1
|
Aslan C, Maralbashi S, Shekari N, Javadian M, Shomali N, Kazemi T. Differential effects of docosahexaenoic acid (DHA) and linoleic acid (LA) on miR-101 and miR-342 tumor suppressor microRNAs in Taxol-treated HER2-positive breast cancer cells. Clin Nutr ESPEN 2024; 63:502-507. [PMID: 39047867 DOI: 10.1016/j.clnesp.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 05/14/2024] [Accepted: 07/06/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND & AIMS Docosahexaenoic acid (DHA) and linoleic acid (LA) have been shown to exhibit anti-proliferative effects against breast cancer cells. However, the mechanisms underlying these effects are not yet fully understood. One potential mechanism is through the regulation of microRNAs (miRs), which are known to play a crucial role in breast cancer development and progression. This study aimed to investigate the expression of miR-342 and miR-101 as tumor-suppressor miRs in the human HER-2 positive breast cancer cell line BT-474 after treatment with DHA, LA, alone or in combination with Taxol, a standard chemotherapy agent. METHODS The human breast cancer cell line BT-474 was cultured, and the IC50 for Taxol was determined using the MTT assay. Cells were then cultured and treated for 24 h with 100 μM DHA and 50 μM LA, alone or in combination with the respective IC50 of Taxol. Cells were harvested, and miRNA extraction and cDNA synthesis were performed using standard methods. Expression levels of miRs were analyzed using quantitative real-time PCR (qRT-PCR), and results were normalized against U6 snRNA expression levels. RESULTS The Taxol IC50 for BT-474 cells was 19 nM. According to the data obtained from our study, it was observed that Taxol treatment resulted in the down-regulation of both miR-101 and miR-342 (3.69 (p < 0.0001) and 1.88 fold, (p < 0.0001) respectively). In addition, DHA, LA and DHA + LA caused up-regulation of miR-101 (0.11, 0.05, 0.03 fold (p < 0.0001) respectively) but not miR-342 (decreased by 1.93 (p < 0.0001), 2.89 (p < 0.0001) and 1.19 fold (p = 0.0029) respectively). Notably, treatment with DHA, LA and DHA + LA was able to restore the down-regulated expression of miR-101 (0.25 (p < 0.0001), 0.05 (p = 0.0012) and 0.06 fold (p < 0.0001) respectively) during Taxol treatment. CONCLUSION Our study demonstrates that DHA and LA can effectively compensate for the reduced expression of miR-101 during Taxol treatment. These findings suggest that dietary fatty acids may play a critical role in modulating the anti-cancer effects of chemotherapy agents. Future studies are needed to investigate the functional aspects of dietary fatty acids on breast cancer development and progression.
Collapse
Affiliation(s)
- Cynthia Aslan
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Maralbashi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Najibeh Shekari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Javadian
- Department of Immunology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Mohebbi H, Esbati R, Hamid RA, Akhavanfar R, Radi UK, Siri G, Yazdani O. EZH2-interacting lncRNAs contribute to gastric tumorigenesis; a review on the mechanisms of action. Mol Biol Rep 2024; 51:334. [PMID: 38393645 DOI: 10.1007/s11033-024-09237-7] [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: 11/13/2023] [Accepted: 01/10/2024] [Indexed: 02/25/2024]
Abstract
Gastric cancer (GC) remains one of the deadliest malignancies worldwide, demanding new targets to improve its diagnosis and treatment. Long non-coding RNAs (lncRNAs) are dysregulated through gastric tumorigenesis and play a significant role in GC progression and development. Recent studies have revealed that lncRNAs can interact with histone-modifying polycomb protein, enhance Zeste Homolog 2 (EZH2), and mediate its site-specific functioning. EZH2, which functions as an oncogene in GC, is the catalytic subunit of the PRC2 complex that induces H3K27 trimethylation and epigenetically represses gene expression. EZH2-interacting lncRNAs can recruit EZH2 to the promoter regions of various tumor suppressor genes and cause their transcriptional deactivation via histone methylation. The interactions between EZH2 and this lncRNA modulate different processes, such as cell cycle, cell proliferation and growth, migration, invasion, metastasis, and drug resistance, in vitro and in vivo GC models. Therefore, EZH2-interacting lncRNAs are exciting targets for developing novel targeted therapies for GC. Subsequently, this review aims to focus on the roles of these interactions in GC progression to understand the therapeutic value of EZH2-interacting lncRNAs further.
Collapse
Affiliation(s)
- Hossein Mohebbi
- Kermanshah University of medical sciences, International branch, Kermanshah, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | | | - Roozbeh Akhavanfar
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Goli Siri
- Department of Internal Medicine, Amir Alam Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
3
|
Hashemi M, Nazdari N, Gholamiyan G, Paskeh MDA, Jafari AM, Nemati F, Khodaei E, Abyari G, Behdadfar N, Raei B, Raesi R, Nabavi N, Hu P, Rashidi M, Taheriazam A, Entezari M. EZH2 as a potential therapeutic target for gastrointestinal cancers. Pathol Res Pract 2024; 253:154988. [PMID: 38118215 DOI: 10.1016/j.prp.2023.154988] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023]
Abstract
Gastrointestinal (GI) cancers continue to be a major cause of mortality and morbidity globally. Understanding the molecular pathways associated with cancer progression and severity is essential for creating effective cancer treatments. In cancer research, there is a notable emphasis on Enhancer of zeste homolog 2 (EZH2), a key player in gene expression influenced by its irregular expression and capacity to attach to promoters and alter methylation status. This review explores the impact of EZH2 signaling on various GI cancers, such as colorectal, gastric, pancreatic, hepatocellular, esophageal, and cholangiocarcinoma. The primary function of EZH2 signaling is to facilitate the accelerated progression of cancer cells. Additionally, EZH2 has the capacity to modulate the reaction of GI cancers to chemotherapy and radiotherapy. Numerous pathways, including long non-coding RNAs and microRNAs, serve as upstream regulators of EZH2 in these types of cancer. EZH2's enzymatic activity enables it to attach to target gene promoters, resulting in methylation that modifies their expression. EZH2 could be considered as an independent prognostic factor, with increased expression correlating with a worse disease prognosis. Additionally, a range of gene therapies including small interfering RNA, and anti-tumor agents are being explored to target EZH2 for cancer treatment. This comprehensive review underscores the current insights into EZH2 signaling in gastrointestinal cancers and examines the prospect of therapies targeting EZH2 to enhance patient outcomes.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Naghmeh Nazdari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ghazaleh Gholamiyan
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fateme Nemati
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Khodaei
- Department of Dermatology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazal Abyari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Behdadfar
- Young Researchers and Elite Club, Buinzahra Branch, Islamic Azad University, Buinzahra, Iran
| | - Behnaz Raei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Matulić M, Gršković P, Petrović A, Begić V, Harabajsa S, Korać P. miRNA in Molecular Diagnostics. Bioengineering (Basel) 2022; 9:bioengineering9090459. [PMID: 36135005 PMCID: PMC9495386 DOI: 10.3390/bioengineering9090459] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
MicroRNAs are a class of small non-coding RNA molecules that regulate gene expression on post-transcriptional level. Their biogenesis consists of a complex series of sequential processes, and they regulate expression of many genes involved in all cellular processes. Their function is essential for maintaining the homeostasis of a single cell; therefore, their aberrant expression contributes to development and progression of many diseases, especially malignant tumors and viral infections. Moreover, they can be associated with certain states of a specific disease, obtained in the least invasive manner for patients and analyzed with basic molecular methods used in clinical laboratories. Because of this, they have a promising potential to become very useful biomarkers and potential tools in personalized medicine approaches. In this review, miRNAs biogenesis, significance in cancer and infectious diseases, and current available test and methods for their detection are summarized.
