|
High Expression and Clinical Significance of Elafin in Colorectal Cancer. Gastroenterol Res Pract 2019; 2019:4946824. [PMID: 31281349 PMCID: PMC6590570 DOI: 10.1155/2019/4946824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/26/2019] [Accepted: 05/13/2019] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The decrease of Elafin is associated with several inflammatory diseases. Exogenous Elafin may be a treatment for IBD. Little data has shown the expression of Elafin in patients of colorectal cancer. Here, we tried to explore Elafin expression in human tissues of colorectal cancer. METHODS We examined the protein expression of Elafin in human tissues of adjacent nontumor and colorectal tumor by immunohistochemistry (IHC) or quantitative real-time polymerase chain reaction (qRT-PCR), then analyzed the clinical and RNA-seq data presented in The Cancer Genome Atlas (TCGA) database to confirm the relationship between Elafin levels and colorectal tumor. RESULTS Of the 88 paired samples, 68 colorectal cancer tissues indicated a high expression of Elafin compared with 52 matched adjacent noncancerous tissues. And the mRNA levels of Elafin in 35 paired tissues showed a similar trend. The RNA-seq and clinical data were available in 438 colorectal cancer tissues and 41 normal tissues in TCGA database. The RNA-seq data showed that Elafin mRNA was upregulated about twofold in colorectal cancer samples as compared to adjacent noncancerous samples (176.42 ± 402.13 vs. 96.75 ± 150.07; P = 0.208). No statistically significant correlation was found between the Elafin expression and the age, gender, tumor invasive stage, lymph node metastasis, and distant metastasis both at the protein and mRNA levels. However, the Elafin expression was correlated with clinical stage based on the AJCC guidelines at protein levels but not mRNA levels. CONCLUSIONS Elafin was upregulated in patients of colorectal cancer, resulting to potential limitations for exogenous Elafin treatment.
Collapse
|
29
|
Gockley AA, Elias KM. Fallopian tube tumorigenesis and clinical implications for ovarian cancer risk-reduction. Cancer Treat Rev 2018; 69:66-71. [DOI: 10.1016/j.ctrv.2018.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/04/2018] [Accepted: 06/09/2018] [Indexed: 12/20/2022]
|
30
|
FBXL14 abolishes breast cancer progression by targeting CDCP1 for proteasomal degradation. Oncogene 2018; 37:5794-5809. [PMID: 29973690 DOI: 10.1038/s41388-018-0372-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/16/2018] [Accepted: 05/29/2018] [Indexed: 01/10/2023]
Abstract
Understanding the molecular mechanisms that underlie the aggressive behavior and relapse of breast cancer may help in the development of novel therapeutic interventions. CUB-domain-containing protein 1 (CDCP1), a transmembrane adaptor protein, is highly maintained and required in the context of cellular metastatic potential in triple-negative breast cancer (TNBC). For this reason, gene expression levels of CDCP1 have been considered as a prognostic marker in TNBC. However, not rarely, transcript levels of genes do not reflect always the levels of proteins, due to the post-transcriptional regulation. Here we show that miR-17/20a control the FBXL14 E3 ligase, establishing FBXL14 as an upstream regulator of the CDCP1 pathway. FBXL14 acts as an novel interaction partner of CDCP1, and facilitates its ubiquitination and proteasomal degradation with an enhanced capacity to suppress CDCP1 protein stability that eventually prevents CDCP1 target genes involved in breast cancer metastasis. Our findings first time uncovers the regulatory mechanism of CDCP-1 protein stabilization, more predictable criteria than gene expression levels for prognosis of breast cancer patients.
