1
|
Situ Y, Deng L, Huang Z, Jiang X, Zhao L, Zhang J, Lu L, Liang Q, Xu Q, Shao Z, Liang M. CDH2 and CDH13 as potential prognostic and therapeutic targets for adrenocortical carcinoma. Cancer Biol Ther 2024; 25:2428469. [PMID: 39545598 PMCID: PMC11572284 DOI: 10.1080/15384047.2024.2428469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/29/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Cadherin 2 (CDH2, N-cadherin) and cadherin 13 (CDH13, T-cadherin, H-cadherin) affect the progress and prognoses of many cancers. However, their roles in adrenocortical carcinoma (ACC), a rare endocrine cancer, remain unclear. To decipher the roles of these proteins in ACC and to identify their regulatory targets, we analyzed their expression levels, gene regulatory networks, prognostic value, and targets in ACC, using various bioinformatic analyses. CDH2 was strongly downregulated and CDH13 was strongly upregulated in patients with ACC; the expression levels of these genes affected the prognosis. In 75 patients, the expression of CDH2 and CDH13 was altered by 8% and 5%, respectively. CDH2 and CDH13, as well as their neighboring genes, were predicted to form a complex network of interactions, mainly through coexpression and physical and genetic interactions. CDH2 and its altered neighboring genes (ANGs) mainly affect tumor-related gene expression, cell cycle, and energy metabolism. The regulation of tumor-related integrin function, gene transcription, metabolism, and amide and phospholipid metabolism are the main functions of CDH13 and its ANGs. MiRNA and kinase targets of CDH2 and CDH13 in ACC were identified. CDH13 expression in patients with ACC was positively associated with immune cell infiltration. Anti-PD1/CTLA-4/PD-L1 immunotherapy significantly downregulated the expression of CDH13 in patients with ACC. Foretinib and elesclomol were predicted to exert strong inhibitory effects on SW13 cells by inhibiting the expression of CDH2 and CDH13. These data indicate that CDH2 and CDH13 are promising targets for precise treatment of ACC and may serve as new biomarkers for ACC prognosis.
Collapse
Affiliation(s)
- Yongli Situ
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| | - Li Deng
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| | - Ziqing Huang
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| | - Xiaoli Jiang
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Liubing Zhao
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Juying Zhang
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Lingling Lu
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| | - Quanyan Liang
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Qinying Xu
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| | - Zheng Shao
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| | - Meng Liang
- Department of Parasitology, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
2
|
Villarreal-García V, Estupiñan-Jiménez JR, Gonzalez-Villasana V, Vivas-Mejía PE, Flores-Colón M, Ancira-Moreno IE, Zapata-Morín PA, Altamirano-Torres C, Vázquez-Guillen JM, Rodríguez-Padilla C, Bayraktar R, Rashed MH, Ivan C, Lopez-Berestein G, Reséndez-Pérez D. Inhibition of microRNA-660-5p decreases breast cancer progression through direct targeting of TMEM41B. Hereditas 2024; 161:53. [PMID: 39709500 DOI: 10.1186/s41065-024-00357-5] [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: 09/17/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Breast cancer is the most prevalent cancer among women worldwide. Most breast cancer-related deaths result from metastasis and drug resistance. Novel therapies are imperative for targeting metastatic and drug-resistant breast cancer cells. Accumulating evidence suggests that dysregulated microRNAs (miRNAs) promote breast cancer progression, metastasis, and drug resistance. Compared with healthy breast tissue, miR-660-5p is notably overexpressed in breast cancer tumor tissues. However, the downstream effectors of miR-660-5p in breast cancer cells have not been fully elucidated. Our aim was to investigate the role of miR-660-5p in breast cancer cell proliferation, migration, invasion, and angiogenesis and to identify its potential targets. RESULTS Our findings revealed significant upregulation of miR-660-5p in MDA-MB-231 and MCF-7 cells compared with MCF-10 A cells. Furthermore, inhibiting miR-660-5p led to notable decreases in the proliferation, migration, and invasion of breast cancer cells, as well as angiogenesis, in HUVEC cells. Through bioinformatics analysis, we identified 15 potential targets of miR-660-5p. We validated TMEM41B as a direct target of miR-660-5p via Western blot and dual-luciferase reporter assays. CONCLUSIONS Our study highlights the upregulation and involvement of miR-660-5p in breast cancer cell proliferation, migration, invasion, and angiogenesis. Additionally, we identified TMEM41B as a direct target of miR-660-5p in breast cancer cells.
Collapse
Affiliation(s)
- Valeria Villarreal-García
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - José Roberto Estupiñan-Jiménez
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - Vianey Gonzalez-Villasana
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México.
| | - Pablo E Vivas-Mejía
- Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
- Comprehensive Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
| | - Marienid Flores-Colón
- Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
- Comprehensive Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Irma Estefanía Ancira-Moreno
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - Patricio Adrián Zapata-Morín
- Facultad de Ciencias Biológicas, Laboratorio de Micología y Fitopatología, Unidad de Manipulación Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - Claudia Altamirano-Torres
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - José Manuel Vázquez-Guillen
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - Cristina Rodríguez-Padilla
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| | - Recep Bayraktar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohamed H Rashed
- Clinical Pharmacy Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Diana Reséndez-Pérez
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Genética, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, México
| |
Collapse
|
3
|
Xiang P, Li P, Yuan X, Zhao X, Xiao Z, Chen B, Liu K, Bischof E, Han J. Exon 1 methylation status of CDH13 is associated with decreased overall survival and distant metastasis in patients with postoperative colorectal cancer. Discov Oncol 2024; 15:725. [PMID: 39611998 DOI: 10.1007/s12672-024-01604-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Cadherin 13 (CDH13) is a member of the cadherin superfamily that exerts tumor-suppressive effects on cancers derived from epithelial cells. Although hypermethylation of CDH13 promoter has been reported in various cancers, its prognostic value for colorectal cancer (CRC) is still controversial. The methylation alterations of CDH13 within exon 1 have not yet been investigated. METHODS A total of 49 CRC patients were recruited for the prospective study. The methylation status of CpG sites was quantified by Bisulfite Amplicon Sequencing (BSAS) in malignant tissues and adjacent normal tissues. The primary endpoint of the study was overall survival (OS) after surgery. The relationship between methylation level with pathological stage and OS was also evaluated. RESULTS Compared with adjacent normal tissues, the overall average methylation level within exon 1 was significantly increased in tumor tissues (p < 0.001). The association study showed that the hypermethylation status of the CpG1 site was non-significantly associated with the presence of distant metastasis (p = 0.032). Moreover, the hypermethylation of two CpG sites, including CpG1 (p = 0.003) and CpG5 (p = 0.032), was associated with worse OS in CRC. Co-hypermethylation of CpG1 and CpG5 sites was significantly associated with a worse clinical outcome (HR: 4.43 [95% CI 1.27-15.46]; p = 0.019) in multivariate Cox regression analysis. CONCLUSION The methylation level of CDH13 exon 1 in CRC tissue was significantly higher than in adjacent normal tissues. Hypermethylation at the CpG1 site suggests a risk of distant metastasis in CRC. The hypermethylation of the CpG1 site and CpG5 site, including the co-hypermethylation of these two sites, may serve as a valuable prognostic biomarker.
Collapse
Affiliation(s)
- PengCheng Xiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - PengJu Li
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Xiaoqi Yuan
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Xiuhao Zhao
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Zitian Xiao
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Bingguan Chen
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China
| | - Kenwen Liu
- Department of Gastrointestinal Surgery, Ji'an Central People's Hospital, Jian, 343000, Jiangxi, China
| | - Evelyne Bischof
- Department of Oncology and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
- Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Junyi Han
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine Shanghai, Shanghai, 200120, China.
| |
Collapse
|
4
|
Shin S, Kim HH, Kim JW, Rim D, An C, Chung YJ, Lee SH. Cellular responses to neoadjuvant FOLFOX6-bevacizumab treatment in colorectal cancers analyzed by single-cell transcriptome analysis. Pathol Res Pract 2024; 263:155681. [PMID: 39471526 DOI: 10.1016/j.prp.2024.155681] [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: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/01/2024]
Abstract
Neoadjuvant chemotherapy combined with bevacizumab is used to treat colorectal cancer (CRC) patients by targeting tumor and vascular cells. However, it is known that other cells in the tumor microenvironment (TME) also change in response to this treatment. To investigate the changes in TME subpopulations in response to neoadjuvant FOLFOX6 plus bevacizumab, we studied pre- and post-treatment CRC tissues in four patients using single-cell RNA sequencing (scRNA-seq). This analysis classified nine cell types, including epithelial, vascular, immune cells, and fibroblasts. The cellular responses were widespread across the cell types, but there were specific subpopulations that altered, especially in vascular, immune, and fibroblast cells. In vascular subpopulations, CDH13-endothelial, arteriole, and CA4 capillary cells were selectively reduced. In immune cells, CD4+, CD8+ T cells, conventional dendritic cell type 1 (cDC1), and CCL19-expressing migrating DC (migDC-1) increased, while Th17, Th22, and tumor-associated macrophage (TAM) cells decreased, indicating that the treatment might be immunostimulatory. In fibroblasts, two major cancer-associated fibroblasts (matrix CAF (mCAF) and inflammatory CAF (iCAF)) increased, while conventional fibroblasts decreased, suggesting that the treatment remodeled the reparative/inflammatory processes, which might lead to reduced aggressiveness from the cancer-associated fibroblasts. In summary, our study reveals that neoadjuvant FOLFOX6 plus bevacizumab leads to alterations in particular subpopulations of vascular, immune, and reparative/inflammatory cells in the TME of CRCs. These alterations include vascular reduction, immunologic stimulation, and reduction of cancer-associated fibroblasts, which may underlie the responsiveness to the therapy in CRC. Our results may provide insights into the mechanisms of responsiveness/resistance to neoadjuvant FOLFOX6 plus bevacizumab therapy in CRCs.
Collapse
Affiliation(s)
- Sun Shin
- Departments of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Hyun Ho Kim
- Departments of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Jae Woong Kim
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Doeun Rim
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Changhyeok An
- Departments of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| | - Yeun-Jun Chung
- Departments of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| | - Sug Hyung Lee
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
5
|
Singh DP, Pathak R, Chintalaramulu N, Pandit A, Kumar A, Ebenezer PJ, Kumar S, Duplooy A, White ME, Jambunathan N, Dharmakumar R, Francis J. Caveolin-1 knockout mitigates breast cancer metastasis to the lungs via integrin α3 dysregulation in 4T1-induced syngeneic breast cancer model. Cancer Gene Ther 2024; 31:1658-1668. [PMID: 39244591 PMCID: PMC11567888 DOI: 10.1038/s41417-024-00821-4] [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: 01/08/2024] [Revised: 07/22/2024] [Accepted: 08/02/2024] [Indexed: 09/09/2024]
Abstract
Caveolin-1 (Cav-1) is a critical lipid raft protein playing dual roles as both a tumor suppressor and promoter. While its role in tumorigenesis, progression, and metastasis has been recognized, the explicit contribution of Cav-1 to the onset of lung metastasis from primary breast malignancies remains unclear. Here, we present the first evidence that Cav-1 knockout in mammary epithelial cells significantly reduces lung metastasis in syngeneic breast cancer mouse models. In vitro, Cav-1 knockout in 4T1 cells suppressed extracellular vesicle secretion, cellular motility, and MMP secretion compared to controls. Complementing this, in vivo analyses demonstrated a marked reduction in lung metastatic foci in mice injected with Cav-1 knockout 4T1 cells as compared to wild-type cells, which was further corroborated by mRNA profiling of the primary tumor. We identified 21 epithelial cell migration genes exhibiting varied expression in tumors derived from Cav-1 knockout and wild-type 4T1 cells. Correlation analysis and immunoblotting further revealed that Cav-1 might regulate metastasis via integrin α3 (ITGα3). In silico protein docking predicted an interaction between Cav-1 and ITGα3, which was confirmed by co-immunoprecipitation. Furthermore, Cav-1 and ITGα3 knockdown corroborated its role in metastasis in the cell migration assay.
Collapse
Affiliation(s)
- Dhirendra Pratap Singh
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rashmi Pathak
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Naveen Chintalaramulu
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abhishek Pandit
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Avinash Kumar
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Philip J Ebenezer
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Sanjay Kumar
- Department of Biological Sciences Louisiana State University, Baton Rouge, LA, USA
| | - Alexander Duplooy
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Mary Evelyn White
- College of Veterinary Medicine, Midwestern University, Glendale, AZ, USA
| | - Nithya Jambunathan
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rohan Dharmakumar
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joseph Francis
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
6
|
Staebler S, Hoechst S, Thongmao A, Schneider N, Bosserhoff AK, Kuphal S. The Role of T-Cadherin (CDH13) in Treatment Options with Garcinol in Melanoma. Cancers (Basel) 2024; 16:1853. [PMID: 38791932 PMCID: PMC11119778 DOI: 10.3390/cancers16101853] [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: 04/18/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Targeted therapies with chemotherapeutic agents and immunotherapy with checkpoint inhibitors are among the systemic therapies recommended in the guidelines for clinicians to treat melanoma. Although there have been constant improvements in the treatment of melanoma, resistance to the established therapies continues to occur. Therefore, the purpose of this study was to explore the function of garcinol with regards to specific cancer properties such as proliferation and apoptosis. Garcinol, a natural compound isolated from the plant also known as mangosteen (Garcinia mangostana), is a newly discovered option for cancer treatment. Numerous pharmaceutical substances are derived from plants. For example, the derivates of camptothecin, extracted from the bark of the Chinese tree of happiness (Camptotheca acuminate), or paclitaxel, extracted from the bark of the Western yew tree (Taxus brevifolia), are used as anti-cancer drugs. Here, we show that garcinol reduced proliferation and induced apoptosis in melanoma cell lines. In addition, we found that those cells that are positive for the expression of the cell-cell adhesion molecule T-cadherin (CDH13) respond more sensitively to treatment with garcinol. After knock-down experiments with an siRNA pool against T-cadherin, the sensitivity to garcinol decreased and proliferation and anti-apoptotic behavior of the cells was restored. We conclude that patients who are T-cadherin-positive could especially benefit from a therapy with garcinol.