Collapse
Affiliation(s)
- Maja Matulić
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Paula Gršković
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Andreja Petrović
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Institute of Clinical Pathology and Cytology, Merkur University Hospital, 10000 Zagreb, Croatia
| | - Valerija Begić
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Primary School “Sesvetski Kraljevec”, 10361 Sesvetski Kraljevec, Croatia
| | - Suzana Harabajsa
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Department of Pathology and Cytology, Division of Pulmonary Cytology Jordanovac, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Petra Korać
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: ; Tel.: +385-1-4606-278
| |
Collapse
|
5
|
Flores AR, Rêma A, Mesquita JR, Taulescu M, Seixas F, Gärtner F, Amorim I. E-cadherin Expression in Canine Gastric Carcinomas: Association with Clinicopathological Parameters. Vet Sci 2022; 9:172. [PMID: 35448670 PMCID: PMC9027758 DOI: 10.3390/vetsci9040172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
E-cadherin (E-cad) is a cell-adhesion molecule known for its tumor-invasion suppressor function. E-cad expression was examined immunohistochemically in a series of canine tissue samples, including normal gastric mucosa (NGM; n = 3), gastric carcinomas (GC; n = 33), adjacent non-neoplastic mucosa (NNM; n = 32), neoplastic emboli (n = 16) and metastatic lesions (n = 9). The relationship between E-cad expression and clinicopathological features were investigated. In NGM, epithelial cells showed strong latero-lateral membranous expression of E-cad, and this pattern was considered normal. The membranous staining was preserved in all specimens of NNM (100%), whereas abnormal E-cad expression was found in 87.9% of the GCs. A marked difference in E-cad expression was observed between normal and malignant tissues (p < 0.0002). Abnormal E-cad expression was significantly more frequent in poorly/undifferentiated carcinomas (96%) and diffuse (95%) and indeterminate carcinomas (100%) than in well-differentiated/intestinal ones (62.5%; p = 0.0115 and p = 0.0392, respectively). There was significant association between abnormal E-cad expression and the depth of invasion (p = 0.0117), and the presence neoplastic emboli (p = 0.0194). No statistically significant differences in E-cad expression were observed concerning tumor location, histological type according to WHO classification, and presence of metastatic lesions. Therefore, deregulation of E-cad expression may play a role in canine gastric carcinogenesis and in tumor progression; moreover, it might be a prognostic tool for canine gastric cancer.
Collapse
Affiliation(s)
- Ana R. Flores
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (A.R.F.); (A.R.); (J.R.M.); (I.A.)
- Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), 4200-465 Porto, Portugal;
- CECAV—Veterinary and Animal Research Center, University pf Trás-os-Montes and Alto Douro (UTAD), 5001-801 Vila Real, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5001-801 Vila Real, Portugal
| | - Alexandra Rêma
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (A.R.F.); (A.R.); (J.R.M.); (I.A.)
| | - João R. Mesquita
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (A.R.F.); (A.R.); (J.R.M.); (I.A.)
- Epidemiology Research Unit (EPIUnit), Instituto de Saúde Pública, Universidade do Porto (ISPUP), 4050-600 Porto, Portugal
| | - Marian Taulescu
- Department of Pathology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
- Synevovet Laboratory, 81 Pache Protopopescu, 021408 Bucharest, Romania
| | - Fernanda Seixas
- CECAV—Veterinary and Animal Research Center, University pf Trás-os-Montes and Alto Douro (UTAD), 5001-801 Vila Real, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5001-801 Vila Real, Portugal
| | - Fátima Gärtner
- Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), 4200-465 Porto, Portugal;
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Irina Amorim
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (A.R.F.); (A.R.); (J.R.M.); (I.A.)
- Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), 4200-465 Porto, Portugal;
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
6
|
Yue Y, Lin X, Qiu X, Yang L, Wang R. The Molecular Roles and Clinical Implications of Non-Coding RNAs in Gastric Cancer. Front Cell Dev Biol 2021; 9:802745. [PMID: 34966746 PMCID: PMC8711095 DOI: 10.3389/fcell.2021.802745] [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: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 01/19/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies in the world. It is also the fifth most common cancer in China. In recent years, a large number of studies have proved that non-coding RNAs (ncRNAs) can regulate cell proliferation, invasion, metastasis, apoptosis, and angiogenesis. NcRNAs also influence the therapeutic resistance of gastric cancer. NcRNAs mainly consist of miRNAs, lncRNAs and circRNAs. In this paper, we summarized ncRNAs as biomarkers and therapeutic targets for gastric cancer, and also reviewed their role in clinical trials and diagnosis. We sum up different ncRNAs and related moleculars and signaling pathway in gastric cancer, like Bcl-2, PTEN, Wnt signaling. In addition, the potential clinical application of ncRNAs in overcoming chemotherapy and radiotherapy resistance in GC in the future were also focused on.
Collapse
Affiliation(s)
- Yanping Yue
- Department of Medical Oncology, Affiliated Cancer Hospital, Nantong University, Nantong, China
| | - Xinrong Lin
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinyue Qiu
- Department of Medical Oncology, Affiliated Cancer Hospital, Nantong University, Nantong, China
| | - Lei Yang
- Department of Medical Oncology, Affiliated Cancer Hospital, Nantong University, Nantong, China
| | - Rui Wang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Loss of E-Cadherin Leads to Druggable Vulnerabilities in Sphingolipid Metabolism and Vesicle Trafficking. Cancers (Basel) 2021; 14:cancers14010102. [PMID: 35008266 PMCID: PMC8749886 DOI: 10.3390/cancers14010102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/23/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Germline loss of the CDH1 gene is the primary genetic basis for hereditary diffuse gastric cancer, a disease resulting in elevated risk of both diffuse gastric cancer and lobular breast cancer. Current preventative treatment consists of prophylactic total gastrectomy, a therapy with several associated long-term morbidities. To address the lack of targeted molecular therapies for hereditary diffuse gastric cancer, we have utilized a synthetic lethal approach to identify candidate compounds that can specifically kill CDH1-null cells. Inhibitors of sphingolipid metabolism and vesicle trafficking pathways were identified as promising candidate compounds in a cell line model of CDH1 loss, then further validated in murine-derived organoid models of hereditary diffuse gastric cancer. With further research, these findings may lead to the development of novel chemoprevention strategies for the treatment of hereditary diffuse gastric cancer. Abstract Germline inactivating variants of CDH1 are causative of hereditary diffuse gastric cancer (HDGC), a cancer syndrome characterized by an increased risk of both diffuse gastric cancer and lobular breast cancer. Because loss of function mutations are difficult to target therapeutically, we have taken a synthetic lethal approach to identify targetable vulnerabilities in CDH1-null cells. We have previously observed that CDH1-null MCF10A cells exhibit a reduced rate of endocytosis relative to wildtype MCF10A cells. To determine whether this deficiency is associated with wider vulnerabilities in vesicle trafficking, we screened isogenic MCF10A cell lines with known inhibitors of autophagy, endocytosis, and sphingolipid metabolism. Relative to wildtype MCF10A cells, CDH1−/− MCF10A cells showed significantly greater sensitivity to several drugs targeting these processes, including the autophagy inhibitor chloroquine, the endocytosis inhibitors chlorpromazine and PP1, and the sphingosine kinase 1 inhibitor PF-543. Synthetic lethality was confirmed in both gastric and mammary organoid models of CDH1 loss, derived from CD44-Cre/Cdh1fl/fl/tdTomato mice. Collectively, these results suggest that both sphingolipid metabolism and vesicle trafficking represent previously unrecognised druggable vulnerabilities in CDH1-null cells and may lead to the development of new therapies for HDGC.
Collapse
|
8
|
Hu S, Liu H, Zhang J, Li S, Zhou H, Gao Y. Effects and prognostic values of miR-30c-5p target genes in gastric cancer via a comprehensive analysis using bioinformatics. Sci Rep 2021; 11:20584. [PMID: 34663825 PMCID: PMC8523699 DOI: 10.1038/s41598-021-00043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/30/2021] [Indexed: 12/02/2022] Open
Abstract
Gastric cancer (GC) is a common cancer and the leading cause of cancer-related death worldwide. To improve the diagnosis and treatment of GC, it is necessary to identify new biomarkers by investigating the cellular and molecular mechanisms. In this study, miR-30c-5p expression was significantly down-regulated in GC tissues by comprehensive analysis using multiple databases. The target genes of miR-30c-5p with up-regulated expression level in GC were identified, including ADAM12 (a disintegrin and metalloproteinase12), EDNRA (the Endothelin receptor type A), STC1 (stanniocalcin 1), and CPNE8 (the calcium-dependent protein, copine 8). The expression level of ADAM12 was significantly related to depth of invasion (p = 0.036) in GC patients. The expression level of EDNRA was significantly related to grade (P = 0.003), depth of invasion (P = 0.019), and lymphatic metastasis (P = 0.001). The expression level of CPNE8 was significantly related to grade (P = 0.043) and TNM stage (P = 0.027).Gene set enrichment analysis showed that they might participate in GC progression through cancer-related pathways. CIBERSORT algorithm analysis showed that their expressions were related to a variety of tumor-infiltrating immune cells. The higher expression of those target genes might be the independent risk factor for poor survival of GC patients, and they might be potential prognostic markers in GC patients.