Collapse
|
31
|
Butera G, Pacchiana R, Donadelli M. Autocrine mechanisms of cancer chemoresistance. Semin Cell Dev Biol 2017; 78:3-12. [PMID: 28751251 DOI: 10.1016/j.semcdb.2017.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/05/2017] [Accepted: 07/17/2017] [Indexed: 02/08/2023]
Abstract
An ever-increasing number of studies highlight the role of cancer secretome in the modification of tumour microenvironment and in the acquisition of cancer cell resistance to therapeutic drugs. The knowledge of the mechanisms underlying the relationship between cancer cell-secreted factors and chemoresistance is becoming fundamental for the identification of novel anticancer therapeutic strategies overcoming drug resistance and novel prognostic secreted biomarkers. In this review, we summarize the novel findings concerning the regulation of secreted molecules by cancer cells compromising drug sensitivity. In particular, we highlight data from available literature describing the involvement of cancer cell-secreted molecules determining chemoresistance in an autocrine manner, including: i) growth factors; ii) glycoproteins; iii) inflammatory cytokines; iv) enzymes and chaperones; and v) tumor-derived exosomes.
Collapse
Affiliation(s)
- Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, Italy.
| |
Collapse
|
32
|
Qiu J, Peng B, Tang Y, Qian Y, Guo P, Li M, Luo J, Chen B, Tang H, Lu C, Cai M, Ke Z, He W, Zheng Y, Xie D, Li B, Yuan Y. CpG Methylation Signature Predicts Recurrence in Early-Stage Hepatocellular Carcinoma: Results From a Multicenter Study. J Clin Oncol 2017; 35:734-742. [PMID: 28068175 DOI: 10.1200/jco.2016.68.2153] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose Early-stage hepatocellular carcinoma (E-HCC) is being diagnosed increasingly, and in one half of diagnosed patients, recurrence will develop. Thus, it is urgent to identify recurrence-related markers. We investigated the effectiveness of CpG methylation in predicting recurrence for patients with E-HCCs. Patients and Methods In total, 576 patients with E-HCC from four independent centers were sorted by three phases. In the discovery phase, 66 tumor samples were analyzed using the Illumina Methylation 450k Beadchip. Two algorithms, Least Absolute Shrinkage and Selector Operation and Support Vector Machine-Recursive Feature Elimination, were used to select significant CpGs. In the training phase, penalized Cox regression was used to further narrow CpGs into 140 samples. In the validation phase, candidate CpGs were validated using an internal cohort (n = 141) and two external cohorts (n = 191 and n =104). Results After combining the 46 CpGs selected by the Least Absolute Shrinkage and Selector Operation and the Support Vector Machine-Recursive Feature Elimination algorithms, three CpGs corresponding to SCAN domain containing 3, Src homology 3-domain growth factor receptor-bound 2-like interacting protein 1, and peptidase inhibitor 3 were highlighted as candidate predictors in the training phase. On the basis of the three CpGs, a methylation signature for E-HCC (MSEH) was developed to classify patients into high- and low-risk recurrence groups in the training cohort ( P < .001). The performance of MSEH was validated in the internal cohort ( P < .001) and in the two external cohorts ( P < .001; P = .002). Furthermore, a nomogram comprising MSEH, tumor differentiation, cirrhosis, hepatitis B virus surface antigen, and antivirus therapy was generated to predict the 5-year recurrence-free survival in the training cohort, and it performed well in the three validation cohorts (concordance index: 0.725, 0.697, and 0.693, respectively). Conclusion MSEH, a three-CpG-based signature, is useful in predicting recurrence for patients with E-HCC.