Collapse
Affiliation(s)
| | | | | | | | | | - Silke Kuphal
- Institute of Biochemistry, Friedrich Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany; (S.S.); (S.H.); (A.T.); (N.S.); (A.-K.B.)
| |
Collapse
|
7
|
Zareifar P, Ahmed HM, Ghaderi P, Farahmand Y, Rahnama N, Esbati R, Moradi A, Yazdani O, Sadeghipour Y. miR-142-3p/5p role in cancer: From epigenetic regulation to immunomodulation. Cell Biochem Funct 2024; 42:e3931. [PMID: 38379239 DOI: 10.1002/cbf.3931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 02/22/2024]
Abstract
MicroRNAs (miRNAs) play critical roles in cancer pathobiology, acting as regulators of gene expression and pivotal drivers of tumorigenesis. It is believed that miRNAs act through canonical mechanisms, involving the binding of mature miRNAs to target messenger RNAs (mRNAs) and subsequent repression of protein translation or degradation of target mRNAs. miR-142-3p/5p has been extensively studied and established as a key regulator in various malignancies. Recent discoveries have revealed miR-142-3p/5p serve as either oncogene or tumor suppressor in cancer. By targeting epigenetic factor and cancer-related signaling pathway, miR-142-3p/5p can regulate wide range of downstream genes. The immune modulatory role of miR-142-3p/5p has been shown in various cancers, which provides significant insight into immunosuppression and tumor escape from the immune response. Exosomes with miR-142-3p/5p facilitate cell communication and can affect cancer cell behavior, offering potential therapeutic, and diagnosis applications in cancer therapy. In this review, for the first time, we comprehensively summarize the current knowledge regarding mentioned functions of miR-142-3p/5p in cancer pathobiology.
Collapse
Affiliation(s)
- Parisa Zareifar
- Golestan University of Medical Science, Gorgan, Golestan, Iran
| | | | - Pouya Ghaderi
- Department of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Yalda Farahmand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Ali Moradi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Yasin Sadeghipour
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Ma G, Li Y, Meng F, Sui C, Wang Y, Cheng D. Hsa_circ_0000119 promoted ovarian cancer development via enhancing the methylation of CDH13 by sponging miR-142-5p. J Biochem Mol Toxicol 2023; 37:e23264. [PMID: 36482494 DOI: 10.1002/jbt.23264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the leading cause of gynecological cancer-related death in women, and is difficult to treat. The aim of our study is to explore the role and action mechanism of hsa_circ_0000119 in ovarian cancer, thus to analyze whether the circular RNA is a potential target for the treatment of the disease. In this present study, our data shows that hsa_circ_0000119 and DNA methyltransferase 1 (DNMT1) was increased, while miR-142-5p was decreased in ovarian cancer. Overexpression of hsa_circ_0000119 promoted tumor growth, while silencing of hsa_circ_0000119 resulted in an opposite effects. Decreasing of hsa_circ_0000119 also notably inhibited the proliferation, migration, and invasion of the ovarian cancer cells. Moreover, the data proves that hsa_circ_0000119 negatively regulated miR-142-5p and cadherin 13 (CDH13) expression, but positively regulated DNMT1 expression. miR-142-5p could interact with hsa_circ_0000119 and DNMT1 3'-UTR. Silencing of DNMT1 could reverse the inhibition of hsa_circ_0000119 to miR-142-5p and CDH13 expression. Importantly, higher level of CDH13 promoter methylation existed in the ovarian tumors than that in matched normal tissues. DNA methyltransferase inhibitor could increase the expression of CDH13 in ovarian cancer cells. In addition, our results also prove that increasing of CDH13 or miR-142-5p effectively reversed the inhibition of hsa _circ_0000119 to the cell malignant phenotypes. Overall, our data demonstrate that hsa_circ_0000119 facilitated ovarian cancer development through increasing CDH13 expression via promoting DNMT1 expression by sponging miR-142-5p. Our data demonstrate the potential role of hsa_circ_0000119 in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Guiyan Ma
- Outpatient Blood Collection Center, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan Li
- Department of Biotherapy, Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fandong Meng
- Department of Biotherapy, Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chengguang Sui
- Department of Biotherapy, Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Wang
- Department of Biotherapy, Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Dali Cheng
- Department of Gynaecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Heng Z, Zhao C, Gao Y. Comparison of urine proteomes from tumor-bearing mice with those from tumor-resected mice. PeerJ 2023; 11:e14737. [PMID: 36718454 PMCID: PMC9884041 DOI: 10.7717/peerj.14737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/22/2022] [Indexed: 01/26/2023] Open
Abstract
Objective This study aimed to address on the most important concern of surgeons-whether to completely resect tumor. Urine can indicate early changes associated with physiological or pathophysiological processes. Based on these ideas, we conducted experiments to explore changes in the urine proteome between tumor-bearing mice and tumor-resected mice. Method The tumor-bearing mouse model was established with MC38 mouse colon cancer cells, and the mice were divided into the control group, tumor-resected group, and tumor-bearing group. Urine was collected 7 and 30 days after tumor resection. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was used to identify the urine proteome, which was analyzed for differentially expressed proteins and functional annotation. Results (1) Seven days after tumor resection, 20 differentially expressed proteins distinguished the tumor-resected group and the tumor-bearing group. The identified biological processes included circadian rhythm, Notch signaling pathway, leukocyte cell-cell adhesion, and heterophilic cell-cell adhesion via plasma membrane cell adhesion molecules. (2) Thirty days after tumor resection, 33 differentially expressed proteins distinguished the tumor-resected group and the tumor-bearing group. The identified biological processes included cell adhesion; complement activation, the alternative pathway; the immune system process; and angiogenesis. (3) The difference in the urine proteome between the tumor-resected group and the healthy control group was smaller 30 days after tumor resection. Conclusion Changes in the urinary proteome can reflect the complete resection of MC38 tumors.
Collapse
Affiliation(s)
- Ziqi Heng
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, China
| | - Chenyang Zhao
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, China
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, China
| |
Collapse
|
10
|
Investigation of Transcriptome Patterns in Endometrial Cancers from Obese and Lean Women. Int J Mol Sci 2022; 23:ijms231911471. [PMID: 36232772 PMCID: PMC9569830 DOI: 10.3390/ijms231911471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 12/04/2022] Open
Abstract
Endometrial cancer is the most common gynaecological malignancy in developed countries. One of the largest risk factors for endometrial cancer is obesity. The aim of this study was to determine whether there are differences in the transcriptome of endometrial cancers from obese vs. lean women. Here we investigate the transcriptome of endometrial cancer between obese and lean postmenopausal women using rRNA-depleted RNA-Seq data from endometrial cancer tissues and matched adjacent non-cancerous endometrial tissues. Differential expression analysis identified 12,484 genes (6370 up-regulated and 6114 down-regulated) in endometrial cancer tissues from obese women, and 6219 genes (3196 up-regulated and 3023 down-regulated) in endometrial cancer tissues from lean women (adjusted p-value < 0.1). A gene ontology enrichment analysis revealed that the top 1000 up-regulated genes (by adjusted p-value) were enriched for growth and proliferation pathways while the top 1000 down-regulated genes were enriched for cytoskeleton restructure networks in both obese and lean endometrial cancer tissues. In this study, we also show perturbations in the expression of protein coding genes (HIST1H2BL, HIST1H3F, HIST1H2BH, HIST1H1B, TTK, PTCHD1, ASPN, PRELP, and CDH13) and the lncRNA MBNL1-AS1 in endometrial cancer tissues. Overall, this study has identified gene expression changes that are similar and also unique to endometrial cancers from obese vs. lean women. Furthermore, some of these genes may serve as prognostic biomarkers or, possibly, therapeutic targets for endometrial cancer.
Collapse
|
11
|
Herrera JA, Dingle L, Montero MA, Venkateswaran RV, Blaikley JF, Lawless C, Schwartz MA. The UIP/IPF fibroblastic focus is a collagen biosynthesis factory embedded in a distinct extracellular matrix. JCI Insight 2022; 7:e156115. [PMID: 35852874 PMCID: PMC9462507 DOI: 10.1172/jci.insight.156115] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Usual interstitial pneumonia (UIP) is a histological pattern characteristic of idiopathic pulmonary fibrosis (IPF). The UIP pattern is patchy with histologically normal lung adjacent to dense fibrotic tissue. At this interface, fibroblastic foci (FF) are present and are sites where myofibroblasts and extracellular matrix (ECM) accumulate. Utilizing laser capture microdissection-coupled mass spectrometry, we interrogated the FF, adjacent mature scar, and adjacent alveoli in 6 fibrotic (UIP/IPF) specimens plus 6 nonfibrotic alveolar specimens as controls. The data were subjected to qualitative and quantitative analysis and histologically validated. We found that the fibrotic alveoli protein signature is defined by immune deregulation as the strongest category. The fibrotic mature scar classified as end-stage fibrosis whereas the FF contained an overabundance of a distinctive ECM compared with the nonfibrotic control. Furthermore, FF were positive for both TGFB1 and TGFB3, whereas the aberrant basaloid cell lining of FF was predominantly positive for TGFB2. In conclusion, spatial proteomics demonstrated distinct protein compositions in the histologically defined regions of UIP/IPF tissue. These data revealed that FF are the main site of collagen biosynthesis and that the adjacent alveoli are abnormal. This essential information will inform future mechanistic studies on fibrosis progression.
Collapse
Affiliation(s)
| | - Lewis Dingle
- Blond McIndoe Laboratories, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - M. Angeles Montero
- Department of Histopathology, Manchester University National Health Service Foundation Trust, Manchester, United Kingdom
| | - Rajamiyer V. Venkateswaran
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Department of Transplant, Manchester University National Health Service Foundation Trust, Manchester, United Kingdom
| | - John F. Blaikley
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Department of Transplant, Manchester University National Health Service Foundation Trust, Manchester, United Kingdom
| | | | - Martin A. Schwartz
- The Wellcome Centre for Cell-Matrix Research and
- Yale Cardiovascular Research Center and
- Departments of Internal Medicine (Cardiology) and Cell Biology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Loaeza-Loaeza J, Illades-Aguiar B, Del Moral-Hernández O, Castro-Coronel Y, Leyva-Vázquez MA, Dircio-Maldonado R, Ortiz-Ortiz J, Hernández-Sotelo D. The CpG island methylator phenotype increases the risk of high-grade squamous intraepithelial lesions and cervical cancer. Clin Epigenetics 2022; 14:4. [PMID: 34991696 PMCID: PMC8740093 DOI: 10.1186/s13148-021-01224-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Background High-risk human papillomavirus (HR-HPV) infection is the main cause of cervical cancer, but additional alterations are necessary for its development. Abnormal DNA methylation has an important role in the origin and dissemination of cervical cancer and other human tumors. In this work, we analyzed the methylation of eight genes (AJAP1, CDH1, CDH13, MAGI2, MGMT, MYOD1, RASSF1A and SOX17) that participate in several biological processes for the maintenance of cell normality. We analyzed DNA methylation by methylation-specific PCR (MSP) and HPV infection using the INNO‑LiPA genotyping kit in 59 samples diagnostic of normal cervical tissue (non-SIL), 107 low-grade squamous intraepithelial lesions (LSILs), 29 high-grade squamous intraepithelial lesions (HSILs) and 51 cervical cancers (CCs). Results We found that all samples of LSIL, HSIL, and CC were HPV-positive, and the genotypes with higher frequencies were 16, 18, 51 and 56. In general, the genes analyzed displayed a significant tendency toward an increase in methylation levels according to increasing cervical lesion severity, except for the CDH13 gene. High CpG island methylator phenotype (CIMP) was associated with a 50.6-fold (95% CI 4.72–2267.3)-increased risk of HSIL and a 122-fold risk of CC (95% CI 10.04–5349.7). Conclusions We found that CIMP high was significantly associated with HSIL and CC risk. These results could indicate that CIMP together with HR-HPV infection and other factors participates in the development of HSIL and CC. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01224-0.