Collapse
Affiliation(s)
- Shangshang Hu
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Huaifeng Liu
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Jinyan Zhang
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China.,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Shujing Li
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Huadong Zhou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.,Department of Neurology, Army Medical Center of PLA, Chongqing, 400038, China
| | - Yu Gao
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China. .,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui, China. .,School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, Anhui, China.
| |
Collapse
|
9
|
Li Z, Zhou Y, Tian G, Song M. Identification of Core Genes and Key Pathways in Gastric Cancer using Bioinformatics Analysis. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421080081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Yang N, Chen H, Huang Y, Song X, Yang P, Zhang S, Yan W, Li N, Feng Z. The Role and Significance of Wnt5a in Regulating Epithelial-Mesenchymal Transition in Endometrioid Adenocarcinoma. Cancer Manag Res 2021; 13:6527-6535. [PMID: 34447270 PMCID: PMC8384146 DOI: 10.2147/cmar.s319346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose As a non-classical ligand of Wnt, the abnormal regulation of Wnt5a contributes to the progression of malignant tumors; however, its effects differ depending on tumor type. Here, we evaluated the expression and significance of Wnt5a in endometrioid adenocarcinoma and its relationship with epithelial–mesenchymal transition (EMT)-related proteins. Patients and Methods Immunohistochemical streptavidin-peroxidase method and reverse transcription polymerase chain reaction (RT-PCR) method were used to analyze the expression and correlation of Wnt5a, and EMT-related protein β-catenin, E-cadherin and enhancer of zeste homolog 2 (EZH2) in endometrial cancer tissues and cell samples of each group. Results The expression of Wnt5a and E-cadherin decreased in the following order, normal endometrium > atypical hyperplasia endometrium > endometrioid adenocarcinoma. In contrast, the expression of β-catenin and EZH2 increased gradually. Moreover, Wnt5a expression was associated with the degree of tissue differentiation, International Federation of Gynecology and Obstetrics (FIGO) stage, and lymph node metastasis (all P<0.05). Wnt5a expression was also negatively correlated with β-catenin and EZH2 expression and positively correlated with E-cadherin expression. RT-PCR results further indicated that E-cadherin mRNA expression was upregulated in a Wnt5a-overexpressing Ishikawa cell line compared to cells transfected with an empty vector or negative control cells (P<0.01). Furthermore, the expression of EZH2 and β-catenin mRNA was downregulated in overexpressing cells compared to empty vector and negative control cells (P<0.01). Conclusion Wnt5a may elicit a suppressive effect on endometrioid adenocarcinoma by inhibiting EMT. This study provides a theoretical basis for the pathological diagnosis and targeted therapy of endometrioid adenocarcinoma and extends our understanding of the Wnt5a signaling pathway.
Collapse
Affiliation(s)
- Ningning Yang
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, Bengbu, 233000, People's Republic of China
| | - Hongchun Chen
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, Bengbu, 233000, People's Republic of China
| | - Yuchen Huang
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, Bengbu, 233000, People's Republic of China
| | - Xuexue Song
- Department of Pathology, First Affiliated Hospital of University of Science and Technology of China, Hefei, 230000, People's Republic of China
| | - Panpan Yang
- Department of Pathology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, People's Republic of China
| | - Shan Zhang
- Department of Pathology, Second People's Hospital of Hefei, Hefei, 230000, People's Republic of China
| | - Wentian Yan
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, Bengbu, 233000, People's Republic of China
| | - Nan Li
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, Bengbu, 233000, People's Republic of China
| | - Zhenzhong Feng
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, Bengbu, 233000, People's Republic of China
| |
Collapse
|
11
|
Zhao H, Hu H, Chen B, Xu W, Zhao J, Huang C, Xing Y, Lv H, Nie C, Wang J, He Y, Wang SQ, Chen XB. Overview on the Role of E-Cadherin in Gastric Cancer: Dysregulation and Clinical Implications. Front Mol Biosci 2021; 8:689139. [PMID: 34422902 PMCID: PMC8371966 DOI: 10.3389/fmolb.2021.689139] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023] Open
Abstract
Gastric cancer is the fifth most common cancer and the third most common cause of cancer death all over the world. E-cadherin encoded by human CDH1 gene plays important roles in tumorigenesis as well as in tumor progression, invasion and metastasis. Full-length E-cadhrin tethered on the cell membrane mainly mediates adherens junctions between cells and is involved in maintaining the normal structure of epithelial tissues. After proteolysis, the extracellular fragment of the full-length E-cadhein is released into the extracellular environment and the blood, which is called soluble E-cadherin (sE-cadherin). sE-cadherin promots invasion and metastasis as a paracrine/autocrine signaling molecule in the progression of various types of cancer including gastric cancer. This review mainly summarizes the dysregulation of E-cadherin and the regulatory roles in the progression, invasion, metastasis, and drug-resistance, as well as its clinical applications in diagnosis, prognosis, and therapeutics of gastric cancer.
Collapse
Affiliation(s)
- Huichen Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Weifeng Xu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jing Zhao
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Chen Huang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yishu Xing
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huifang Lv
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Caiyun Nie
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jianzheng Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yunduan He
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Sai-Qi Wang
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Xiao-Bing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Ramos MFKP, Pereira MA, de Mello ES, Cirqueira CDS, Zilberstein B, Alves VAF, Ribeiro-Junior U, Cecconello I. Gastric cancer molecular classification based on immunohistochemistry and in situ hybridization: Analysis in western patients after curative-intent surgery. World J Clin Oncol 2021; 12:688-701. [PMID: 34513602 PMCID: PMC8394162 DOI: 10.5306/wjco.v12.i8.688] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/09/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is a highly heterogeneous disease, and the identification of molecular subtyping of gastric adenocarcinoma emerged as a promising option to define therapeutic strategies and prognostic subgroups. However, the costs and technical complexity of molecular methodologies remains an obstacle to its adoption, and their clinical significance by other approaches needs further evidence.
AIM To evaluate the clinicopathological characteristics and long-term survival of GC based on the subgroups of molecular classification by immunohistochemistry (IHC) and in situ hybridization (ISH).
METHODS We retrospectively evaluated all patients who underwent D2-gastrectomy between 2009 and 2016 in a Western cohort of GC patients treated with curative intent. Microsatellite instability (MSI) status, E-cadherin, and p53 expression were analyzed by IHC, and Epstein-Barr virus (EBV) by ISH. Tissue microarrays were constructed for analysis. Clinicopathological characteristics and survival of GC were evaluated according to subtypes defined by The Cancer Genome Atlas (TCGA) Research Network Group and Asian Cancer Research Group (ACRG) classification systems.
RESULTS A total of 287 GC patients were included. Based on IHC and ISH analysis, five profiles were defined as follows: E-cadherin aberrant (9.1%), MSI (20.9%), p53 aberrant (36.6%), EBV positivity (10.5%), and p53 normal (31%), which corresponded to tumors that showed no alteration in another profile. A flowchart according to the TCGA and ACRG classifications were used to define the subtypes, where clinical and pathological characteristics associated with GC subtypes were evidenced. Proximal location (P < 0.001), total gastrectomy (P = 0.001), and intense inflammatory infiltrate (P < 0.001) were characteristics related to EBV subtype. MSI subtype was predominantly associated with advanced age (P = 0.017) and the presence of comorbidities (P = 0.011). While Laurén diffuse type (P < 0.001) and advanced stage (P = 0.029) were related to genomically stable (GS) subtype. GS tumors and microsatellite stable/epithelial to mesenchymal transition phenotype subtype had worse disease-free survival (DFS) and overall survival (OS) than other subtypes. Conversely, MSI subtype of GC had better survival in both classifications. Type of gastrectomy, pT and the TCGA subtypes were independent factors associated to DFS and OS.
CONCLUSION The IHC/ISH analysis was able to distinguish immunophenotypic groups of GC with distinct characteristics and prognosis, resembling the subtypes of the molecular classifications. Accordingly, this method of classification may represent a viable option for use in a clinical setting.
Collapse
Affiliation(s)
- Marcus Fernando Kodama Pertille Ramos
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01249000, Brazil
| | - Marina Alessandra Pereira
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01249000, Brazil
| | - Evandro Sobroza de Mello
- Department of Pathology, Instituto do Cancer, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 01249000, Brazil
| | | | - Bruno Zilberstein
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01249000, Brazil
| | - Venancio Avancini Ferreira Alves
- Department of Pathology, Instituto do Cancer, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 01249000, Brazil
| | - Ulysses Ribeiro-Junior
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01249000, Brazil
| | - Ivan Cecconello
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01249000, Brazil
| |
Collapse
|
13
|
Peng W, Du J, Dai W, Lan W. Predicting miRNA-Disease Association Based on Modularity Preserving Heterogeneous Network Embedding. Front Cell Dev Biol 2021; 9:603758. [PMID: 34178973 PMCID: PMC8223753 DOI: 10.3389/fcell.2021.603758] [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: 09/07/2020] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are a category of small non-coding RNAs that profoundly impact various biological processes related to human disease. Inferring the potential miRNA-disease associations benefits the study of human diseases, such as disease prevention, disease diagnosis, and drug development. In this work, we propose a novel heterogeneous network embedding-based method called MDN-NMTF (Module-based Dynamic Neighborhood Non-negative Matrix Tri-Factorization) for predicting miRNA-disease associations. MDN-NMTF constructs a heterogeneous network of disease similarity network, miRNA similarity network and a known miRNA-disease association network. After that, it learns the latent vector representation for miRNAs and diseases in the heterogeneous network. Finally, the association probability is computed by the product of the latent miRNA and disease vectors. MDN-NMTF not only successfully integrates diverse biological information of miRNAs and diseases to predict miRNA-disease associations, but also considers the module properties of miRNAs and diseases in the course of learning vector representation, which can maximally preserve the heterogeneous network structural information and the network properties. At the same time, we also extend MDN-NMTF to a new version (called MDN-NMTF2) by using modular information to improve the miRNA-disease association prediction ability. Our methods and the other four existing methods are applied to predict miRNA-disease associations in four databases. The prediction results show that our methods can improve the miRNA-disease association prediction to a high level compared with the four existing methods.