Collapse
Affiliation(s)
- Jiliang Qiu
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Baogang Peng
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunqiang Tang
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yeben Qian
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Pi Guo
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengfeng Li
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junhang Luo
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bin Chen
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Tang
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Canliang Lu
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Muyan Cai
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zunfu Ke
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei He
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yun Zheng
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dan Xie
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Binkui Li
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunfei Yuan
- Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, Sun Yat-sen University Cancer Center; Baogang Peng, Junhang Luo, Bin Chen, and Zunfu Ke, First Affiliated Hospital of Sun Yat-sen University; Pi Guo and Mengfeng Li, Sun Yat-sen University; Jiliang Qiu, Muyan Cai, Wei He, Yun Zheng, Dan Xie, Binkui Li, and Yunfei Yuan, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine; Jiliang Qiu, Yunqiang Tang, and Hui Tang, Guangzhou Medical University Cancer Center, Guangzhou; and Yeben Qian and Canliang Lu, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
33
|
Oh S, Kim HS. Emerging power of proteomics for delineation of intrinsic tumor subtypes and resistance mechanisms to anti-cancer therapies. Expert Rev Proteomics 2016; 13:929-939. [PMID: 27599289 DOI: 10.1080/14789450.2016.1233063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Despite extreme genetic heterogeneity, tumors often show similar alterations in the expression, stability, and activation of proteins important in oncogenic signaling pathways. Thus, classifying tumor samples according to shared proteomic features may help facilitate the identification of cancer subtypes predictive of therapeutic responses and prognostic for patient outcomes. Meanwhile, understanding mechanisms of intrinsic and acquired resistance to anti-cancer therapies at the protein level may prove crucial to devising reversal strategies. Areas covered: Herein, we review recent advances in quantitative proteomic technology and their applications in studies to identify intrinsic tumor subtypes of various tumors, to illuminate mechanistic aspects of pharmacological and oncogenic adaptations, and to highlight interaction targets for anti-cancer compounds and cancer-addicted proteins. Expert commentary: Quantitative proteomic technologies are being successfully employed to classify tumor samples into distinct intrinsic subtypes, to improve existing DNA/RNA based classification methods, and to evaluate the activation status of key signaling pathways.
Collapse
Affiliation(s)
- Sejin Oh
- a Brain Korea 21 Project for Medical Science, Severance Biomedical Science Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Hyun Seok Kim
- a Brain Korea 21 Project for Medical Science, Severance Biomedical Science Institute , Yonsei University College of Medicine , Seoul , Korea
| |
Collapse
|
34
|
Elias KM, Emori MM, Westerling T, Long H, Budina-Kolomets A, Li F, MacDuffie E, Davis MR, Holman A, Lawney B, Freedman ML, Quackenbush J, Brown M, Drapkin R. Epigenetic remodeling regulates transcriptional changes between ovarian cancer and benign precursors. JCI Insight 2016; 1. [PMID: 27617304 DOI: 10.1172/jci.insight.87988] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Regulation of lineage-restricted transcription factors has been shown to influence malignant transformation in several types of cancer. Whether similar mechanisms are involved in ovarian cancer pathogenesis is unknown. PAX8 is a nuclear transcription factor that controls the embryologic development of the Müllerian system, including the fallopian tubes. Recent studies have shown that fallopian tube secretory epithelial cells (FTSECs) give rise to the most common form of ovarian cancer, high-grade serous ovarian carcinomas (HGSOCs). We designed the present study in order to understand whether changes in gene expression between FTSECs and HGSOCs relate to alterations in PAX8 binding to chromatin. Using whole transcriptome shotgun sequencing (RNA-Seq) after PAX8 knockdown and ChIP-Seq, we show that FTSECs and HGSOCs are distinguished by marked reprogramming of the PAX8 cistrome. Genes that are significantly altered between FTSECs and HGSOCs are enriched near PAX8 binding sites. These sites are also near TEAD binding sites, and these transcriptional changes may be related to PAX8 interactions with the TEAD/YAP1 signaling pathway. These data suggest that transcriptional changes after transformation in ovarian cancer are closely related to epigenetic remodeling in lineage-specific transcription factors.