Collapse
Affiliation(s)
- Jaqueline Loaeza-Loaeza
- Laboratory of Cancer Epigenetics, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Laboratory of Molecular Biomedicine, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Oscar Del Moral-Hernández
- Laboratory of Cancer Virology, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Yaneth Castro-Coronel
- Laboratory of Cytopathology and Histochemistry, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Marco A Leyva-Vázquez
- Laboratory of Molecular Biomedicine, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Roberto Dircio-Maldonado
- Laboratory of Molecular Biomedicine, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Julio Ortiz-Ortiz
- Laboratory of Molecular Biomedicine, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico
| | - Daniel Hernández-Sotelo
- Laboratory of Cancer Epigenetics, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N Col. Haciendita, 39070, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
13
|
Epigenetics of Cutaneous Sarcoma. Int J Mol Sci 2021; 23:ijms23010422. [PMID: 35008848 PMCID: PMC8745302 DOI: 10.3390/ijms23010422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/25/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Epigenetic changes influence various physiological and pathological conditions in the human body. Recent advances in epigenetic studies of the skin have led to an appreciation of the importance of epigenetic modifications in skin diseases. Cutaneous sarcomas are intractable skin cancers, and there are no curative therapeutic options for the advanced forms of cutaneous sarcomas. In this review, we discuss the detailed molecular effects of epigenetic modifications on skin sarcomas, such as dermatofibrosarcoma protuberans, angiosarcoma, Kaposi's sarcoma, leiomyosarcoma, and liposarcoma. We also discuss the application of epigenetic-targeted therapy for skin sarcomas.
Collapse
|
14
|
Decidua Parietalis Mesenchymal Stem/Stromal Cells and Their Secretome Diminish the Oncogenic Properties of MDA231 Cells In Vitro. Cells 2021; 10:cells10123493. [PMID: 34944000 PMCID: PMC8700435 DOI: 10.3390/cells10123493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to suppress tumor growth, inhibit angiogenesis, regulate cellular signaling, and induce apoptosis in cancer cells. We have earlier reported that placenta-derived decidua parietalis mesenchymal stem/stromal cells (DPMSCs) not only retained their functional characteristics in the cancer microenvironment but also exhibited increased expression of anti-apoptotic genes, demonstrating their anti-tumor properties in the tumor setting. In this study, we have further evaluated the effects of DPMSCs on the functional outcome of human breast cancer cell line MDA231. MDA231 cells were exposed to DPMSCs, and their biological functions, including adhesion, proliferation, migration, and invasion, were evaluated. In addition, genomic and proteomic modifications of the MDA231 cell line, in response to the DPMSCs, were also evaluated. MDA231 cells exhibited a significant reduction in proliferation, migration, and invasion potential after their treatment with DPMSCs. Furthermore, DPMSC treatment diminished the angiogenic potential of MDA231 cells. DPMSC treatment modulated the expression of various pro-apoptotic as well as oncogenes in MDA231 cells. The properties of DPMSCs to inhibit the invasive characteristics of MDA231 cells demonstrate that they may be a useful candidate in a stem-cell-based therapy against cancer.
Collapse
|
15
|
Baranova I, Kovarikova H, Laco J, Sedlakova I, Vrbacky F, Kovarik D, Hejna P, Palicka V, Chmelarova M. Identification of a four-gene methylation biomarker panel in high-grade serous ovarian carcinoma. Clin Chem Lab Med 2021; 58:1332-1340. [PMID: 32145055 DOI: 10.1515/cclm-2019-1319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/04/2020] [Indexed: 12/31/2022]
Abstract
Background The lack of effective biomarkers for the screening and early detection of ovarian cancer (OC) is one of the most pressing problems in oncogynecology. Because epigenetic alterations occur early in the cancer development, they provide great potential to serve as such biomarkers. In our study, we investigated a potential of a four-gene methylation panel (including CDH13, HNF1B, PCDH17 and GATA4 genes) for the early detection of high-grade serous ovarian carcinoma (HGSOC). Methods For methylation detection we used methylation sensitive high-resolution melting analysis and real-time methylation specific analysis. We also investigated the relation between gene hypermethylation and gene relative expression using the 2-ΔΔCt method. Results The sensitivity of the examined panel reached 88.5%. We were able to detect methylation in 85.7% (12/14) of early stage tumors and in 89.4% (42/47) of late stage tumors. The total efficiency of the panel was 94.4% and negative predictive value reached 90.0%. The specificity and positive predictive value achieved 100% rates. Our results showed lower gene expression in the tumor samples in comparison to control samples. The more pronounced downregulation was measured in the group of samples with detected methylation. Conclusions In our study we designed the four-gene panel for HGSOC detection in ovarian tissue with 100% specificity and sensitivity of 88.5%. The next challenge is translation of the findings to the less invasive source for biomarker examination, such as plasma. Our results indicate that combination of examined genes deserve consideration for further testing in clinical molecular diagnosis of HGSOC.
Collapse
Affiliation(s)
- Ivana Baranova
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Helena Kovarikova
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Laco
- The Fingerland Department of Pathology, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Iva Sedlakova
- Department of Obstetrics and Gynecology, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Filip Vrbacky
- The 4th Department of Internal Medicine - Hematology, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Dalibor Kovarik
- Department of Forensic Medicine, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Petr Hejna
- Department of Forensic Medicine, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marcela Chmelarova
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
16
|
Chankeaw W, Lignier S, Richard C, Ntallaris T, Raliou M, Guo Y, Plassard D, Bevilacqua C, Sandra O, Andersson G, Humblot P, Charpigny G. Analysis of the transcriptome of bovine endometrial cells isolated by laser micro-dissection (1): specific signatures of stromal, glandular and luminal epithelial cells. BMC Genomics 2021; 22:451. [PMID: 34139994 PMCID: PMC8212485 DOI: 10.1186/s12864-021-07712-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 04/11/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND A number of studies have examined mRNA expression profiles of bovine endometrium at estrus and around the peri-implantation period of pregnancy. However, to date, these studies have been performed on the whole endometrium which is a complex tissue. Consequently, the knowledge of cell-specific gene expression, when analysis performed with whole endometrium, is still weak and obviously limits the relevance of the results of gene expression studies. Thus, the aim of this study was to characterize specific transcriptome of the three main cell-types of the bovine endometrium at day-15 of the estrus cycle. RESULTS In the RNA-Seq analysis, the number of expressed genes detected over 10 transcripts per million was 6622, 7814 and 8242 for LE, GE and ST respectively. ST expressed exclusively 1236 genes while only 551 transcripts were specific to the GE and 330 specific to LE. For ST, over-represented biological processes included many regulation processes and response to stimulus, cell communication and cell adhesion, extracellular matrix organization as well as developmental process. For GE, cilium organization, cilium movement, protein localization to cilium and microtubule-based process were the only four main biological processes enriched. For LE, over-represented biological processes were enzyme linked receptor protein signaling pathway, cell-substrate adhesion and circulatory system process. CONCLUSION The data show that each endometrial cell-type has a distinct molecular signature and provide a significantly improved overview on the biological process supported by specific cell-types. The most interesting result is that stromal cells express more genes than the two epithelial types and are associated with a greater number of pathways and ontology terms.
Collapse
Affiliation(s)
- Wiruntita Chankeaw
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, SLU, PO Box 7054, 750 07, Uppsala, Sweden
- Faculty of Veterinary Science, Rajamangala University of Technolgy Srivijaya (RUTS), Thungyai, Nakhon si thammarat, 80240, Thailand
| | - Sandra Lignier
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
| | - Christophe Richard
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
| | - Theodoros Ntallaris
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, SLU, PO Box 7054, 750 07, Uppsala, Sweden
| | - Mariam Raliou
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
| | - Yongzhi Guo
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, SLU, PO Box 7054, 750 07, Uppsala, Sweden
| | - Damien Plassard
- GenomEast Platform CERBM GIE, IGBMC, 67404, Illkirch, Cedex, France
| | - Claudia Bevilacqua
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy en Josas, France
| | - Olivier Sandra
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
| | - Göran Andersson
- Department of Animal Breeding and Genetics, Molecular Genetics, Swedish University of Agricultural Sciences, SLU, PO Box 7023, 750 07, Uppsala, Sweden
| | - Patrice Humblot
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, SLU, PO Box 7054, 750 07, Uppsala, Sweden
| | - Gilles Charpigny
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France.
- Department of Animal Breeding and Genetics, Molecular Genetics, Swedish University of Agricultural Sciences, SLU, PO Box 7023, 750 07, Uppsala, Sweden.
| |
Collapse
|
17
|
Di Palo A, Siniscalchi C, Polito R, Nigro E, Russo A, Daniele A, Potenza N. microRNA-377-3p downregulates the oncosuppressor T-cadherin in colorectal adenocarcinoma cells. Cell Biol Int 2021; 45:1797-1803. [PMID: 33818827 PMCID: PMC8360034 DOI: 10.1002/cbin.11605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is the second leading cause of death of malignant tumors worldwide. Recent studies point to a role for the adiponectin‐receptor axis in colorectal carcinogenesis, and in particular to the oncosuppressive properties of the T‐cadherin receptor. In addition, the loss of T‐cadherin expression in tumor tissues has been linked to cancer progression and attributed to aberrant methylation of its promoter. Recognizing the pivotal role of microRNAs in CRC, this study explores their possible contribution to the downregulation of T‐cadherin. A systematic bioinformatics analysis, restricted by microRNA expression data in the colon or in cultured colorectal cell lines, predicted twelve top‐ranking target miRNA sites within the 3ʹ UTR of T‐cadherin. Experimental validation analyses based on luciferase reporter constructs and miRNA mimic or miRNA inhibitor transfections toward colorectal adenocarcinoma cell lines indicated that miR‐377‐3p was able to directly bind to the T‐cadherin sequence, and thus downregulating its expression. Given the oncogenic activity of miR‐377 and the oncosuppressive activity of T‐cadherin in CRC, the regulatory circuit highlighted in this study may add new insights into molecular mechanisms driving colorectal carcinogenesis, and perspectively it could be exploited to identify novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Armando Di Palo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Chiara Siniscalchi
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Rita Polito
- CEINGE-Biotecnologie avanzate, Napoli, Italy
| | | | - Aniello Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Aurora Daniele
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy.,CEINGE-Biotecnologie avanzate, Napoli, Italy
| | - Nicoletta Potenza
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| |
Collapse
|
18
|
Lin HY, Huang PY, Cheng CH, Tung HY, Fang Z, Berglund AE, Chen A, French-Kwawu J, Harris D, Pow-Sang J, Yamoah K, Cleveland JL, Awasthi S, Rounbehler RJ, Gerke T, Dhillon J, Eeles R, Kote-Jarai Z, Muir K, Schleutker J, Pashayan N, Neal DE, Nielsen SF, Nordestgaard BG, Gronberg H, Wiklund F, Giles GG, Haiman CA, Travis RC, Stanford JL, Kibel AS, Cybulski C, Khaw KT, Maier C, Thibodeau SN, Teixeira MR, Cannon-Albright L, Brenner H, Kaneva R, Pandha H, Srinivasan S, Clements J, Batra J, Park JY. KLK3 SNP-SNP interactions for prediction of prostate cancer aggressiveness. Sci Rep 2021; 11:9264. [PMID: 33927218 PMCID: PMC8084951 DOI: 10.1038/s41598-021-85169-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
Risk classification for prostate cancer (PCa) aggressiveness and underlying mechanisms remain inadequate. Interactions between single nucleotide polymorphisms (SNPs) may provide a solution to fill these gaps. To identify SNP-SNP interactions in the four pathways (the angiogenesis-, mitochondria-, miRNA-, and androgen metabolism-related pathways) associated with PCa aggressiveness, we tested 8587 SNPs for 20,729 cases from the PCa consortium. We identified 3 KLK3 SNPs, and 1083 (P < 3.5 × 10-9) and 3145 (P < 1 × 10-5) SNP-SNP interaction pairs significantly associated with PCa aggressiveness. These SNP pairs associated with PCa aggressiveness were more significant than each of their constituent SNP individual effects. The majority (98.6%) of the 3145 pairs involved KLK3. The 3 most common gene-gene interactions were KLK3-COL4A1:COL4A2, KLK3-CDH13, and KLK3-TGFBR3. Predictions from the SNP interaction-based polygenic risk score based on 24 SNP pairs are promising. The prevalence of PCa aggressiveness was 49.8%, 21.9%, and 7.0% for the PCa cases from our cohort with the top 1%, middle 50%, and bottom 1% risk profiles. Potential biological functions of the identified KLK3 SNP-SNP interactions were supported by gene expression and protein-protein interaction results. Our findings suggest KLK3 SNP interactions may play an important role in PCa aggressiveness.