Collapse
Affiliation(s)
- Wei Peng
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology, Kunming, China.,Computer Technology Application Key Laboratory of Yunnan Province, Kunming University of Science and Technology, Kunming, China
| | - Jielin Du
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology, Kunming, China
| | - Wei Dai
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology, Kunming, China.,Computer Technology Application Key Laboratory of Yunnan Province, Kunming University of Science and Technology, Kunming, China
| | - Wei Lan
- Guangxi Key Laboratory of Multimedia Communications and Network Technology, Guangxi University, Nanning, China
| |
Collapse
|
14
|
Danilova NV, Mikhailov IA, Oleynikova NA, Malkov PG. [E-cadherin expression in tumor emboli in gastric cancer]. Arkh Patol 2021; 83:11-19. [PMID: 34041891 DOI: 10.17116/patol20218303111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the level of E-cadherin expression in tumor emboli, to compare it with expression in a tumor, to determine the dependence of E-cadherin expression in tumor emboli on the clinical and morphological characteristics of gastric cancer. MATERIAL AND METHODS We used samples of surgical material from 280 patients with a verified diagnosis of gastric cancer. E-cadherin expression was determined by immunohistochemical method. The results of the reactions were assessed semi-quantitatively and compared with the main clinical and morphological characteristics of gastric cancer (histological type according to the WHO classification 2019, histological type according to the classification of P. Lauren, clinical stage, depth of invasion (T), number of metastases in lymph nodes (N), presence or/absence of distant metastases (M), tumor localization in the stomach). RESULTS Among 280 cases of cancer, emboli were detected only in 67 cases, used for further analysis. The rest of the samples were excluded from the analysis, since emboli did not get into the sections during the cutting of immunohistochemical preparations. The expression of E-cadherin in tumor emboli was significantly higher (p<0.001) than in tumor tissue. At the same time, no cases identified where the level of E-cadherin decreased in emboli compared to the tumor. A significant increase in the expression of E-cadherin in tumor emboli compared to the primary tumor was noted for all histological types according to WHO 2019, for intermediate and diffuse types according to the P. Lauren classification (p<0.001). Comparison of expression in emboli and tumors for neoplasms with different depths of invasion (T), different stages and different localizations did not reveal statistically significant differences. An increase in the expression of E-cadherin in emboli compared to tumors was characterized by a higher level of significance in the presence of metastases (N1, N2, N3a, N3b; p<0.001) than in the absence of metastases (N0; p=0.016). CONCLUSION The study revealed a statistically significant increase in the expression of E-cadherin in tumor emboli compared to the primary tumor, which is evidence of its important role in maintaining the integrity of emboli and tumor dissemination.
Collapse
Affiliation(s)
- N V Danilova
- Lomonosov Moscow State University, Moscow, Russia
| | | | | | - P G Malkov
- Lomonosov Moscow State University, Moscow, Russia.,Russian Medical Academy for Continuous Professional Education, Moscow, Russia
| |
Collapse
|
15
|
Genetic and Epigenetic Alterations of CDH1 Regulatory Regions in Hereditary and Sporadic Gastric Cancer. Pharmaceuticals (Basel) 2021; 14:ph14050457. [PMID: 34066170 PMCID: PMC8151134 DOI: 10.3390/ph14050457] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
E-cadherin is a key player in gastric cancer (GC) and germline alterations of CDH1, its encoding gene, are responsible for Hereditary Diffuse Gastric Cancer (HDGC) syndrome. This study aimed at elucidating the role of genetic variants and DNA methylation of CDH1 promoter and enhancers in the regulation of gene expression. For this purpose, we analyzed genetic variants of the CDH1 gene through Next-Generation Sequencing (NGS) in a series of GC cell lines (NCI-N87, KATO-III, SNU-1, SNU-5, GK2, AKG, KKP) and the corresponding CDH1 expression levels. By bisulfite genomic sequencing, we analyzed the methylation status of CDH1 regulatory regions in 8 GC cell lines, in a series of 13 sporadic GC tissues and in a group of 20 HDGC CDH1-negative patients and 6 healthy controls. The NGS analysis on CDH1 coding and regulatory regions detected genetic alterations in 3 out of 5 GC cell lines lacking functional E-cadherin. CDH1 regulatory regions showed different methylation patterns in patients and controls, GC cell lines and GC tissues, expressing different E-cadherin levels. Our results showed that alterations in terms of genetic variants and DNA methylation patterns of both promoter and enhancers are associated with CDH1 expression levels and have a role in its regulation.
Collapse
|
16
|
Deshmukh AP, Vasaikar SV, Tomczak K, Tripathi S, den Hollander P, Arslan E, Chakraborty P, Soundararajan R, Jolly MK, Rai K, Levine H, Mani SA. Identification of EMT signaling cross-talk and gene regulatory networks by single-cell RNA sequencing. Proc Natl Acad Sci U S A 2021; 118:e2102050118. [PMID: 33941680 PMCID: PMC8126782 DOI: 10.1073/pnas.2102050118] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) plays a critical role during normal development and in cancer progression. EMT is induced by various signaling pathways, including TGF-β, BMP, Wnt-β-catenin, NOTCH, Shh, and receptor tyrosine kinases. In this study, we performed single-cell RNA sequencing on MCF10A cells undergoing EMT by TGF-β1 stimulation. Our comprehensive analysis revealed that cells progress through EMT at different paces. Using pseudotime clustering reconstruction of gene-expression profiles during EMT, we found sequential and parallel activation of EMT signaling pathways. We also observed various transitional cellular states during EMT. We identified regulatory signaling nodes that drive EMT with the expression of important microRNAs and transcription factors. Using a random circuit perturbation methodology, we demonstrate that the NOTCH signaling pathway acts as a key driver of TGF-β-induced EMT. Furthermore, we demonstrate that the gene signatures of pseudotime clusters corresponding to the intermediate hybrid EMT state are associated with poor patient outcome. Overall, this study provides insight into context-specific drivers of cancer progression and highlights the complexities of the EMT process.