Collapse
Affiliation(s)
- Kevin M Elias
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts, USA.; Harvard Medical School, Boston, Massachusetts, USA
| | - Megan M Emori
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.; Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Thomas Westerling
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Henry Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Anna Budina-Kolomets
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fugen Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Emily MacDuffie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michelle R Davis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts, USA.; Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander Holman
- Computational Biology and Functional Genomics Laboratory, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Brian Lawney
- Computational Biology and Functional Genomics Laboratory, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.; Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - John Quackenbush
- Computational Biology and Functional Genomics Laboratory, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Myles Brown
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ronny Drapkin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.; Harvard Medical School, Boston, Massachusetts, USA.; Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Ichinose J, Shinozaki-Ushiku A, Nagayama K, Nitadori JI, Anraku M, Fukayama M, Nakajima J, Takai D. Immunohistochemical pattern analysis of squamous cell carcinoma: Lung primary and metastatic tumors of head and neck. Lung Cancer 2016; 100:96-101. [PMID: 27597287 DOI: 10.1016/j.lungcan.2016.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/01/2016] [Accepted: 08/09/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVES This study aimed to develop an immunohistochemical (IHC) diagnostic algorithm for primary lung squamous cell carcinoma (LSCC) and pulmonary metastasis of head and neck SCC (HNSCC). MATERIALS AND METHODS We selected three antibodies (CK19, MMP3, and PI3) from a web-based gene expression database and an IHC analysis available online. We developed an IHC diagnostic algorithm using tissue microarrays from 39 LSCCs and 48 HNSCCs as the training set. It was validated using whole tumor sections of 32 LSCCs and 23 HNSCCs. The algorithm was applied to 28 cases with a history of HNSCC and who underwent resection of pulmonary squamous cell tumors. RESULTS The sensitivity, specificity, and accuracy of the algorithm were 90%, 62%, and 77%, respectively, in the training set and 96%, 44%, and 65%, respectively, in the validation set. Twenty-three of 28 SCCs were diagnosed as metastasis of HNSCC; the remaining five tumors were diagnosed as LSCC. Among the patients in the HNSCC group, 18 developed postoperative recurrence and 11 died of the disease, whereas only one patient in the LSCC group had recurrence. Compared with the LSCC group, the HNSCC group had poorer prognosis (P=0.07). IHC diagnosis coincided with the retrospective diagnosis in 22 (79%) of the 28 patients (sensitivity, 95%; specificity, 44%). CONCLUSION The IHC diagnostic algorithm may be clinically useful for distinguishing between LSCC and pulmonary metastasis of HNSCC.
Collapse
Affiliation(s)
- Junji Ichinose
- Department of Thoracic Surgery, University of Tokyo Hospital, Tokyo, Japan.
| | | | - Kazuhiro Nagayama
- Department of Thoracic Surgery, University of Tokyo Hospital, Tokyo, Japan
| | - Jun-Ichi Nitadori
- Department of Thoracic Surgery, University of Tokyo Hospital, Tokyo, Japan
| | - Masaki Anraku
- Department of Thoracic Surgery, University of Tokyo Hospital, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, University of Tokyo Hospital, Tokyo, Japan
| | - Jun Nakajima
- Department of Thoracic Surgery, University of Tokyo Hospital, Tokyo, Japan
| | - Daiya Takai
- Department of Clinical Laboratory, University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
36
|
Gu X, Li B, Jiang M, Fang M, Ji J, Wang A, Wang M, Jiang X, Gao C. RNA sequencing reveals differentially expressed genes as potential diagnostic and prognostic indicators of gallbladder carcinoma. Oncotarget 2016; 6:20661-71. [PMID: 25970782 PMCID: PMC4653033 DOI: 10.18632/oncotarget.3861] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/14/2015] [Indexed: 12/13/2022] Open
Abstract