Collapse
Affiliation(s)
- Hui-Yi Lin
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| | - Po-Yu Huang
- Computational Intelligence Technology Center, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chia-Ho Cheng
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Heng-Yuan Tung
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Zhide Fang
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Ann Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jennifer French-Kwawu
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Darian Harris
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Julio Pow-Sang
- Department of Genitourinary Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Kosj Yamoah
- Department of Radiation Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Shivanshu Awasthi
- Department of Cancer Epidemiology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Robert J Rounbehler
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Travis Gerke
- Department of Cancer Epidemiology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jasreman Dhillon
- Department of Pathology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Rosalind Eeles
- The Institute of Cancer Research, London, SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | | | - Kenneth Muir
- Division of Population Health, Health Services Research, and Primary Care, University of Manchester, Oxford Road, Manchester, M139PT, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Johanna Schleutker
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
- Department of Medical Genetics, Genomics, Laboratory Division, Turku University Hospital, PO Box 52, 20521, Turku, Finland
| | - Nora Pashayan
- Department of Applied Health Research, University College London, London, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge, CB1 8RN, UK
- Department of Applied Health Research, University College London, London, WC1E 7HB, UK
| | - David E Neal
- Nuffield Department of Surgical Sciences, University of Oxford, Room 6603, Level 6, John Radcliffe Hospital, Headley Way, Headington, Oxford, OX3 9DU, UK
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Hills Road, Box 279, Cambridge, CB2 0QQ, UK
| | - Sune F Nielsen
- Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2200, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, 2200, Copenhagen, Denmark
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, 615 St Kilda Road, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Grattan Street, Parkville, VIC, 3010, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3168, Australia
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, 90015, USA
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, OX3 7LF, UK
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, 98195, USA
| | - Adam S Kibel
- Division of Urologic Surgery, Brigham and Womens Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, University of Cambridge, Cambridge, CB2 2QQ, UK
| | - Christiane Maier
- Humangenetik Tuebingen, Paul-Ehrlich-Str 23, 72076, Tuebingen, Germany
| | - Stephen N Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
- Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Lisa Cannon-Albright
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, 84148, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Radka Kaneva
- Department of Medical Chemistry and Biochemistry, Molecular Medicine Center, Medical University of Sofia, Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria
| | - Hardev Pandha
- University of Surrey, Guildford, GU2 7XH, Surrey, UK
| | - Srilakshmi Srinivasan
- Translational Research Institute, Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Judith Clements
- Translational Research Institute, Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Jyotsna Batra
- Translational Research Institute, Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| |
Collapse
|
19
|
Ebrahimpour A, Sarfi M, Rezatabar S, Tehrani SS. Novel insights into the interaction between long non-coding RNAs and microRNAs in glioma. Mol Cell Biochem 2021; 476:2317-2335. [PMID: 33582947 DOI: 10.1007/s11010-021-04080-x] [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: 10/07/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Glioma is the most common brain tumor of the central nervous system. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have been identified to play a vital role in the initiation and progression of glioma, including tumor cell proliferation, survival, apoptosis, invasion, and therapy resistance. New documents emerged, which indicated that the interaction between long non-coding RNAs and miRNAs contributes to the tumorigenesis and pathogenesis of glioma. LncRNAs can act as competing for endogenous RNA (ceRNA), and molecular sponge/deregulator in regulating miRNAs. These interactions stimulate different molecular signaling pathways in glioma, including the lncRNAs/miRNAs/Wnt/β-catenin molecular signaling pathway, the lncRNAs/miRNAs/PI3K/AKT/mTOR molecular signaling pathway, the lncRNAs-miRNAs/MAPK kinase molecular signaling pathway, and the lncRNAs/miRNAs/NF-κB molecular signaling pathway. In this paper, the basic roles and molecular interactions of the lncRNAs and miRNAs pathway glioma were summarized to better understand the pathogenesis and tumorigenesis of glioma.
Collapse
Affiliation(s)
- Anahita Ebrahimpour
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Sarfi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Rezatabar
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Shiu BH, Lu WY, Tantoh DM, Chou MC, Nfor ON, Huang CC, Liaw YP. Interactive association between dietary fat and sex on CDH13 cg02263260 methylation. BMC Med Genomics 2021; 14:13. [PMID: 33407434 PMCID: PMC7788866 DOI: 10.1186/s12920-020-00858-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND DNA methylation of Cadherin 13 (CDH13), a tumor suppressor gene is associated with gene repression and carcinogenesis. We determined the relation of dietary fat and sex with CDH13 cg02263260 methylation in Taiwanese adults. METHODS Data of 870 eligible participants (430 men and 440 women) between 30 and 70 years were obtained from the Taiwan Biobank (TWB) database. The association of dietary fat and sex with CDH13 cg02263260 methylation was determined using multiple linear regression. RESULTS The association between sex and cg02263260 methylation was significant: beta-coefficient (β) = 0.00532; 95% confidence interval (CI) = 0.00195-0.00868. Moreover, the interaction between sex and dietary fat on cg02263260 methylation was significant (P-value = 0.0145). After stratification by sex, the association of dietary fat with cg02263260 methylation was significant only in women. Specifically, high dietary fat was positively associated with cg02263260 methylation in women (β = 0.00597; 95% CI = 0.00061-0.01133) and the test for trend was significant (P-value = 0.0283). CONCLUSION High fat intake was significantly associated with higher cg02263260 methylation in women and the test for trend was significant. These findings suggest that the association of fat intake and CDH13 cg02263260 might vary by sex and CDH13 cg02263260 methylation levels in women might increase as fat intake increases.
Collapse
Affiliation(s)
- Bei-Hao Shiu
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Division of Colon-Rectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Wen-Yu Lu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan
| | - Disline Manli Tantoh
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan
| | - Chi-Chou Huang
- Division of Colon-Rectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan.
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110 Sec. 1 Jianguo N. Road, Taichung City, 40201, Taiwan.
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
| |
Collapse
|
21
|
Kisoda S, Shao W, Fujiwara N, Mouri Y, Tsunematsu T, Jin S, Arakaki R, Ishimaru N, Kudo Y. Prognostic value of partial EMT-related genes in head and neck squamous cell carcinoma by a bioinformatic analysis. Oral Dis 2020; 26:1149-1156. [PMID: 32277532 DOI: 10.1111/odi.13351] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/24/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Recent studies have revealed that the ability of cancer cells to undergo intermediate state of epithelial-to-mesenchymal transition (EMT), partial EMT (p-EMT), poses a higher metastatic risk rather than complete EMT. Here, we examined the prognostic value of p-EMT-related genes in head and neck squamous cell carcinoma (HNSCC) by bioinformatic approaches. MATERIALS AND METHODS We used RNA-seq data of 519 primary HNSCC cases obtained from TCGA database. We compared the expression of p-EMT-related genes in HNSCC tissues with normal tissues. We evaluated the prognostic value of p-EMT-related genes in HNSCC cases by log-rank test. We examined the expression of p-EMT-, EMT-, and epithelial differentiation-related genes by qPCR. RESULTS Among p-EMT-related genes that were highly expressed in HNSCC cases, high expression of SERPINE1, ITGA5, TGFBI, P4HA2, CDH13, and LAMC2 was significantly correlated with poor survival of HNSCC patients. By gene expression pattern, HNSCC cell lines were classified into three groups: epithelial phenotype, EMT phenotype, and p-EMT phenotype. CONCLUSIONS Our findings suggest that p-EMT program may be involved in poor prognosis of HNSCC. SERPINE1, ITGA5, TGFBI, P4HA2, CDH13, and LAMC2 can be used for a prognostic marker. Moreover, HNSCC cells with p-EMT phenotype can be a useful model for investigating a nature of p-EMT.
Collapse
Affiliation(s)
- Satoru Kisoda
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Wenhua Shao
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Natsumi Fujiwara
- Department of Oral Health Care Promotion, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasuhiro Mouri
- Department of Biochemistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shengjian Jin
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
22
|
Losi L, Zanocco-Marani T, Grande A. Cadherins down-regulation: towards a better understanding of their relevance in colorectal cancer. Histol Histopathol 2020; 35:1391-1402. [PMID: 32567668 DOI: 10.14670/hh-18-236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The down-regulation of cadherin expression in colorectal cancer (CRC) has been widely studied. However, existing data on cadherin expression are highly variable and its relevance to CRC development has not been completely established. This review examines published studies on cadherins whose down-regulation has been already demonstrated in CRC, trying to establish a relationship with promoter methylation, the capacity to influence the Wnt / CTNNB1 (catenin beta 1, beta-catenin) signalling pathway and the clinical implications for disease outcome. Moreover, it also analyses factors that may explain data variability and highlights the importance of considering the altered subcellular localization of the examined cadherins. The results of this survey reveal that thirty of one hundred existing cadherins appear to be down-regulated in CRC. Among these, ten are cadherins, sixteen are protocadherins, equally divided between clustered and non clustered, and four are cadherin - related. These findings suggest that, to better define the role played by cadherin down-regulation in CRC pathogenesis, the expression of multiple rather than individual cadherins should be taken into account and further functional studies are necessary to clarify the relative ability of individual cadherins to inhibit CTNNB1 therefore acting as tumor suppressors.
Collapse
Affiliation(s)
- Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | | | - Alexis Grande
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
23
|
Deng W, Liu H, Luo S, Clarke J, Glass C, Su L, Lin L, Christiani DC, Wei Q. APOB Genotypes and CDH13 Haplotypes in the Cholesterol-Related Pathway Genes Predict Non-Small Cell Lung Cancer Survival. Cancer Epidemiol Biomarkers Prev 2020; 29:1204-1213. [PMID: 32238407 PMCID: PMC7269811 DOI: 10.1158/1055-9965.epi-19-1262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/07/2020] [Accepted: 03/20/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Several oncogenic signals are involved in the synthesis, metabolism, transportation, and modulation of cholesterol. However, the roles of genetic variants of the cholesterol pathway genes in cancer survival remain unclear. METHODS We investigated associations between 26,781 common SNPs in 209 genes of the cholesterol pathway and non-small cell lung cancer (NSCLC) survival by utilizing genotyping data from two published genome-wide association studies. We used multivariate Cox proportional hazards regression and expression quantitative trait loci analyses to identify survival-associated SNPs and their correlations with the corresponding mRNA expression, respectively. We also used the Kaplan-Meier survival analysis and bioinformatics functional prediction to further evaluate the identified independent SNPs. RESULTS We found five independent SNPs (APOB rs1801701C>T; CDH13 rs35859010 C>T, rs1833970 T>A, rs254315 T>C, and rs425904 T>C) to be significantly associated with NSCLC survival in both discovery and replication datasets. When the unfavorable genotype (APOB rs1801701CC) and haplotypes (CDH13 rs35859010-rs1833970-rs254315-rs425904 C-A-T-C and T-T-T-T) were combined into a genetic score as the number of unfavorable genotypes/haplotypes (NUGH) in the multivariate analysis, an increased NUGH was associated with worse survival (P trend < 0.0001). In addition, both APOB rs1801701T CONCLUSIONS Genetic variants of APOB and CDH13 in the cholesterol pathway were associated with NSCLC survival, possibly by affecting their gene expression. IMPACT Genetic variants of APOB and CDH13 in the cholesterol pathway may provide new scientific insights into NSCLC prognosis.
Collapse
Affiliation(s)
- Wei Deng
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Jeffrey Clarke
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Carolyn Glass
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina
| | - Li Su
- Departments of Environmental Health and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Lijuan Lin
- Departments of Environmental Health and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - David C Christiani
- Departments of Environmental Health and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
24
|
Xu D, Yuan H, Meng Z, Yang C, Li Z, Li M, Zhang Z, Gan Y, Tu H. Cadherin 13 Inhibits Pancreatic Cancer Progression and Epithelial-mesenchymal Transition by Wnt/β-Catenin Signaling. J Cancer 2020; 11:2101-2112. [PMID: 32127937 PMCID: PMC7052920 DOI: 10.7150/jca.37762] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/21/2019] [Indexed: 12/16/2022] Open
Abstract
Cadherin 13 (CDH13) is an atypical cadherin that exerts tumor-suppressive effects on cancers derived from epithelial cells. Although the CDH13 promoter is frequently hypermethylated in pancreatic cancer (PC), the direct impact of CDH13 on PC is unknown. Accordingly, the expression of CDH13 in PC cell lines and paired PC tissues was examined by immunohistochemistry, quantitative real-time PCR and western blotting. Our findings showed that CDH13 was downregulated in PC tissues and cell lines. Moreover, cell proliferation, migration and invasion were detected by CCK-8 assay, transwell migration assay and transwell invasion assay, respectively. Xenograft tumor experiments were used to determine the biological function of CDH13 in vivo. As revealed by our data, CDH13 overexpression significantly inhibited the proliferation, migration and invasion of human PC cells in vitro. The inhibitory effect of CDH13 on PC was further confirmed in animal models. Mice subcutaneously or orthotopically transplanted with CDH13-overexpressing CFPAC-1 cells developed significantly smaller tumors with less liver metastases and mesenteric metastases than those of the control group. Next, transcriptomics and western blot analysis were used to identify the underlying mechanisms. Further molecular mechanism studies showed that CDH13 overexpression inhibited the activation of the Wnt/β-catenin signaling pathway and regulated the expression of epithelial-mesenchymal transition (EMT)-related markers. Our results indicated that CDH13 displayed an inhibitory effect on PC and suggested that CDH13 might be a potential biomarker and a new therapeutic target for PC.