Collapse
Affiliation(s)
- Abhijeet P Deshmukh
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Suhas V Vasaikar
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Katarzyna Tomczak
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Shubham Tripathi
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02115
| | - Petra den Hollander
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Emre Arslan
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Priyanka Chakraborty
- Centre for BioSystems Science and Engineering, Indian Institute of Science, 560012 Bangalore, India
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, 560012 Bangalore, India
| | - Kunal Rai
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030;
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02115;
- Department of Physics, Northeastern University, Boston, MA 02115
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030;
| |
Collapse
|
17
|
Liu X, Pu K, Wang Y, Chen Y, Zhou Y. Gastric cancer-associated microRNA expression signatures: integrated bioinformatics analysis, validation, and clinical significance. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:797. [PMID: 34268410 PMCID: PMC8246217 DOI: 10.21037/atm-21-1631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/29/2021] [Indexed: 12/21/2022]
Abstract
Background Gastric cancer (GC) is one of the common gastrointestinal malignancy worldwide and exhibits a poor prognosis. Increasing studies have indicated that microRNAs play critical roles in the cancer progression and have shown great potential as useful biomarkers. The search for potential diagnostic and prognostic biomarkers of gastric cancer (GC) with integrated bioinformatics analyses has been undertaken in previous studies. Methods In this study, the robust rank aggregation (RRA) method was used to perform an integrated analysis of differentially expressed miRNAs (DEMs) from five microarray datasets in the Gene Expression Omnibus (GEO) database to find robust biomarkers for GC. Ultimately, seven miRNAs were filtered from fourteen primary miRNAs using the validation set of The Cancer Genome Atlas (TCGA) database. Based on these results, diagnostic and survival analyses were performed, and logistic regression and Cox regression were used to determine the clinicopathological characteristics of the DEM expression and overall survival. Results Nine eligible miRNA datasets related to GC were selected from the GEO database for integrated analysis in this study. Diagnostic analysis implied that these miRNAs could be regarded as promising candidate diagnostic biomarkers in GC tissues, but whether the results of the tissue analysis are consistent with those of peripheral blood analysis requires further validation. The logistic regression indicated that the ectopic expression of these DEMs was relevant to the histological type, anatomical region, and pathological grade of GC. However, the survival and Cox regression analyses suggested that the poor prognosis of GC patients was not strongly dependent on the ectopic expression of the seven miRNAs, but rather, a poor prognosis was associated with age, metastasis, and histological grade. Conclusions Based on the results presented in this study it can be concluded that these miRNAs (miR-455-3p, miR-135b-5p, let-7a-3p, miR-195-5p, miR-204-5p, miR-149-5p, and miR-143-3p) might be potential biomarkers for the early diagnosis of GC patients, but this finding should be regarded with caution. A large-scale, prospective, and multicenter cohort study should be performed.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China.,Department of Rheumatology and Immunology, The First Hospital of Lanzhou University, Lanzhou, China.,Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ke Pu
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China.,Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China.,Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yanfei Chen
- Department of Rheumatology and Immunology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou University, Lanzhou, China.,Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Fernandes Â, Dias AM, Silva MC, Gaifem J, Azevedo CM, Carballo I, Pinho SS. The Role of Glycans in Chronic Inflammatory Gastrointestinal and Liver Disorders and Cancer. COMPREHENSIVE GLYCOSCIENCE 2021:444-470. [DOI: 10.1016/b978-0-12-819475-1.00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
MicroRNAs: Diverse Mechanisms of Action and Their Potential Applications as Cancer Epi-Therapeutics. Biomolecules 2020; 10:biom10091285. [PMID: 32906681 PMCID: PMC7565521 DOI: 10.3390/biom10091285] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/10/2020] [Accepted: 09/02/2020] [Indexed: 12/23/2022] Open
Abstract
Usually, miRNAs function post-transcriptionally, by base-pairing with the 3′UTR of target mRNAs, repressing protein synthesis in the cytoplasm. Furthermore, other regions including gene promoters, as well as coding and 5′UTR regions of mRNAs are able to interact with miRNAs. In recent years, miRNAs have emerged as important regulators of both translational and transcriptional programs. The expression of miRNA genes, similar to protein-coding genes, can be epigenetically regulated, in turn miRNA molecules (named epi-miRs) are able to regulate epigenetic enzymatic machinery. The most recent line of evidence indicates that miRNAs can influence physiological processes, such as embryonic development, cell proliferation, differentiation, and apoptosis as well as pathological processes (e.g., tumorigenesis) through epigenetic mechanisms. Some tumor types show repression of tumor-suppressor epi-miRs resulting in cancer progression and metastasis, hence these molecules have become novel therapeutic targets in the last few years. This review provides information about miRNAs involvement in the various levels of transcription and translation regulation, as well as discusses therapeutic potential of tumor-suppressor epi-miRs used in in vitro and in vivo anti-cancer therapy.
Collapse
|
20
|
Bakare OO, Fadaka AO, Keyster M, Pretorius A. Structural and Molecular Docking Analytical Studies of the Predicted Ligand Binding Sites of Cadherin-1 in Cancer Prognostics. Adv Appl Bioinform Chem 2020; 13:1-9. [PMID: 32821128 PMCID: PMC7419610 DOI: 10.2147/aabc.s253851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Several studies have explored the design of antimicrobial peptides (AMPs) for the development of therapeutic and diagnostic molecules for the treatment and identification of pathogenic diseases as well as cancer. Human cadherin-1 protein has been identified to be involved in adhesion-mediated signalling pathways in normal cells and its loss through genetic and epigenetic alterations can result in an enhanced invasion and metastasis of malignancy in tumours. Therefore, the identification of cadherin during treatment of cancer can be used as prognostic biomarker to establish the responsiveness of patients to treatment regimen. Antimicrobial peptides (AMPs) offer several compensatory advantages in biomedical applications and have been used for treatment of diseases, dietary supplements and diagnosis of diseases. The aim of this research work was to use in silico approaches to analyse retrieved human cadherin-1 as prognostic targets in cancer treatments using modelled putative anticancer AMPs. METHODS The structures of the putative AMPs and cadherin-1 were modelled using I-TASSER server and the protein overall quality was validated using PROCHECK. Thereafter, the protein motifs were predicted and the molecular interaction between the putative anticancer AMPs and protein was carried out using PatchDock. RESULTS The results revealed that all the AMPs were good prognostic molecules for cancer with BOO1 having the highest binding affinity of 15,874. CONCLUSION This study revealed that all the generated AMPs have good prognostic value for monitoring the progress of cancer treatment using human cadherin-1 as receptor. This is the first report where AMPs were used in prognostics of cancer using human cadherin-1.
Collapse
Affiliation(s)
| | | | - Marshall Keyster
- Biotechnology Department, University of the Western Cape, Cape Town7535, South Africa
| | - Ashley Pretorius
- Bioinformatics Research Group, University of the Western Cape, Cape Town7535, South Africa
| |
Collapse
|
21
|
Zeng W, Zhang S, Yang L, Wei W, Gao J, Guo N, Wu F. Serum miR-101-3p combined with pepsinogen contributes to the early diagnosis of gastric cancer. BMC MEDICAL GENETICS 2020; 21:28. [PMID: 32041551 PMCID: PMC7011268 DOI: 10.1186/s12881-020-0967-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND This study aimed to explore the diagnostic value of serum miR-101-3p combined with pepsinogen (PG) on early diagnosis of gastric cancer (GC). METHODS A total of 61 atrophic gastritis (AG) and 86 GC patients, and 50 healthy volunteers were enrolled. The serum expression of miR-101-3p was measured by qRT-PCR. The serum content of carcinoembryonic antigen (CEA) was measured by Electrochemiluminescence immunoassay. The serum contents of PGI and PGII were measured by Enzyme linked immunosorbent assay. The diagnostic value of serum markers on AG and GC was analyzed by receiver operating characteristic (ROC) analysis. RESULTS The expression of miR-101-3p, the content of PGI and the ratio of PGI/II were significantly decreased, and the content of PGII was significantly increased in AG patients compared with those in normal controls. The changes of the above serum indicators were more obvious in GC patients than those in AG patients. The content of CEA was significantly higher in GC patients than that in AG patients. In addition, the expression of miR-101-3p was negatively associated with the submucosal infiltration in GC patients. MiR-101-3p exhibited high diagnostic value on AG (AUC 0.8493, sensitivity 80.33%, specificity 80%) and GC (AUC 0.8749, sensitivity 72.09%, specificity 86.49%). MiR-101-3p + PGI + PGI/II (AUC 0.856, sensitivity 80.23%, specificity 77.05%) exhibited a high diagnostic value in distinguishing between AG and GC. CONCLUSIONS MiR-101-3p was a potential diagnostic marker for AG and GC. MiR-101-3p + PGI + PGI/II was effective in distinguishing between AG and GC.
Collapse
Affiliation(s)
- Weiwei Zeng
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China.
| | - Shuxiang Zhang
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Lei Yang
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Wenchao Wei
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Jie Gao
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Ni Guo
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| | - Fengting Wu
- Department of Gastroenterology, Dongying People's Hospital, No. 317, Chengnan First Road, Dongying City, 257091, Shandong Province, China
| |
Collapse
|
22
|
Moridnia A, Tabatabaiefar MA, Zeinalian M, Minakari M, Kheirollahi M, Moghaddam NA. Novel Variants and Copy Number Variation in CDH1 Gene in Iranian Patients with Sporadic Diffuse Gastric Cancer. J Gastrointest Cancer 2020; 50:420-427. [PMID: 29577179 DOI: 10.1007/s12029-018-0082-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The aim of this study was to survey the nucleotide changes and copy number variations (CNV) in the CDH1 gene in Iranian patients with sporadic diffuse gastric cancer (SDGC). MATERIALS AND METHODS In this study, 28 patients were examined who upon gastrectomy had been diagnosed with SDGC according to the familial history and histopathological criteria which was confirmed by the pathologist. DNA extraction was performed from formalin-fixed paraffin-embedded tissues using a phenol-chloroform method following xylene deparaffinization. Determination of DNA sequence by Sanger was performed using PCR amplification of 16 exons and boundaries of intron/exon of CDH1 gene. Multiplex ligation-dependent probe amplification (MLPA) was performed on patients with pathogenic disorders in the sequence. RESULTS In total, patients included 20 males and 8 females. Of all patients, 12 patients were under 45 years old (early onset gastric cancer, EODC) and 16 patients were older. The tumor was diagnosed in the early TNM stage (I, II) in six patients and in late stages (III, IV) in 19 cases. Altogether, 16 variants (three exonic with one new variant and 13 intronic with nine new variants) were found in DNA sequencing of the CDH1 gene in five samples. Also, using MLPA, a new duplication in exon 9 and one deletion in exon 2 were detected in two other patients. Altogether, CDH1 variants were identified in seven out of 28 patients (25%). CONCLUSION Our study revealed several novel somatic variants in the CDH1 gene in Iranian patients with sporadic diffuse GC. Our data supports the hypothesis that mutations in CDH1 gene, and particularly the mutations we describe, should be considered, even in sporadic cases of gastric cancer. The presence of these mutations in patients raises important issues regarding genetic counseling and diagnostic test in DGC patients.