Gallbladder carcinoma (GBC) is a rare tumor with a dismal survival rate overall. Hence, there is an urgent need for exploring more specific and sensitive biomarkers for the diagnosis and treatment of GBC. At first, amplified total RNAs from two paired GBC tumors and adjacent non-tumorous tissues (ANTTs) were subjected to RNA sequencing. 161 genes were identified differentially expressed between tumors and ANTTs. Functional enrichment analysis indicated that the up-regulated genes in tumor were primarily associated with signaling molecules and enzyme modulators, and mainly involved in cell cycles and pathways in cancer. Twelve differentially expressed genes (DEGs) were further confirmed in another independent cohort of 35 GBC patients. Expression levels of BIRC5, TK1, TNNT1 and MMP9 were found to be positively related to postoperative relapse. There was also a significant correlation between BIRC5 expression and tumor-node-metastasis (TNM) stage. Besides, we observed a positive correlation between serum CA19-9 concentration and the expression levels of TNNT1, MMP9 and CLIC3. Survival analysis revealed that GBC patients with high TK1 and MMP9 expression levels had worse prognosis. These identified DEGs might not only be promising biomarkers for GBC diagnosis and prognosis, but also expedite the discovery of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xing Gu
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Bin Li
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Mingming Jiang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Meng Fang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Jun Ji
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Aihua Wang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Mengmeng Wang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Xiaoqing Jiang
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| | - Chunfang Gao
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, PR China
| |
Collapse
|
37
|
Yuan S, Yu Z, Liu Q, Zhang M, Xiang Y, Wu N, Wu L, Hu Z, Xu B, Cai T, Ma X, Zhang Y, Liao C, Wang L, Yang P, Bai L, Li Y. GPC5, a novel epigenetically silenced tumor suppressor, inhibits tumor growth by suppressing Wnt/β-catenin signaling in lung adenocarcinoma. Oncogene 2016; 35:6120-6131. [DOI: 10.1038/onc.2016.149] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 03/19/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
|
38
|
Caruso JA, Karakas C, Zhang J, Yi M, Albarracin C, Sahin A, Bondy M, Liu J, Hunt KK, Keyomarsi K. Elafin is downregulated during breast and ovarian tumorigenesis but its residual expression predicts recurrence. Breast Cancer Res 2015; 16:3417. [PMID: 25551582 PMCID: PMC4326485 DOI: 10.1186/s13058-014-0497-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Elafin is an endogenous serine protease inhibitor. The majority of breast cancer cell lines lack elafin expression compared to human mammary epithelial cells. In this study, we hypothesized that elafin is downregulated during breast and ovarian tumorigenesis. METHODS We examined elafin expression by immunohistochemistry (IHC) in specimens of normal breast tissue (n = 24), ductal carcinoma in situ (DCIS) (n = 54), and invasive breast cancer (n = 793). IHC analysis of elafin expression was also performed in normal fallopian tube tissue (n = 20), ovarian cystadenomas (n = 9), borderline ovarian tumors (n = 21), and invasive ovarian carcinomas (n = 216). To understand the significance of elafin in luminal breast cancer cell lines, wild-type or M25G elafin (lacking the protease inhibitory function) were exogenously expressed in MCF-7 and T47D cells. RESULTS Elafin expression was downregulated in 24% of DCIS and 83% of invasive breast tumors when compared to elafin expression in the normal mammary epithelium. However, the presence of elafin-positive cells in invasive breast tumors, even at low frequency, correlated with poor recurrence-free survival (RFS), reduced overall survival (OS), and clinicopathological markers of aggressive tumor behavior. Elafin-positive cells were an especially strong and independent prognostic marker of reduced RFS in IHC-defined luminal A-like tumors. Elafin was also downregulated in 33% of ovarian cystadenomas, 43% of borderline ovarian tumors, and 86% of invasive ovarian carcinomas when compared to elafin expression in the normal fallopian tube. In ovarian tumors, elafin-positive cells were correlated with reduced RFS, OS and disease-specific survival (DSS) only in stage I/II patients and not in stage III/IV patients. Notably, exogenous expression of elafin or elafin M25G in the luminal breast cancer cell lines MCF-7 and T47D significantly decreased cell proliferation in a protease inhibitory domain-independent manner. CONCLUSIONS Elafin predicts poor outcome in breast and ovarian cancer patients and delineates a subset of endocrine receptor-positive breast cancer patients susceptible to recurrence who could benefit from more aggressive intervention. Our in vitro results suggest that elafin arrests luminal breast cancer cells, perhaps suggesting a role in tumor dormancy.