Collapse
Affiliation(s)
- Dengfei Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Hui Yuan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.,Department of Thoracic Surgery, Cancer Research Center, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zihong Meng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Chunmei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Zefang Li
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mengge Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Yu Gan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Hong Tu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
25
|
Mahmoud NN, Abu-Dahab R, Hamadneh LA, Abuarqoub D, Jafar H, Khalil EA. Insights into the Cellular Uptake, Cytotoxicity, and Cellular Death Modality of Phospholipid-Coated Gold Nanorods toward Breast Cancer Cell Lines. Mol Pharm 2019; 16:4149-4164. [PMID: 31398052 DOI: 10.1021/acs.molpharmaceut.9b00470] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gold nanorods (GNRs) have gained pronounced recognition in the diagnosis and treatment of cancers driven by their distinctive properties. Herein, a gold-based nanosystem was prepared by utilizing a phospholipid moiety linked to thiolated polyethylene glycol, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-PEG-SH, as a surface decorating agent. The synthesized phospholipid-PEG-GNRs displayed good colloidal stability upon exposure to the tissue culture medium. Cytotoxicity of phospholipid-PEG-GNRs was investigated toward MCF-7 and T47D breast cancer cells using sulforhodamine B test. The results revealed that phospholipid-PEG-GNRs demonstrated high cytotoxicity to MCF-7 cells compared to T47D cells, and minimal cytotoxicity to human dermal fibroblasts. The cellular uptake studies performed by imaging and quantitative analysis demonstrated massive internalization of phospholipid-coated GNRs into MCF-7 cells in comparison to T47D cells. The cellular death modality of cancer cells after treatment with phospholipid-PEG-GNRs was evaluated using mitochondrial membrane potential assay (JC-1 dye), gene expression analysis, and flow cytometry study. The overall results suggest that phospholipid-modified GNRs enhanced mainly the cellular apoptotic events in MCF-7 cells in addition to necrosis, whereas cellular necrosis and suppression of cellular invasion contributed to the cellular death modality in the T47D cell line upon treatment with phospholipid-PEG-GNRs. The phospholipid-coated GNRs interact in a different manner with breast cancer cell lines and could be considered for breast cancer treatment.
Collapse
Affiliation(s)
- Nouf N Mahmoud
- Faculty of Pharmacy , Al-Zaytoonah University of Jordan , Amman 11733 , Jordan
| | | | - Lama A Hamadneh
- Faculty of Pharmacy , Al-Zaytoonah University of Jordan , Amman 11733 , Jordan
| | | | | | | |
Collapse
|
26
|
Yuan S, Li L, Xiang S, Jia H, Luo T. Cadherin-11 is inactivated due to promoter methylation and functions in colorectal cancer as a tumour suppressor. Cancer Manag Res 2019; 11:2517-2529. [PMID: 31114321 PMCID: PMC6497840 DOI: 10.2147/cmar.s193921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/28/2019] [Indexed: 12/24/2022] Open
Abstract
Background: The cadherin-11 (CDH11, OB-cadherin) gene is a member of the cadherin family and is located on chromosome 16q22.1. Previous studies have revealed that cadherins play significant roles in the development of many human malignancies. Increasing evidence has identified CDH11 as a functional tumour suppressor, which is commonly silenced by promoter methylation, but the functions of this gene in colorectal cancer (CRC) have been unclear. Methods: The CDH11 expression in primary CRC tissues and cell lines was investigated by qRT-PCR, RT-PCR and immunohistochemistry. The promoter methylation status of CDH11 was measured by methylation-specific PCR (MSP). Cell proliferation assay, colony formation assay, flow cytometry analysis, wound-healing assay, transwell assay and in vivo experiments were used to investigate the function of CDH11 in CRC. The mechanisms of CDH11 also were explored by western blots. Results: Our study suggests that CDH11 downregulation in CRC due to its promoter methylation and induced cell cycle arrest in G0/G1 phase and apoptosis, suppressing tumor cell proliferation, colony formation, migration and invasion by affecting the NF-kB signaling pathway. Conclusion: Overall, CDH11 may be considered as a functional tumour suppressor gene (TSG) in CRC, CDH11 has the potential to serve as a valuable prognostic marker for colorectal cancer.
Collapse
Affiliation(s)
- Shiyun Yuan
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lin Li
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Shili Xiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hexun Jia
- Office of academic, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Tao Luo
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|
27
|
Importance of Cadherins Methylation in Ovarian Cancer: a Next Generation Sequencing Approach. Pathol Oncol Res 2018; 25:1457-1465. [DOI: 10.1007/s12253-018-0500-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 10/15/2018] [Indexed: 12/21/2022]
|
28
|
Liu J, Liu Z, Liu Q, Li L, Fan X, Wen T, An G. CLEC3B is downregulated and inhibits proliferation in clear cell renal cell carcinoma. Oncol Rep 2018; 40:2023-2035. [PMID: 30066941 PMCID: PMC6111577 DOI: 10.3892/or.2018.6590] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/19/2018] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of C-Type Lectin Domain Family 3 Member B (CLEC3B) in serum or tumor tissues has been reported in patients with various cancer types. However, the expression and function of CLEC3B in clear cell renal cell carcinoma (ccRCC) remain unknown. To examine the function of CLEC3B in ccRCC, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were examined to determine the expression of CLEC3B at the transcriptional level and it was demonstrated that CLEC3B mRNA was significantly downregulated in ccRCC compared with normal tissues (P<0.0001 and P=0.0392 in TCGA and GEO databases, respectively). The downregulation of CLEC3B was further validated at the protein level in 78.9% of ccRCCs by immunohistochemistry. To investigate the potential genetic mechanism for CLEC3B downregulation in ccRCC, copy number analysis was performed by profiling the copy number variation data from the TCGA project and it was revealed that the copy number loss of CLEC3B was prevalent in up to 88.1% of patients with ccRCC. CLEC3B genetic deletion was coupled with the well-known genetic loss of the von Hippel-Lindau tumor suppressor, which is a characteristic oncogenic event during ccRCC carcinogenesis. The downregulation of CLEC3B was associated with tumor progression and predicted unfavorable prognostic outcomes in the TCGA cohort. Real-time cell analyzer system technology revealed that CLEC3B inhibited the proliferation of ccRCC cell lines in vitro and that the mitogen-activated protein kinase pathway may contribute to this process. CLEC3B demonstrated substantial positive associations with proliferation inhibitors, but inverse associations with proliferation inducers and markers in two large ccRCC cohorts, suggesting that CLEC3B was able to identify ccRCCs with a lower proliferation capacity. In conclusion, the results of the present study propose that CLEC3B is a promising target for therapeutic intervention in ccRCC.
Collapse
Affiliation(s)
- Jian Liu
- Medical Research Center, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Zhe Liu
- Department of Oncology, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Qun Liu
- Department of Obstetrics and Gynecology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Lina Li
- Medical Research Center, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiaona Fan
- Medical Research Center, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Tao Wen
- Medical Research Center, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Guangyu An
- Department of Oncology, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
29
|
Chattopadhyay S, Thomsen H, da Silva Filho MI, Weinhold N, Hoffmann P, Nöthen MM, Marina A, Jöckel KH, Schmidt B, Pechlivanis S, Langer C, Goldschmidt H, Hemminki K, Försti A. Enrichment of B cell receptor signaling and epidermal growth factor receptor pathways in monoclonal gammopathy of undetermined significance: a genome-wide genetic interaction study. Mol Med 2018; 24:30. [PMID: 30134812 PMCID: PMC6016882 DOI: 10.1186/s10020-018-0031-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/27/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recent identification of 10 germline variants predisposing to monoclonal gammopathy of undetermined significance (MGUS) explicates genetic dependency of this asymptomatic precursor condition with multiple myeloma (MM). Yet much of genetic burden as well as functional links remain unexplained. We propose a workflow to expand the search for susceptibility loci with genome-wide interaction and for subsequent identification of genetic clusters and pathways. METHODS Polygenic interaction analysis on 243 cases/1285 controls identified 14 paired risk loci belonging to unique chromosomal bands which were then replicated in two independent sets (case only study, 82 individuals; case/control study 236 cases/ 2484 controls). Further investigation on gene-set enrichment, regulatory pathway and genetic network was carried out with stand-alone in silico tools separately for both interaction and genome-wide association study-detected risk loci. RESULTS Intronic-PREX1 (20q13.13), a reported locus predisposing to MM was confirmed to have contribution to excess MGUS risk in interaction with SETBP1, a well-established candidate predisposing to myeloid malignancies. Pathway enrichment showed B cell receptor signaling pathway (P < 5.3 × 10- 3) downstream to allograft rejection pathway (P < 5.6 × 10- 4) and autoimmune thyroid disease pathway (P < 9.3 × 10- 4) as well as epidermal growth factor receptor regulation pathway (P < 2.4 × 10- 2) to be differentially regulated. Oncogene ALK and CDH2 were also identified to be moderately interacting with rs10251201 and rs16966921, two previously reported risk loci for MGUS. CONCLUSIONS We described novel pathways and variants potentially causal for MGUS. The methodology thus proposed to facilitate our search streamlines risk locus-based interaction, genetic network and pathway enrichment analyses.
Collapse
Affiliation(s)
- Subhayan Chattopadhyay
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany.
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany.
| | - Hauke Thomsen
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Miguel Inacio da Silva Filho
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Research Center, University of Bonn, Bonn, Germany
| | - Arendt Marina
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Börge Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sonali Pechlivanis
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Langer
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- National Centre of Tumor Diseases, Heidelberg, Germany
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| |
Collapse
|
30
|
Gene methylation as a powerful biomarker for detection and screening of non-small cell lung cancer in blood. Oncotarget 2018; 8:31692-31704. [PMID: 28404957 PMCID: PMC5458240 DOI: 10.18632/oncotarget.15919] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 01/10/2017] [Indexed: 11/25/2022] Open
Abstract
DNA methylation has been reported to become a potential powerful tool for cancer detection and diagnosis. However, the possibilities for the application of blood-based gene methylation as a biomarker for non-small cell lung cancer (NSCLC) detection and screening remain unclear. Hence, we performed this meta-analysis to evaluate the value of gene methylation detected in blood samples as a noninvasive biomarker in NSCLC. A total of 28 genes were analyzed from 37 case-control studies. In the genes with more than three studies, we found that the methylation of P16, RASSF1A, APC, RARβ, DAPK, CDH13, and MGMT was significantly associated with risks of NSCLC. The methylation statuses of P16, RASSF1A, APC, RARβ, DAPK, CDH13, and MGMT were not linked to age, gender, smoking behavior, and tumor stage and histology in NSCLC. Therefore, the use of the methylation status of P16, RASSF1A, APC, RARβ, DAPK, CDH13, and MGMT could become a promising and powerful biomarker for the detection and screening of NSCLC in blood in clinical settings. Further large-scale studies with large sample sizes are necessary to confirm our findings in the future.
Collapse
|
31
|
Parikh N, Shuck RL, Gagea M, Shen L, Donehower LA. Enhanced inflammation and attenuated tumor suppressor pathways are associated with oncogene-induced lung tumors in aged mice. Aging Cell 2018; 17. [PMID: 29047229 PMCID: PMC5771401 DOI: 10.1111/acel.12691] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2017] [Indexed: 01/09/2023] Open
Abstract
Aging is often accompanied by a dramatic increase in cancer susceptibility. To gain insights into how aging affects tumor susceptibility, we generated a conditional mouse model in which oncogenic KrasG12D was activated specifically in lungs of young (3–5 months) and old (19–24 months) mice. Activation of KrasG12D in old mice resulted in shorter survival and development of higher‐grade lung tumors. Six weeks after KrasG12D activation, old lung tissues contained higher numbers of adenomas than their young tissue counterparts. Lung tumors in old mice displayed higher proliferation rates, as well as attenuated DNA damage and p53 tumor suppressor responses. Gene expression comparison of lung tumors from young and old mice revealed upregulation of extracellular matrix‐related genes in young tumors, indicative of a robust cancer‐associated fibroblast response. In old tumors, numerous inflammation‐related genes such as Ccl7,IL‐1β, Cxcr6, and IL‐15ra were consistently upregulated. Increased numbers of immune cells were localized around the periphery of lung adenomas from old mice. Our experiments indicate that more aggressive lung tumor formation in older KrasG12D mice may be in part the result of subdued tumor suppressor and DNA damage responses, an enhanced inflammatory milieu, and a more accommodating tissue microenvironment.