Collapse
Affiliation(s)
- Abbas Moridnia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable disease and Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, P.O.Box: 81746-73461, Isfahan, Iran
| | - Mohammad Amin Tabatabaiefar
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable disease and Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, P.O.Box: 81746-73461, Isfahan, Iran
| | - Mehrdad Zeinalian
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable disease and Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, P.O.Box: 81746-73461, Isfahan, Iran
| | - Mohammad Minakari
- Internal medicine department, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Majid Kheirollahi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable disease and Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, P.O.Box: 81746-73461, Isfahan, Iran.
| | - Noushin Afshar Moghaddam
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
23
|
Varanda AS, Santos M, Soares AR, Vitorino R, Oliveira P, Oliveira C, Santos MAS. Human cells adapt to translational errors by modulating protein synthesis rate and protein turnover. RNA Biol 2020; 17:135-149. [PMID: 31570039 PMCID: PMC6948982 DOI: 10.1080/15476286.2019.1670039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 02/08/2023] Open
Abstract
Deregulation of tRNAs, aminoacyl-tRNA synthetases (aaRS) or tRNA modifying enzymes, increase the level of protein synthesis errors (PSE) and are associated with several diseases, but the cause-effect mechanisms of these pathologies remain elusive. To clarify the role of PSE in human biology, we have engineered a HEK293 cell line to overexpress a wild type (Wt) tRNASer and two tRNASer mutants that misincorporate serine at non-cognate codon sites. Then, we followed long-term adaptation to PSE of such recombinant cells by analysing cell viability, protein synthesis rate and activation of protein quality control mechanisms (PQC). Engineered cells showed higher level of misfolded and aggregated proteins; activated the ubiquitin-proteasome system (UPS) and the unfolded protein response (UPR), indicative of proteotoxic stress. Adaptation to PSE involved increased protein turnover, UPR up-regulation and altered protein synthesis rate. Gene expression analysis showed that engineered cells presented recurrent alterations in the endoplasmic reticulum, cell adhesion and calcium homeostasis. Herein, we unveil new phenotypic consequences of protein synthesis errors in human cells and identify the protein quality control processes that are necessary for long-term adaptation to PSE and proteotoxic stress. Our data provide important insight on how chronic proteotoxic stress may cause disease and highlight potential biological pathways that support the association of PSE with disease.
Collapse
Affiliation(s)
- Ana Sofia Varanda
- Department of Medical Sciences and Institute of Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Expression Regulation in Cancer, Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Mafalda Santos
- Department of Medical Sciences and Institute of Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Expression Regulation in Cancer, Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Ana R. Soares
- Department of Medical Sciences and Institute of Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- Department of Medical Sciences and Institute of Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| | - Patrícia Oliveira
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Expression Regulation in Cancer, Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Carla Oliveira
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Expression Regulation in Cancer, Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Manuel A. S. Santos
- Department of Medical Sciences and Institute of Biomedicine – iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
24
|
Huang Z, Wu X, Li J. miR-101 suppresses colon cancer cell migration through regulation of EZH2. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2020; 113:255-260. [DOI: 10.17235/reed.2020.6800/2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Rossi T, Tedaldi G, Petracci E, Abou Khouzam R, Ranzani GN, Morgagni P, Saragoni L, Monti M, Calistri D, Ulivi P, Molinari C. E-cadherin Downregulation and microRNAs in Sporadic Intestinal-Type Gastric Cancer. Int J Mol Sci 2019; 20:ijms20184452. [PMID: 31509966 PMCID: PMC6769612 DOI: 10.3390/ijms20184452] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022] Open
Abstract
CDH1 gene, encoding E-cadherin, is a tumor suppressor gene frequently altered in gastric cancers (GCs) of both diffuse (DGC) and intestinal (IGC) histotypes, albeit through different mechanisms. The study aimed to characterize CDH1 expression in sporadic IGC and to investigate whether microRNAs (miRs) are involved in its transcriptional control. We evaluated CDH1 expression by quantitative real-time PCR (RT-qPCR) in 33 IGC patients and found a significant downregulation in tumor tissues compared to normal counterparts (p-value = 0.025). Moreover, 14 miRs, predicted to be involved in CDH1 regulation in both a direct and indirect manner, were selected and analyzed by RT-qPCR in an independent case series of 17 IGCs and matched normal tissues. miR-101, miR-26b, and miR-200c emerged as significantly downregulated and were confirmed in the case series of 33 patients (p-value < 0.001). Finally, we evaluated EZH2 expression, a target of both miR-101 and miR-26b, which showed significant upregulation in IGCs (p-value = 0.005). A significant inverse correlation was observed between EZH2 overexpression and CDH1, miR-101, and miR-26b levels (p-value < 0.001). Our results reinforce the link between CDH1 and IGC, highlighting the role of miRs in its transcriptional control and improving our understanding of GC subtypes and biomarkers.
Collapse
Affiliation(s)
- Tania Rossi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Gianluca Tedaldi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Raefa Abou Khouzam
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | | | - Paolo Morgagni
- General and Oncologic Surgery, Department of Surgery, G.B. Morgagni L.Pierantoni General Hospital, AUSL Romagna, 47121 Forlì, Italy.
| | - Luca Saragoni
- Department of Pathology, AUSL Romagna, Morgagni-Pierantoni Hospital, 47121 Forlì, Italy.
| | - Manlio Monti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Chiara Molinari
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| |
Collapse
|
26
|
Zare A, Ganji M, Omrani MD, Alipoor B, Ghaedi H. Gastric Cancer MicroRNAs Meta-signature. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:94-102. [PMID: 32215261 DOI: 10.22088/ijmcm.bums.8.2.94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022]
Abstract
Gastric cancer (GC) is one of the most common types of cancer and the second leading cause of cancer-associated mortality. Identification of novel biomarkers is critical to prolonging patient survival. MicroRNAs (miRNAs) proved to play diverse roles in the physiological and pathological state in cancers including GC. Herein we aimed at performing a meta-analysis on miRNA profiling studies that used microarray platforms. Relevant studies were retrieved from PubMed and GEO databases. We used the robust rank aggregation to perform the meta-analysis. Moreover, for meta-signature miRNAs target genes, we performed pathway enrichment and GO molecular function enrichment analysis. A total of 19 upregulated miRNAs and seven downregulated miRNAs in GC samples were identified. However, only three upregulated and one downregulated miRNA reached statistical significance after multiple test correction. Here we showed that hsa-miR-21-5p, hsa-miR-93-5p, hsa-miR-25-3p, and hsa-miR-375 are differentially expressed in GC samples.
Collapse
Affiliation(s)
- Ali Zare
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maziar Ganji
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Alipoor
- Department of Laboratory Sciences, Faculty of Para medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Chen Z, Ju H, Zhao T, Yu S, Li P, Jia J, Li N, Jing X, Tan B, Li Y. hsa_circ_0092306 Targeting miR-197-3p Promotes Gastric Cancer Development by Regulating PRKCB in MKN-45 Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:617-626. [PMID: 31689616 PMCID: PMC6838893 DOI: 10.1016/j.omtn.2019.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 07/11/2019] [Accepted: 08/07/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide and is thus a global cancer burden. Here, we focused on a novel circular RNA hsa_circ_0092306 and explored the potential molecular mechanism to provide a new target for and novel insights into GC treatment. The GEO microarray was mined and analyzed with R software. Sanger sequencing and RNase R assay were applied to verify the identification of hsa_circ_0092306. Quantitative real-time PCR and western blot were performed to measure the mRNA and protein levels. Pull-down and luciferase reporter assays were conducted to confirm the target relationships. Annexin V-PI apoptosis flow cytometry, 3-(4,5Dimethylthiazol- yl)-2,5Dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide (MTT), wound healing, and Transwell assays were applied to detect cell apoptosis, viability, migration, and invasion in MKN-45 cells, respectively. A xenograft in vivo experiment was conducted to confirm the cell experiment results. hsa_circ_0092306 was upregulated in GC tissues and GC cells, and promoted GC development in MKN-45 cells. hsa_circ_0092306 inhibited tumor suppressor miR-197-3p expression but promoted tumor promotor protein kinase C beta (PRKCB) expression in MKN-45 cells. hsa_circ_0092306 and PRKCB had a common target (miR-197-3p) and were negatively related to miR-197-3p expression. hsa_circ_0092306 promoted the development of GC by regulating the pathway of miR-197-3p/PRKCB in MKN-45 cells.