Collapse
|
39
|
|
40
|
Jiang X, Nguyen TT, Tian W, Sung YK, Yuan K, Qian J, Rajadas J, Sallenave JM, Nickel NP, de Jesus Perez V, Rabinovitch M, Nicolls MR. Cyclosporine Does Not Prevent Microvascular Loss in Transplantation but Can Synergize With a Neutrophil Elastase Inhibitor, Elafin, to Maintain Graft Perfusion During Acute Rejection. Am J Transplant 2015; 15:1768-81. [PMID: 25727073 PMCID: PMC4474772 DOI: 10.1111/ajt.13189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/10/2014] [Accepted: 12/26/2014] [Indexed: 01/25/2023]
Abstract
The loss of a functional microvascular bed in rejecting solid organ transplants is correlated with fibrotic remodeling and chronic rejection; in lung allografts, this pathology is predicted by bronchoalveolar fluid neutrophilia which suggests a role for polymorphonuclear cells in microcirculatory injury. In a mouse orthotopic tracheal transplant model, cyclosporine, which primarily inhibits T cells, failed as a monotherapy for preventing microvessel rejection and graft ischemia. To target neutrophil action that may be contributing to vascular injury, we examined the effect of a neutrophil elastase inhibitor, elafin, on the microvascular health of transplant tissue. We showed that elafin monotherapy prolonged microvascular perfusion and enhanced tissue oxygenation while diminishing the infiltration of neutrophils and macrophages and decreasing tissue deposition of complement C3 and the membrane attack complex, C5b-9. Elafin was also found to promote angiogenesis through activation of the extracellular signal-regulated kinase (ERK) signaling pathway but was insufficient as a single agent to completely prevent tissue ischemia during acute rejection episodes. However, when combined with cyclosporine, elafin effectively preserved airway microvascular perfusion and oxygenation. The therapeutic strategy of targeting neutrophil elastase activity alongside standard immunosuppression during acute rejection episodes may be an effective approach for preventing the development of irreversible fibrotic remodeling.
Collapse
Affiliation(s)
- Xinguo Jiang
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Tom T. Nguyen
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Wen Tian
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Yon K. Sung
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ke Yuan
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | - Jin Qian
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Jean-Michel Sallenave
- Unité de Défense Innée et Inflammation, Institut Pasteur, Paris, France,INSERM U884, Paris, France
| | - Nils P. Nickel
- Cardiovascular Institute and Department of Pediatrics, Stanford, CA
| | - Vinicio de Jesus Perez
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Mark R. Nicolls
- Veterans’ Affairs Palo Alto Health Care System, Medical Service, Palo Alto, CA,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
41
|
Abstract
Personalized medicine is the cornerstone of medical practice. It tailors treatments for specific conditions of an affected individual. The borders of personalized medicine are defined by limitations in technology and our understanding of biology, physiology and pathology of various conditions. Current advances in technology have provided physicians with the tools to investigate the molecular makeup of the disease. Translating these molecular make-ups to actionable targets has led to the development of small molecular inhibitors. Also, detailed understanding of genetic makeup has allowed us to develop prognostic markers, better known as companion diagnostics. Current attempts in the development of drug delivery systems offer the opportunity of delivering specific inhibitors to affected cells in an attempt to reduce the unwanted side effects of drugs.
Collapse
Affiliation(s)
- Gayane Badalian-Very
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline ave, Boston, MA 02115, United States. Tel.: + 1 617 513 7940; fax: + 1 617 632 5998.
| |
Collapse
|