Collapse
Affiliation(s)
- Neha Parikh
- Department of Molecular Virology and Microbiology; Baylor College of Medicine; Houston TX 77030 USA
| | - Ryan L. Shuck
- Department of Molecular Virology and Microbiology; Baylor College of Medicine; Houston TX 77030 USA
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery; UT MD Anderson Cancer Center; Houston TX 77030 USA
| | - Lanlan Shen
- Children's Nutrition Research Center; Houston TX 77030 USA
| | - Lawrence A. Donehower
- Department of Molecular Virology and Microbiology; Baylor College of Medicine; Houston TX 77030 USA
| |
Collapse
|
32
|
Zhu Q, Wang Z, Zhou L, Ren Y, Gong Y, Qin W, Bai L, Hu J, Wang T. The role of cadherin-11 in microcystin-LR-induced migration and invasion in colorectal carcinoma cells. Oncol Lett 2017; 15:1417-1422. [PMID: 29399188 PMCID: PMC5774544 DOI: 10.3892/ol.2017.7458] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/05/2017] [Indexed: 12/20/2022] Open
Abstract
The present study aimed to explore whether microcystin-LR (MC-LR; a well-known cyanobacterial toxin produced in eutrophic lakes or reservoirs) induced tumor progression by activating cadherin-11(CDH11). A previous tumor metastasis PCR array demonstrated that MC-LR exposure resulted in a significant increase in the expression of CDH11. In the present study, to confirm the effect of the MC-LR treatment on CDH11 expression, HT-29 cell migration and invasion following MC-LR treatment were tested by Transwell assays, and protein levels of CDH11 were tested by immunofluorescence and western blot analysis. The results demonstrated that MC-LR activated CDH11 expression in addition to cell migration and invasion in HT-29 cells. To further investigate the association between MC-LR-induced CDH11 upregulation, and higher motility and invasiveness in HT-29 cells, knockdown of CDH11 using small interfering RNA (siRNA) in HT-29 cells was performed. Subsequent Transwell assays confirmed that MC-LR-induced enhancement of migration and invasion was significantly decreased following CDH11 knockdown by CDH11-siRNA in HT-29 cells. The results from the present study indicate that MC-LR may act as a CDH11 activator to promote HT-29 cell migration and invasion.
Collapse
Affiliation(s)
- Qiangqiang Zhu
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Zhen Wang
- Department of Pathology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lihua Zhou
- Clinical Medicine School, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Yan Ren
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Ying Gong
- Department of Pharmacy, The Fourth People's Hospital of Jinan City, Jinan, Shandong 250000, P.R. China
| | - Wei Qin
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Lin Bai
- Clinical Medicine School, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Jun Hu
- Clinical Medicine School, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Ting Wang
- Department of Cell Biology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
33
|
Singh AN, Sharma N. Identification of key pathways and genes with aberrant methylation in prostate cancer using bioinformatics analysis. Onco Targets Ther 2017; 10:4925-4933. [PMID: 29066912 PMCID: PMC5644600 DOI: 10.2147/ott.s144725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Prostate cancer (PCa), a multifocal clinically heterogeneous disease, is the most commonly diagnosed non-cutaneous neoplasm in men worldwide. The epigenome of PCa is a typical representation of catastrophic model of epigenetic alterations during tumorigenesis and its progression. Alterations in methylation patterns in tumor suppressors, cell cycle, oncogenes and metabolism-related genes are the most commonly observed epigenetic alterations in PCa. In this study, we have developed a computational strategy to identify methylated biomarker signature panels as potential targets of PCa by screening >160 genes reported to be epigenetically dysregulated, and shortlisted 26 differentially methylated genes (DMGs) that significantly contribute to oncogenesis. The gene ontology and functional enrichment analysis were performed, which showed that identified DMGs contribute to cellular and metabolic processes such as cell communication, cell cycle, response to drugs, apoptosis and p53 signaling. The top hub genes AR, CDH13, CDKN2A, DAPK1, GSTP1, CD44 and RASSF1 identified from protein-protein interaction network construction using Search Tool for the Retrieval of Interacting Genes contributed to hormonal response, inflammatory response, cell cycle, reactive oxygen species activity and receptor kinase activity, which are related to hallmarks of cancer as revealed by their functional enrichment analysis by Cytoscape. These genes were further scrutinized for CpG islands, transcription start sites and positions of methylated cytosines to study their methylation profiles. Our analysis revealed high negative correlation values between methylation frequencies and gene expressions of the hub genes, namely, AR, CDH13, CDKN2A, DAPK1, CD44, GSTP1 and RASSF1, which can be used as potential diagnostic biomarkers for PCa.
Collapse
Affiliation(s)
- Anshika N Singh
- Symbiosis School of Biological Sciences, Symbiosis International University, Gram – Lavale, Taluka – Mulshi, Pune, India
| | - Neeti Sharma
- Symbiosis School of Biological Sciences, Symbiosis International University, Gram – Lavale, Taluka – Mulshi, Pune, India
| |
Collapse
|
34
|
Kong D, Wang MH, Yang J, Li L. Association of T-cadherin levels with the response to neoadjuvant chemotherapy in locally advanced breast cancer. Oncotarget 2017; 8:13747-13753. [PMID: 28099918 PMCID: PMC5355134 DOI: 10.18632/oncotarget.14630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/07/2017] [Indexed: 11/25/2022] Open
Abstract
Purpose To examine the association of T-cadherin with pathologic complete response (pCR) after neoadjuvant chemotherapy for locally advanced breast cancer. Results T-cadherin expression before and after neoadjuvant chemotherapy was similar (P = 0.162). The multivariable analysis indicated that negative T-cadherin expression was independently associated with pCR after neoadjuvant TAC chemotherapy (P = 0.001). Materials and Methods A total of 136 patients with locally advanced breast cancer received four cycles of neoadjuvant TAC chemotherapy (docetaxel + epirubicin + cyclophosphamide), followed by surgery. T-cadherin, estrogen receptor (ER), progesterone receptor (PR), HER-2, and Ki-67 were analyzed by immunohistochemistry. The association between T-cadherin expression and pCR after neoadjuvant chemotherapy was analyzed using multivariable logistic analysis. Conclusions Negative T-cadherin expression before and after neoadjuvant chemotherapy for locally advanced breast cancer was similar. T-cadherin could be considered an independent factor associated with the efficacy of such therapy.
Collapse
Affiliation(s)
- Dedi Kong
- Department of Thyroid and Breast Surgery, Jining No.1 People's Hospital, Jiningy 272011, Shandong, People's Republic of China
| | - Mei-Hong Wang
- Department of Thyroid and Breast Surgery, Jining No.1 People's Hospital, Jiningy 272011, Shandong, People's Republic of China
| | - Jie Yang
- Department of Pathology, Jining No.1 People's Hospital, Jiningy 272011, Shandong, People's Republic of China
| | - Liang Li
- Department of Pharmacy, Jining No.1 People's Hospital, Jiningy 272011, Shandong, People's Republic of China
| |
Collapse
|
35
|
Lin J, Chen Z, Huang Z, Chen F, Ye Z, Lin S, Wang W. Upregulation of T-cadherin suppresses cell proliferation, migration and invasion of gastric cancer in vitro. Exp Ther Med 2017; 14:4194-4200. [PMID: 29104635 PMCID: PMC5658734 DOI: 10.3892/etm.2017.5090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/31/2017] [Indexed: 12/12/2022] Open
Abstract
As a unique member of the cadherin superfamily, T-cadherin (T-cad) has been demonstrated to be associated with gastric cancer (GC) prognosis. To elucidate the function of T-cad in GC in vitro, the present study firstly examined T-cad protein expression in normal and gastric cancer tissues and cell lines, and it was demonstrated to be significantly downregulated in gastric cancer samples compared with normal samples. Control and T-cad expression vectors were then transfected into the MGC8-03 and AGS GC cell lines. Utilizing MTT, clonogenic, flow cytometry, wound healing and Transwell invasion assays in addition to Western blotting, the present study demonstrated that the overexpression of T-cad suppressed GC cell growth and colony formation via cell cycle arrest at the G0/G1 phase via downregulating the expression of cyclin dependent kinase 4 and Cyclin D1. In addition, overexpression of T-cad significantly inhibited GC cell migration and invasion by increasing E-cadherin and decreasing Vimentin expression. These findings suggest T-cad may be important in GC cell proliferation and metastasis and serve as a promising target for the treatment of GC in the future.
Collapse
Affiliation(s)
- Jianqing Lin
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Zhiyao Chen
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Zhijun Huang
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Feng Chen
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Zeyi Ye
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Shaoze Lin
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Weidong Wang
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| |
Collapse
|
36
|
Kong DD, Wang MH, Yang J, Li L, Wang W, Wang SB, Zhou YZ. T-cadherin is associated with prognosis in triple-negative breast cancer. Oncol Lett 2017; 14:2975-2981. [PMID: 28928835 PMCID: PMC5588130 DOI: 10.3892/ol.2017.6505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 05/26/2017] [Indexed: 12/25/2022] Open
Abstract
The purpose of the present study was to assess the prognostic impact of T-cadherin expression in patients with triple-negative breast cancer (TNBC). On the basis of the results of immunohistochemical analysis, 106 patients with operable TNBC were divided into two groups, the T-cadherin-positive group and T-cadherin-negative group. Fisher's exact and χ2 tests were employed to analyze clinical data, which included the association between T-cadherin expression and clinicopathological features and prognosis. The log-rank test was used to examine the impact of T-cadherin expression on the 5-year disease-free survival (DFS) and the 5-year overall survival (OS) of these patients. Kaplan-Meier and Cox regression analyses were introduced to analyze DFS and OS. Compared with the T-cadherin-positive group (58.3, 52.8 and 47.2, respectively; P=0.018, P=0.017, and P=0.047), tumor size >2 cm, grade II and III (Elston-Ellis modification of Bloom-Richardson grading system), and positive lymph node status were significantly more common in the T-cadherin-negative group compared with the T-cadherin-positive group (80.0 vs. 58.3%, 75.7 vs. 52.8% and 67.1 vs. 47.2%, respectively) (P=0.018, P=0.017, and P=0.047). Compared with the T-cadherin-positive group, 5-year DFS and OS levels were significantly lower in the T-cadherin-negative group (Z=6.233, P=0.013; Z=5.366, P=0.021). Multivariate analysis revealed that negative T-cadherin expression was an independent prognostic factor for DFS (P=0.009) and OS (P=0.048). The results of the present study indicated that negative T-cadherin expression indicated a worse prognosis for patients with TNBC.
Collapse
Affiliation(s)
- De-Di Kong
- Department of Thyroid and Breast Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Mei-Hong Wang
- Department of Thyroid and Breast Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Jie Yang
- Department of Pharmacy, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Liang Li
- Department of Pathology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Wei Wang
- Department of Thyroid and Breast Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Shi-Bing Wang
- Department of Thyroid and Breast Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Yan-Zhen Zhou
- Department of Thyroid and Breast Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
37
|
Estrogen receptor β, a regulator of androgen receptor signaling in the mouse ventral prostate. Proc Natl Acad Sci U S A 2017; 114:E3816-E3822. [PMID: 28439009 DOI: 10.1073/pnas.1702211114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
As estrogen receptor β-/- (ERβ-/-) mice age, the ventral prostate (VP) develops increased numbers of hyperplastic, fibroplastic lesions and inflammatory cells. To identify genes involved in these changes, we used RNA sequencing and immunohistochemistry to compare gene expression profiles in the VP of young (2-mo-old) and aging (18-mo-old) ERβ-/- mice and their WT littermates. We also treated young and old WT mice with an ERβ-selective agonist and evaluated protein expression. The most significant findings were that ERβ down-regulates androgen receptor (AR) signaling and up-regulates the tumor suppressor phosphatase and tensin homolog (PTEN). ERβ agonist increased expression of the AR corepressor dachshund family (DACH1/2), T-cadherin, stromal caveolin-1, and nuclear PTEN and decreased expression of RAR-related orphan receptor c, Bcl2, inducible nitric oxide synthase, and IL-6. In the ERβ-/- mouse VP, RNA sequencing revealed that the following genes were up-regulated more than fivefold: Bcl2, clusterin, the cytokines CXCL16 and -17, and a marker of basal/intermediate cells (prostate stem cell antigen) and cytokeratins 4, 5, and 17. The most down-regulated genes were the following: the antioxidant gene glutathione peroxidase 3; protease inhibitors WAP four-disulfide core domain 3 (WFDC3); the tumor-suppressive genes T-cadherin and caveolin-1; the regulator of transforming growth factor β signaling SMAD7; and the PTEN ubiquitin ligase NEDD4. The role of ERβ in opposing AR signaling, proliferation, and inflammation suggests that ERβ-selective agonists may be used to prevent progression of prostate cancer, prevent fibrosis and development of benign prostatic hyperplasia, and treat prostatitis.
Collapse
|
38
|
Klein RH, Lin Z, Hopkin AS, Gordon W, Tsoi LC, Liang Y, Gudjonsson JE, Andersen B. GRHL3 binding and enhancers rearrange as epidermal keratinocytes transition between functional states. PLoS Genet 2017; 13:e1006745. [PMID: 28445475 PMCID: PMC5425218 DOI: 10.1371/journal.pgen.1006745] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 05/10/2017] [Accepted: 04/05/2017] [Indexed: 11/19/2022] Open
Abstract
Transcription factor binding, chromatin modifications and large scale chromatin re-organization underlie progressive, irreversible cell lineage commitments and differentiation. We know little, however, about chromatin changes as cells enter transient, reversible states such as migration. Here we demonstrate that when human progenitor keratinocytes either differentiate or migrate they form complements of typical enhancers and super-enhancers that are unique for each state. Unique super-enhancers for each cellular state link to gene expression that confers functions associated with the respective cell state. These super-enhancers are also enriched for skin disease sequence variants. GRHL3, a transcription factor that promotes both differentiation and migration, binds preferentially to super-enhancers in differentiating keratinocytes, while during migration, it binds preferentially to promoters along with REST, repressing the expression of migration inhibitors. Key epidermal differentiation transcription factor genes, including GRHL3, are located within super-enhancers, and many of these transcription factors in turn bind to and regulate super-enhancers. Furthermore, GRHL3 represses the formation of a number of progenitor and non-keratinocyte super-enhancers in differentiating keratinocytes. Hence, chromatin relocates GRHL3 binding and enhancers to regulate both the irreversible commitment of progenitor keratinocytes to differentiation and their reversible transition to migration.