Collapse
Affiliation(s)
- Zihao Chen
- Graduate School of Hebei Medical University, Shijiazhuang 050017, Hebei, China; The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Hongping Ju
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China; The Respiratory System Disease Prevention and Control of Public Service Platform of Science and Technology in Yunnan Province, Kunming 650214, Yunnan, China.
| | - Ting Zhao
- Graduate School of Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Shan Yu
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Ping Li
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Jing Jia
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Nan Li
- School of Medicine, Kunming University, Kunming 650214, Yunnan, China
| | - Xiaojie Jing
- Department of Medicine, The People's Hospital of Economic and Technological Development Zone, Kunming 650217, Yunnan, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China.
| |
Collapse
|
28
|
Bure IV, Nemtsova MV, Zaletaev DV. Roles of E-cadherin and Noncoding RNAs in the Epithelial-mesenchymal Transition and Progression in Gastric Cancer. Int J Mol Sci 2019; 20:ijms20122870. [PMID: 31212809 PMCID: PMC6627057 DOI: 10.3390/ijms20122870] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023] Open
Abstract
The epithelial–mesenchymal transition (EMT) is thought to be at the root of invasive and metastatic cancer cell spreading. E-cadherin is an important player in this process, which forms the structures that establish and maintain cell–cell interactions. A partial or complete loss of E-cadherin expression in the EMT is presumably mediated by mechanisms that block the expression of E-cadherin regulators and involve the E-cadherin-associated transcription factors. The protein is involved in several oncogenic signaling pathways, such as the Wnt/β-catenin, Rho GTPase, and EGF/EGFR, whereby it plays a role in many tumors, including gastric cancer. Such noncoding transcripts as microRNAs and long noncoding RNAs—critical components of epigenetic control of gene expression in carcinogenesis—contribute to regulation of the E-cadherin function by acting directly or through numerous factors controlling transcription of its gene, and thus affecting not only cancer cell proliferation and metastasis, but also the EMT. This review focuses on the role of E-cadherin and the non-coding RNAs-mediated mechanisms of its expressional control in the EMT during stomach carcinogenesis.
Collapse
Affiliation(s)
- Irina V Bure
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia.
| | - Marina V Nemtsova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia.
- Research Centre for Medical Genetics, Moskvorechie st., 1, Moscow 115522, Russia.
| | - Dmitry V Zaletaev
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia.
- Research Centre for Medical Genetics, Moskvorechie st., 1, Moscow 115522, Russia.
| |
Collapse
|
29
|
Rocha S, Teles SP, Azevedo M, Oliveira P, Carvalho J, Oliveira C. Gastric Cancer Extracellular Vesicles Tune the Migration and Invasion of Epithelial and Mesenchymal Cells in a Histotype-Dependent Manner. Int J Mol Sci 2019; 20:ijms20112608. [PMID: 31141946 PMCID: PMC6600627 DOI: 10.3390/ijms20112608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/26/2022] Open
Abstract
Extracellular vesicles (EVs) secreted by tumor cells modulate recipient cells' behavior, but their effects in normal cells from the tumor microenvironment remain poorly known. In this study, we dissected the functional impact of gastric cancer cell-derived EVs (GC-EVs), representative of distinct GC histotypes, on the behavior of normal isogenic epithelial and mesenchymal cells. GC-EVs were isolated by differential centrifugation and characterized by transmission electron microscopy, nanoparticle tracking analysis, and imaging flow-cytometry. Epithelial and mesenchymal cells were challenged with GC-EVs and submitted to proliferation, migration, and invasion assays. Expression of epithelial and mesenchymal markers was followed by immunofluorescence and flow-cytometry. Our results indicated that GC-EVs secreted by diffuse-type cancer cells decrease the migration of recipient cells. This effect was more prominent and persistent for mesenchymal recipient cells, which also increased Fibronectin expression in response to EVs. GC-EVs secreted by cancer cells derived from tumors with an intestinal component increased invasion of recipient epithelial cells, without changes in EMT markers. In summary, this study demonstrated that GC-EVs modulate the migration and invasion of epithelial and mesenchymal cells from the tumor microenvironment, in a histotype-dependent manner, highlighting new features of intestinal and diffuse-type GC cells, which may help explaining differential metastasis patterns and aggressiveness of GC histotypes.
Collapse
Affiliation(s)
- Sara Rocha
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| | - Sara Pinto Teles
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
| | - Mafalda Azevedo
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Patrícia Oliveira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
| | - Joana Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
| | - Carla Oliveira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
- Department Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
30
|
Li T, Gao X, Han L, Yu J, Li H. Identification of hub genes with prognostic values in gastric cancer by bioinformatics analysis. World J Surg Oncol 2018; 16:114. [PMID: 29921304 PMCID: PMC6009060 DOI: 10.1186/s12957-018-1409-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Background Gastric cancer (GC) is a prevalent malignant cancer of digestive system. To identify key genes in GC, mRNA microarray GSE27342, GSE29272, and GSE33335 were downloaded from GEO database. Methods Differentially expressed genes (DEGs) were obtained using GEO2R. DAVID database was used to analyze function and pathways enrichment of DEGs. Protein-protein interaction (PPI) network was established by STRING and visualized by Cytoscape software. Then, the influence of hub genes on overall survival (OS) was performed by the Kaplan-Meier plotter online tool. Module analysis of the PPI network was performed using MCODE. Additionally, potential stem loop miRNAs of hub genes were predicted by miRecords and screened by TCGA dataset. Transcription factors (TFs) of hub genes were detected by NetworkAnalyst. Results In total, 67 DEGs were identified; upregulated DEGs were mainly enriched in biological process (BP) related to angiogenesis and extracellular matrix organization and the downregulated DEGs were mainly enriched in BP related to ion transport and response to bacterium. KEGG pathways analysis showed that the upregulated DEGs were enriched in ECM-receptor interaction and the downregulated DEGs were enriched in gastric acid secretion. A PPI network of DEGs was constructed, consisting of 43 nodes and 87 edges. Twelve genes were considered as hub genes owing to high degrees in the network. Hsa-miR-29c, hsa-miR-30c, hsa-miR-335, hsa-miR-33b, and hsa-miR-101 might play a crucial role in hub genes regulation. In addition, the transcription factors-hub genes pairs were displayed with 182 edges and 102 nodes. The high expression of 7 out of 12 hub genes was associated with worse OS, including COL4A1, VCAN, THBS2, TIMP1, COL1A2, SERPINH1, and COL6A3. Conclusions The miRNA and TFs regulation network of hub genes in GC may promote understanding of the molecular mechanisms underlying the development of gastric cancer and provide potential targets for GC diagnosis and treatment. Electronic supplementary material The online version of this article (10.1186/s12957-018-1409-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Xujie Gao
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Lei Han
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Hui Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China. .,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China. .,National Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
31
|
Lima JF, Carvalho J, Pinto-Ribeiro I, Almeida C, Wengel J, Cerqueira L, Figueiredo C, Oliveira C, Azevedo NF. Targeting miR-9 in gastric cancer cells using locked nucleic acid oligonucleotides. BMC Mol Biol 2018; 19:6. [PMID: 29879907 PMCID: PMC5992815 DOI: 10.1186/s12867-018-0107-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 05/28/2018] [Indexed: 12/21/2022] Open
Abstract
Background Gastric cancer is the third leading cause of cancer-related mortality worldwide. Recently, it has been demonstrated that gastric cancer cells display a specific miRNA expression profile, with increasing evidence of the role of miRNA-9 in this disease. miRNA-9 upregulation has been shown to influence the expression of E-cadherin-encoding gene, triggering cell motility and invasiveness. Results In this study, we designed LNA anti-miRNA oligonucleotides with a complementary sequence to miRNA-9 and tested their properties to both detect and silence the target miRNA. We could identify and visualize the in vitro uptake of low-dosing LNA-based anti-miRNA oligonucleotides without any carrier or transfection agent, as early as 2 h after the addition of the oligonucleotide sequence to the culture medium. Furthermore, we were able to assess the silencing potential of miRNA-9, using different LNA anti-miRNA oligonucleotide designs, and to observe its subsequent effect on E-cadherin expression. Conclusions The administration of anti-miRNA sequences even at low-doses, rapidly repressed the target miRNA, and influenced the expression of E-cadherin by significantly increasing its levels.