Collapse
Affiliation(s)
- Rachel Herndon Klein
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Ziguang Lin
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Amelia Soto Hopkin
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - William Gordon
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yun Liang
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California Irvine, Irvine, California, United States of America
- Department of Medicine, School of Medicine, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
39
|
Zhu C, Feng X, Ye G, Huang T. Meta-analysis of possible role of cadherin gene methylation in evolution and prognosis of hepatocellular carcinoma with a PRISMA guideline. Medicine (Baltimore) 2017; 96:e6650. [PMID: 28422868 PMCID: PMC5406084 DOI: 10.1097/md.0000000000006650] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cadherins (CDHs) have been reported to be associated with cancer. However, the clinical significance of CDH gene methylation in hepatocellular carcinoma (HCC) remains unclear. METHODS Based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement criteria, available studies were identified from online electronic database. The overall odds ratio (OR) and the corresponding 95% confidence interval (95% CI) were calculated and analyzed. RESULTS A total of 29 eligible studies with 2562 HCC samples and 1685 controls were included. E-cadherin (CDH1) hypermethylation was observed to be significantly higher in HCC than in benign, adjacent, or normal samples. Moreover, CDH1 hypermethylation was not associated with gender, tumor grade, clinical stage, hepatitis B virus (HBV), or hepatitis C virus (HCV) infection in HCC patients. H-cadherin (CDH13), protocadherin-10 (PCDH10), P-cadherin (CDH3), and M-cadherin (CDH15) methylation may have an increased risk of HCC in fewer than 4 studies, and methylated cadherin 8, type 2 (CDH8) and OB-cadherin (CDH11) had a similar OR in HCC and adjacent samples. When HCC samples were compared with normal samples, the analysis of sample type revealed a significantly higher OR in normal blood samples than in normal tissues for hypermethylated CDH1 (50.82 vs 4.44). CONCLUSION CDH1 hypermethylation may play a key role in the carcinogenesis of HCC. However, CDH1 hypermethylation was not correlated with clinicopathological features. Methylated CDH13, PCDH10, CDH3, and CDH15, but not methylated CDH8 or CDH11, may lead to an increased risk of HCC. Hypermethylated CDH1 may become a noninvasive blood biomarker. Further studies with more data are necessary.
Collapse
|
40
|
Ye M, Huang T, Li J, Zhou C, Yang P, Ni C, Chen S. Role of CDH13 promoter methylation in the carcinogenesis, progression, and prognosis of colorectal cancer: A systematic meta-analysis under PRISMA guidelines. Medicine (Baltimore) 2017; 96:e5956. [PMID: 28121942 PMCID: PMC5287966 DOI: 10.1097/md.0000000000005956] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND H-cadherin (CDH13) is commonly downregulated through promoter methylation in various cancers. However, the role of CDH13 promoter methylation status in patients with colorectal cancer (CRC) remains to be clarified. METHODS Eligible articles were identified from online electronic database based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement criteria. The pooled odds ratio (OR) and the corresponding 95% confidence interval (95% CI) were calculated and analyzed. RESULTS Eventually, a total of nine studies were included in this meta-analysis, including 488 CRC, 298 adjacent, 144 normal, 68 premalignant tissues. The results demonstrated that CDH13 promoter methylation was notably higher in CRC than in normal, adjacent, and premalignant tissues (cancer tissues vs normal tissues: OR = 16.94, P < 0.001; cancer tissues vs adjacent tissues: OR = 20.06, P < 0.001; cancer tissues vs premalignant tissues: OR = 2.23, P = 0.038). CDH13 promoter methylation had a significantly increased risk for poorly differentiated CRC (OR = 4.07, P = 0.001). CDH13 promoter methylation was not associated with sex status, tumor stage, and lymph node status (all P > 0.05). One study with 85 CRC patients reported that CDH13 promoter methylation was correlated with poor prognosis in overall survival (OS). CONCLUSIONS CDH13 promoter methylation may play an important role in the initiation and progression of CRC, and may be correlated with OS of patients with CRC. Additional studies with large sample sizes are needed to further confirm our findings in the future.
Collapse
Affiliation(s)
- Meng Ye
- The Affiliated Hospital of Ningbo University
| | - Tao Huang
- The Affiliated Hospital of Ningbo University
| | - Jinyun Li
- The Affiliated Hospital of Ningbo University
| | - Chongchang Zhou
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Ping Yang
- The Affiliated Hospital of Ningbo University
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Chao Ni
- The Affiliated Hospital of Ningbo University
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, People's Republic of China
| | - Si Chen
- The Affiliated Hospital of Ningbo University
| |
Collapse
|
41
|
Zhao JW, Fang F, Guo Y, Zhu TL, Yu YY, Kong FF, Han LF, Chen DS, Li F. HPV16 integration probably contributes to cervical oncogenesis through interrupting tumor suppressor genes and inducing chromosome instability. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:180. [PMID: 27884161 PMCID: PMC5123399 DOI: 10.1186/s13046-016-0454-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/09/2016] [Indexed: 12/11/2022]
Abstract
Background The integration of human papilloma virus (HPV) into host genome is one of the critical steps that lead to the progression of precancerous lesion into cancer. However, the mechanisms and consequences of such integration events are poorly understood. This study aims to explore those questions by studying high risk HPV16 integration in women with cervical intraepithelial neoplasia (CIN) and cervical squamous cell carcinoma (SCC). Methods Specifically, HPV integration status of 13 HPV16-infected patients were investigated by ligation-mediated PCR (DIPS-PCR) followed by DNA sequencing. Results In total, 8 HPV16 integration sites were identified inside or around genes associated with cancer development. In particular, the well-studied tumor suppressor genes SCAI was found to be integrated by HPV16, which would likely disrupt its expression and therefore facilitate the migration of tumor. On top of that, we observed several cases of chromosome translocation events coincide with HPV integration, which suggests the existence of chromosome instability. Additionally, short overlapping sequences were observed between viral derived and host derived fragments in viral-cellular junctions, indicating that integration was mediated by micro homology-mediated DNA repair pathway. Conclusions Overall, our study suggests a model in which HPV16 might contribute to oncogenesis not only by disrupting tumor suppressor genes, but also by inducing chromosome instability. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0454-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jun-Wei Zhao
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Fang Fang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Yi Guo
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Tai-Lin Zhu
- Abbey College Cambridge, Homerton Gardens, Cambridge, CB2 8EB, UK
| | - Yun-Yun Yu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Fan-Fei Kong
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Ling-Fei Han
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Dong-Sheng Chen
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK. .,Fitzwilliam College, University of Cambridge, Storey's Way, Cambridge, CB3 0DG, UK.
| | - Fang Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China.
| |
Collapse
|
42
|
Alkebsi L, Handa H, Yokohama A, Saitoh T, Tsukamoto N, Murakami H. Chromosome 16q genes CDH1, CDH13 and ADAMTS18 are correlated and frequently methylated in human lymphoma. Oncol Lett 2016; 12:3523-3530. [PMID: 27900031 DOI: 10.3892/ol.2016.5116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/19/2016] [Indexed: 12/31/2022] Open
Abstract
The products of the E-cadherin (CDH1), H-cadherin (CDH13) and a disintegrin and metalloproteinase with thrombospondin motif 18 (ADAMTS18) genes are proteins displaying structural features and functions on the cell surface membrane, and have been reported to be involved in cancer progression. Using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and methylation-specific PCR (MSP) analysis, the promoter methylation status and messenger RNA (mRNA) expression levels of CDH1, CDH13 and ADAMTS18, which are putative tumor-suppressor genes located on chromosome 16q, were evaluated. In addition, the mRNA expression levels of DNA methyltransferases (DNMTs) 1, 3A and 3B were examined, and the correlations among the different parameters analyzed were studied in 36 lymphomas and 16 non-malignant lymphoid tissue samples. A significant positive correlation was identified between the expression levels of CDH1 and CDH13 (r=0.735, P<0.01). ADAMTS18 expression also exhibited a significant positive correlation with both CDH1 and CDH13 mRNA expression levels (r=0.625, P<0.01; and r=0.720, P<0.01, respectively). Our results indicated that CDH1, CDH13 and ADAMTS18, which are localized on chromosome 16q, are remarkably correlated and frequently methylated in human lymphomas, and their methylation could not be explained solely by the mRNA expression level of DNMTs.
Collapse
Affiliation(s)
- Lobna Alkebsi
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Gunma 371-8511, Japan
| | - Hiroshi Handa
- Department of Medicine and Clinical Sciences, Graduate School of Medicine, Gunma University Hospital, Gunma 371-8511, Japan
| | - Akihiko Yokohama
- Blood Transfusion Service, Gunma University Hospital, Gunma 371-8511, Japan
| | - Takayuki Saitoh
- Oncology Center, Gunma University Hospital, Gunma 371-8511, Japan
| | | | - Hirokazu Murakami
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, Gunma 371-8511, Japan
| |
Collapse
|
43
|
Chen F, Huang T, Ren Y, Wei J, Lou Z, Wang X, Fan X, Chen Y, Weng G, Yao X. Clinical significance of CDH13 promoter methylation as a biomarker for bladder cancer: a meta-analysis. BMC Urol 2016; 16:52. [PMID: 27578166 PMCID: PMC5004266 DOI: 10.1186/s12894-016-0171-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/23/2016] [Indexed: 12/21/2022] Open
Abstract
Background Methylation of the tumor suppressor gene H-cadherin (CDH13) has been reported in many cancers. However, the clinical effect of the CDH13 methylation status of patients with bladder cancer remains to be clarified. Methods A systematic literature search was performed to identify eligible studies in the PubMed, Embase, EBSCO, CKNI and Wanfang databases. The pooled odds ratio (OR) and the corresponding 95 % confidence interval (95 % CI) was calculated and summarized. Results Nine eligible studies were included in the present meta-analysis consisting of a total of 1017 bladder cancer patients and 265 non-tumor controls. A significant association was found between CDH13 methylation levels and bladder cancer (OR = 21.71, P < 0.001). The results of subgroup analyses based on sample type suggested that CDH13 methylation was significantly associated with bladder cancer risk in both the tissue and the urine (OR = 53.94, P < 0.001; OR = 7.71, P < 0.001; respectively). A subgroup analysis based on ethnic population showed that the OR value of methylated CDH13 was higher in Asians than in Caucasians (OR = 35.18, P < 0.001; OR = 8.86, P < 0.001; respectively). The relationships between CDH13 methylation and clinicopathological features were also analyzed. A significant association was not observed between CDH13 methylation status and gender (P = 0.053). Our results revealed that CDH13 methylation was significantly associated with high-grade bladder cancer, multiple bladder cancer and muscle invasive bladder cancer (OR = 2.22, P < 0.001; OR = 1.45, P = 0.032; OR = 3.42, P < 0.001; respectively). Conclusion Our study indicates that CDH13 methylation may play an important role in the carcinogenesis, development and progression of bladder cancer. In addition, CDH13 methylation has the potential to be a useful biomarker for bladder cancer screening in urine samples and to be a prognostic biomarker in the clinic. Electronic supplementary material The online version of this article (doi:10.1186/s12894-016-0171-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Feng Chen
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Tao Huang
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Yu Ren
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Junjun Wei
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Zhongguan Lou
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Xue Wang
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Xiaoxiao Fan
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Yirun Chen
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Guobin Weng
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China
| | - Xuping Yao
- Laboratory of Kidney Carcinoma, Ningbo urology & Nephrology Hospital, Ningbo, 315040, Zhejiang, China.
| |
Collapse
|
44
|
Stem cell and neurogenic gene-expression profiles link prostate basal cells to aggressive prostate cancer. Nat Commun 2016; 7:10798. [PMID: 26924072 PMCID: PMC4773505 DOI: 10.1038/ncomms10798] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/21/2016] [Indexed: 12/26/2022] Open
Abstract
The prostate gland mainly contains basal and luminal cells constructed as a pseudostratified epithelium. Annotation of prostate epithelial transcriptomes provides a foundation for discoveries that can impact disease understanding and treatment. Here we describe a genome-wide transcriptome analysis of human benign prostatic basal and luminal epithelial populations using deep RNA sequencing. Through molecular and biological characterizations, we show that the differential gene-expression profiles account for their distinct functional properties. Strikingly, basal cells preferentially express gene categories associated with stem cells, neurogenesis and ribosomal RNA (rRNA) biogenesis. Consistent with this profile, basal cells functionally exhibit intrinsic stem-like and neurogenic properties with enhanced rRNA transcription activity. Of clinical relevance, the basal cell gene-expression profile is enriched in advanced, anaplastic, castration-resistant and metastatic prostate cancers. Therefore, we link the cell-type-specific gene signatures to aggressive subtypes of prostate cancer and identify gene signatures associated with adverse clinical features. Gene-expression profiles can be used to predict the prognosis of cancer patients. Here, the authors describe gene expression profiles of human prostate epithelial lineages and show that basal cells have intrinsic stem and neurogenic properties, and molecularly resemble aggressive prostate cancer.
Collapse
|
45
|
Matouk IJ, Halle D, Raveh E, Gilon M, Sorin V, Hochberg A. The role of the oncofetal H19 lncRNA in tumor metastasis: orchestrating the EMT-MET decision. Oncotarget 2016; 7:3748-65. [PMID: 26623562 PMCID: PMC4826167 DOI: 10.18632/oncotarget.6387] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/15/2015] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNA (lncRNA) genes are emerging as key players in the metastatic cascade. Current evidence indicate that H19 lncRNA and the microRNA(miRNA) miR-675, which is processed from it, play crucial roles in metastasis, through the regulation of critical events specifically the epithelial to mesenchymal (EMT) and the mesenchymal to epithelial transitions (MET). This review summarizes recent mechanistic pathways and tries to put together seemingly conflicting data from different reports under one proposed general scheme underlying the various roles of H19/miR-675 in the metastatic cascade. We propose several approaches to harnessing this knowledge for translational medicine.
Collapse
Affiliation(s)
- Imad J. Matouk
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biological Sciences, Faculty of Science and Technology, Al-Quds University, Jerusalem, West Bank
| | - David Halle
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eli Raveh
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Gilon
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vladimir Sorin
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avraham Hochberg
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
46
|
Eiring AM, Khorashad JS, Anderson DJ, Yu F, Redwine HM, Mason CC, Reynolds KR, Clair PM, Gantz KC, Zhang TY, Pomicter AD, Kraft IL, Bowler AD, Johnson K, Mac Partlin M, O’Hare T, Deininger MW. β-Catenin is required for intrinsic but not extrinsic BCR-ABL1 kinase-independent resistance to tyrosine kinase inhibitors in chronic myeloid leukemia. Leukemia 2015; 29:2328-37. [PMID: 26202934 PMCID: PMC4675686 DOI: 10.1038/leu.2015.196] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/06/2015] [Accepted: 07/13/2015] [Indexed: 12/26/2022]
Abstract
Activation of nuclear β-catenin and expression of its transcriptional targets promotes chronic myeloid leukemia (CML) progression, tyrosine kinase inhibitor (TKI) resistance, and leukemic stem cell self-renewal. We report that nuclear β-catenin has a role in leukemia cell-intrinsic but not -extrinsic BCR-ABL1 kinase-independent TKI resistance. Upon imatinib inhibition of BCR-ABL1 kinase activity, β-catenin expression was maintained in intrinsically resistant cells grown in suspension culture and sensitive cells cultured in direct contact (DC) with bone marrow (BM) stromal cells. Thus, TKI resistance uncouples β-catenin expression from BCR-ABL1 kinase activity. In β-catenin reporter assays, intrinsically resistant cells showed increased transcriptional activity versus parental TKI-sensitive controls, and this was associated with restored expression of β-catenin target genes. In contrast, DC with BM stromal cells promoted TKI resistance, but had little effects on Lef/Tcf reporter activity and no consistent effects on cytoplasmic β-catenin levels, arguing against a role for β-catenin in extrinsic TKI resistance. N-cadherin or H-cadherin blocking antibodies abrogated DC-based resistance despite increasing Lef/Tcf reporter activity, suggesting that factors other than β-catenin contribute to extrinsic, BM-derived TKI resistance. Our data indicate that, while nuclear β-catenin enhances survival of intrinsically TKI-resistant CML progenitors, it is not required for extrinsic resistance mediated by the BM microenvironment.
Collapse
Affiliation(s)
- Anna M. Eiring
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | | | - David J. Anderson
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Fan Yu
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
- Beijing Tsinghua Chang Gung Hospital, Tsinghua University, Beijing, China
| | - Hannah M. Redwine
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Clinton C. Mason
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | | | - Phillip M. Clair
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Kevin C. Gantz
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Tian Y. Zhang
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Anthony D. Pomicter
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Ira L. Kraft
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Amber D. Bowler
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
| | - Kara Johnson
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, OR, U.S.A
| | - Mary Mac Partlin
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, OR, U.S.A
| | - Thomas O’Hare
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
- Department of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT, U.S.A
| | - Michael W. Deininger
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, U.S.A
- Department of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT, U.S.A
| |
Collapse
|
47
|
Thomas SN, Harlan R, Chen J, Aiyetan P, Liu Y, Sokoll LJ, Aebersold R, Chan DW, Zhang H. Multiplexed Targeted Mass Spectrometry-Based Assays for the Quantification of N-Linked Glycosite-Containing Peptides in Serum. Anal Chem 2015; 87:10830-8. [PMID: 26451657 DOI: 10.1021/acs.analchem.5b02063] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protein glycosylation is one of the most common protein modifications, and the quantitative analysis of glycoproteins has the potential to reveal biological functions and their association with disease. However, the high throughput accurate quantification of glycoproteins is technically challenging due to the scarcity of robust assays to detect and quantify glycoproteins. Here we describe the development of multiplexed targeted MS assays to quantify N-linked glycosite-containing peptides in serum using parallel reaction monitoring (PRM). Each assay was characterized by its performance metrics and criteria established by the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium (NCI CPTAC) to facilitate the widespread adoption of the assays in studies designed to confidently detect changes in the relative abundance of these analytes. An in-house developed software program, MRMPlus, was used to compute assay performance parameters including specificity, precision, and repeatability. We show that 43 selected N-linked glycosite-containing peptides identified in prostate cancer tissue studies carried out in our group were detected in the sera of prostate cancer patients within the quantitative range of the developed PRM assays. A total of 41 of these formerly N-linked glycosite-containing peptides (corresponding to 37 proteins) were reproducibly quantified based on their relative peak area ratios in human serum during PRM assay development, with 4 proteins showing differential significance in serum from nonaggressive (NAG) vs aggressive (AG) prostate cancer patient serum (n = 50, NAG vs AG). The data demonstrate that the assays can be used for the high throughput and reproducible quantification of a panel of formerly N-linked glycosite-containing peptides. The developed assays can also be used for the quantification of formerly N-linked glycosite-containing peptides in human serum irrespective of disease state.
Collapse
Affiliation(s)
- Stefani N Thomas
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| | - Robert Harlan
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| | - Jing Chen
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| | - Paul Aiyetan
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| | - Yansheng Liu
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland
| | - Lori J Sokoll
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland.,Faculty of Science, University of Zurich , 8057 Zurich, Switzerland
| | - Daniel W Chan
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| | - Hui Zhang
- Department of Pathology, Clinical Chemistry Division, Johns Hopkins University School of Medicine , 1550 Orleans Street, CRBII Room 3M03, Baltimore, Maryland 21231, United States
| |
Collapse
|
48
|
T-Cadherin Expression in Melanoma Cells Stimulates Stromal Cell Recruitment and Invasion by Regulating the Expression of Chemokines, Integrins and Adhesion Molecules. Cancers (Basel) 2015. [PMID: 26197340 PMCID: PMC4586773 DOI: 10.3390/cancers7030840] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
T-cadherin is a glycosyl-phosphatidylinositol (GPI) anchored member of the cadherin superfamily involved in the guidance of migrating cells. We have previously shown that in vivo T-cadherin overexpression leads to increased melanoma primary tumor growth due to the recruitment of mesenchymal stromal cells as well as the enhanced metastasis. Since tumor progression is highly dependent upon cell migration and invasion, the aim of the present study was to elucidate the mechanisms of T-cadherin participation in these processes. Herein we show that T-cadherin expression results in the increased invasive potential due to the upregulated expression of pro-oncogenic integrins, chemokines, adhesion molecules and extracellular matrix components. The detected increase in chemokine expression could be responsible for the stromal cell recruitment. At the same time our previous data demonstrated that T-cadherin expression inhibited neoangiogenesis in the primary tumors. We demonstrate that T-cadherin overexpression leads to the increase in the expression of anti-angiogenic molecules and reduction in pro-angiogenic factors. Thus, T-cadherin plays a dual role in melanoma growth and progression: T-cadherin expression results in anti-angiogenic effects in melanoma, however, this also stimulates transcription of genes responsible for migration and invasion of melanoma cells.
Collapse
|
49
|
CDH13 promoter SNPs with pleiotropic effect on cardiometabolic parameters represent methylation QTLs. Hum Genet 2014; 134:291-303. [PMID: 25543204 PMCID: PMC4318987 DOI: 10.1007/s00439-014-1521-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Abstract
CDH13 encodes T-cadherin, a receptor for high molecular weight (HMW) adiponectin and low-density lipoprotein, promoting proliferation and migration of endothelial cells. Genome-wide association studies have mapped multiple variants in CDH13 associated with cardiometabolic traits (CMT) with variable effects across studies. We hypothesized that this heterogeneity might reflect interplay with DNA methylation within the region. Resequencing and EpiTYPER™ assay were applied for the HYPertension in ESTonia/Coronary Artery Disease in Czech (HYPEST/CADCZ; n = 358) samples to identify CDH13 promoter SNPs acting as methylation Quantitative Trait Loci (meQTLs) and to investigate their associations with CMT. In silico data were extracted from genome-wide DNA methylation and genotype datasets of the population-based sample Estonian Genome Center of the University of Tartu (EGCUT; n = 165). HYPEST–CADCZ meta-analysis identified a rare variant rs113460564 as highly significant meQTL for a 134-bp distant CpG site (P = 5.90 × 10−6; β = 3.19 %). Four common SNPs (rs12443878, rs12444338, rs62040565, rs8060301) exhibited effect on methylation level of up to 3 neighboring CpG sites in both datasets. The strongest association was detected in EGCUT between rs8060301 and cg09415485 (false discovery rate corrected P value = 1.89 × 10−30). Simultaneously, rs8060301 showed association with diastolic blood pressure, serum high-density lipoprotein and HMW adiponectin (P < 0.005). Novel strong associations were identified between rare CDH13 promoter meQTLs (minor allele frequency <5 %) and HMW adiponectin: rs2239857 (P = 5.50 × 10−5, β = −1,841.9 ng/mL) and rs77068073 (P = 2.67 × 10−4, β = −2,484.4 ng/mL). Our study shows conclusively that CDH13 promoter harbors meQTLs associated with CMTs. It paves the way to deeper understanding of the interplay between DNA variation and methylation in susceptibility to common diseases.
Collapse
|
50
|
Wang Z, Wang B, Guo H, Shi G, Hong X. Clinicopathological significance and potential drug target of T-cadherin in NSCLC. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 9:207-16. [PMID: 25565774 PMCID: PMC4278732 DOI: 10.2147/dddt.s74259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background Previous studies demonstrate that T-cadherin is a candidate tumor suppressor in several types of human tumors, including non-small cell lung cancer (NSCLC). Lack of protein expression of T-cadherin by hypermethylation has been found to play an important role in lung alveolar differentiation regulation and epithelial tumorigenesis. However, the correlation between T-cadherin hypermethylation and clinicopathological characteristics of NSCLC remains unclear. Here we conducted a systematic review and meta-analysis to quantitatively evaluate the effects of T-cadherin hypermethylation on the incidence of NSCLC and clinicopathological characteristics. Methods A detailed literature search was carried out for related research publications. Analyses of pooled data were performed. Odds ratio (OR) and hazard ratio (HR) were calculated and summarized, respectively. Results Final analysis of 1,172 NSCLC patients from 15 eligible studies was performed. T-cadherin hypermethylation was observed to be significantly higher in NSCLC than in normal lung tissue, based on the pooled OR from nine studies including 532 NSCLC and 372 normal lung tissue samples (OR=8.19, 95% confidence interval [CI]=5.41–12.39, P<0.00001). T-cadherin hypermethylation may also be associated with pathological types. The pooled OR was obtained from four studies including 111patients with squamous cell carcinoma and 106 with adenocarcinoma (OR=0.35, 95% CI=0.19–0.66, P=0.001), which indicated that T-cadherin hypermethylation plays a more important role in the pathogenesis of adenocarcinoma. We did not find that T-cadherin hypermethylation was correlated with the sex or smoking status, clinical stages, or epidermal growth factor receptor (EGFR) mutation status. However, T-cadherin hypermethylation was found to be significantly higher in poorly differentiated NSCLC than in moderately and highly differentiated NSCLC, and NSCLC patients with T-cadherin hypermethylation had a lower survival rate than those without T-cadherin hypermethylation. Conclusion The results of this meta-analysis suggest that T-cadherin hypermethylation is associated with an increased risk and worse survival in NSCLC. T-cadherin hypermethylation, which induces the inactivation of T-cadherin gene, plays an important role in the carcinogenesis, cancer progression, as well as clinical outcome.
Collapse
Affiliation(s)
- Zhidong Wang
- Oncology Department, Eighth Hospital of Changsha, Changsha, People's Republic of China
| | - Bin Wang
- Oncology Department, Eighth Hospital of Changsha, Changsha, People's Republic of China
| | - Huanchen Guo
- Department of Respiratory Medicine, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, People's Republic of China
| | - Guoyu Shi
- Department of Respiratory Medicine, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, People's Republic of China
| | - Xiuqin Hong
- Institute of Gerontology, Hunan Geriatric Hospital, Changsha, People's Republic of China
| |
Collapse
|