Collapse
Affiliation(s)
- Joana Filipa Lima
- Department of Chemical Engineering, LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal. .,Biomode, 2 S.A., Braga, Portugal. .,i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.
| | - Joana Carvalho
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Inês Pinto-Ribeiro
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,FMUP, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Carina Almeida
- National Institute for Agricultural and Veterinary Research (INIAV), Vairão, Vila do Conde, Portugal
| | - Jesper Wengel
- Department of Physics, Chemistry and Pharmacy, Nucleic Acid Center, University of Southern Denmark, Odense, Denmark
| | - Laura Cerqueira
- Department of Chemical Engineering, LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal.,Biomode, 2 S.A., Braga, Portugal
| | - Céu Figueiredo
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,FMUP, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Carla Oliveira
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,FMUP, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Nuno Filipe Azevedo
- Department of Chemical Engineering, LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| |
Collapse
|
32
|
Antognelli C, Cecchetti R, Riuzzi F, Peirce MJ, Talesa VN. Glyoxalase 1 sustains the metastatic phenotype of prostate cancer cells via EMT control. J Cell Mol Med 2018; 22:2865-2883. [PMID: 29504694 PMCID: PMC5908125 DOI: 10.1111/jcmm.13581] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/23/2018] [Indexed: 01/07/2023] Open
Abstract
Metastasis is the primary cause of death in prostate cancer (PCa) patients. Effective therapeutic intervention in metastatic PCa is undermined by our poor understanding of its molecular aetiology. Defining the mechanisms underlying PCa metastasis may lead to insights into how to decrease morbidity and mortality in this disease. Glyoxalase 1 (Glo1) is the detoxification enzyme of methylglyoxal (MG), a potent precursor of advanced glycation end products (AGEs). Hydroimidazolone (MG-H1) and argpyrimidine (AP) are AGEs originating from MG-mediated post-translational modification of proteins at arginine residues. AP is involved in the control of epithelial to mesenchymal transition (EMT), a crucial determinant of cancer metastasis and invasion, whose regulation mechanisms in malignant cells are still emerging. Here, we uncover a novel mechanism linking Glo1 to the maintenance of the metastatic phenotype of PCa cells by controlling EMT by engaging the tumour suppressor miR-101, MG-H1-AP and TGF-β1/Smad signalling. Moreover, circulating levels of Glo1, miR-101, MG-H1-AP and TGF-β1 in patients with metastatic compared with non-metastatic PCa support our in vitro results, demonstrating their clinical relevance. We suggest that Glo1, together with miR-101, might be potential therapeutic targets for metastatic PCa, possibly by metformin administration.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Rodolfo Cecchetti
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Francesca Riuzzi
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | - Matthew J. Peirce
- Department of Experimental MedicineUniversity of PerugiaPerugiaItaly
| | | |
Collapse
|
33
|
Segovia C, Martínez-Fernández M, Dueñas M, Rubio C, López-Calderón FF, Costa C, Saiz-Ladera C, Fernández-Grajera M, Duarte J, Muñoz HG, de la Rosa F, Villacampa F, Castellano D, Paramio JM. Opposing roles of PIK3CA gene alterations to EZH2 signaling in non-muscle invasive bladder cancer. Oncotarget 2018; 8:10531-10542. [PMID: 28060766 PMCID: PMC5354678 DOI: 10.18632/oncotarget.14453] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/12/2016] [Indexed: 12/03/2022] Open
Abstract
The high rates of tumor recurrence and progression represent a major clinical problem in non-muscle invasive bladder cancer. Previous data showed that EZH2-dependent signaling mediates these processes, whereas the frequent alterations of PIK3CA gene (copy gains and mutations) are predictive of reduced recurrence. Here we show, using clinical samples and bladder cancer cell lines, a functional interaction between EZH2- and PIK3CA-dependent signaling pathways. PIK3CA alterations mediated, on the one hand, the increased expression of two miRNAs, miR-101 and miR-138, which posttranscriptionally downregulate EZH2 expression. On the other hand, PIK3CA alterations facilitate the activation of Akt which phosphorylates EZH2 on Ser21, precluding the trimethylation of histone H3 in K27. Remarkably the increased expression of miR101 or miR138 and the expression of Ser21-phosphorylated EZH2 are good prognostic factors regarding non-muscle invasive bladder cancer recurrence and progression. Collectively, this study provides molecular evidences indicating that the gene expression rewiring occurring in primary bladder tumors, associated with increased EZH2 expression and activity and mediating the increased recurrence and progression risk, are prevented by PIK3CA-dependent signaling. This molecular process may have deep implications in the management of bladder cancer patients and in the design of novel molecularly targeted therapeutic approaches.
Collapse
Affiliation(s)
- Cristina Segovia
- Unidad de Oncología Molecular, CIEMAT (ed70A), Madrid, Spain.,Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Mónica Martínez-Fernández
- Unidad de Oncología Molecular, CIEMAT (ed70A), Madrid, Spain.,Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Marta Dueñas
- Unidad de Oncología Molecular, CIEMAT (ed70A), Madrid, Spain.,Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Carolina Rubio
- Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | | | - Clotilde Costa
- Unidad de Oncología Molecular, CIEMAT (ed70A), Madrid, Spain.,Unidad Mixta Roche-CHUS, Hospital Universitario de Santiago de Compostela, Travesía de Choupana, s/n, Santiago de Compostela, A Coruña, Spain
| | - Cristina Saiz-Ladera
- Unidad de Oncología Molecular, CIEMAT (ed70A), Madrid, Spain.,Departamento de Bioquímica y Biología Molecular I, Facultad de Biología, Universidad Complutense, Madrid, Spain
| | | | - José Duarte
- Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Huberto García Muñoz
- Servicio de Anatomía Patológica, Hospital Universitario 12 de Octubre, Instituto de Investigación 12 de Octubre i+12, UCM, Av Cordoba s/n, Madrid, Spain
| | - Federico de la Rosa
- Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Felipe Villacampa
- Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Daniel Castellano
- Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| | - Jesús M Paramio
- Unidad de Oncología Molecular, CIEMAT (ed70A), Madrid, Spain.,Grupo de Oncología celular y Molecular, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER ONC), Spain
| |
Collapse
|
34
|
de Brot S, S. Rutland C, P. Mongan N, James V. Epigenetic Control of MicroRNA Expression and Cancer. CANCER AND NONCODING RNAS 2018:373-380. [DOI: 10.1016/b978-0-12-811022-5.00020-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Integrated MicroRNA-mRNA Analysis Reveals miR-204 Inhibits Cell Proliferation in Gastric Cancer by Targeting CKS1B, CXCL1 and GPRC5A. Int J Mol Sci 2017; 19:ijms19010087. [PMID: 29283424 PMCID: PMC5796037 DOI: 10.3390/ijms19010087] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/23/2017] [Accepted: 12/25/2017] [Indexed: 01/07/2023] Open
Abstract
Gastric cancer (GC) is the second most frequent cause of cancer-related deaths worldwide. MicroRNAs are single-stranded RNA molecules of 21–23 nucleotides that regulate target gene expression through specific base-pairing interactions between miRNA and untranslated regions of targeted mRNAs. In this study, we generated a multistep approach for the integrated analysis of miRNA and mRNA expression. First, both miRNA and mRNA expression profiling datasets in gastric cancer from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) identified 79 and 1042 differentially expressed miRNAs and mRNAs, respectively, in gastric cancer. Second, inverse correlations between miRNA and mRNA expression levels identified 3206 miRNA–mRNA pairs combined with 79 dysregulated miRNAs and their 774 target mRNAs predicted by three prediction tools, miRanda, PITA, and RNAhybrid. Additionally, miR-204, which was found to be down-regulated in gastric cancer, was ectopically over-expressed in the AGS gastric cancer cell line and all down-regulated targets were identified by RNA sequencing (RNA-seq) analysis. Over-expression of miR-204 reduced the gastric cancer cell proliferation and suppressed the expression of three targets which were validated by qRT-PCR and luciferase assays. For the first time, we identified that CKS1B, CXCL1, and GPRC5A are putative targets of miR-204 and elucidated that miR-204 acted as potential tumor suppressor and, therefore, are useful as a promising therapeutic target for gastric cancer.
Collapse
|
36
|
Predicting the Functional Impact of CDH1 Missense Mutations in Hereditary Diffuse Gastric Cancer. Int J Mol Sci 2017; 18:ijms18122687. [PMID: 29231860 PMCID: PMC5751289 DOI: 10.3390/ijms18122687] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022] Open
Abstract
The role of E-cadherin in Hereditary Diffuse Gastric Cancer (HDGC) is unequivocal. Germline alterations in its encoding gene (CDH1) are causative of HDGC and occur in about 40% of patients. Importantly, while in most cases CDH1 alterations result in the complete loss of E-cadherin associated with a well-established clinical impact, in about 20% of cases the mutations are of the missense type. The latter are of particular concern in terms of genetic counselling and clinical management, as the effect of the sequence variants in E-cadherin function is not predictable. If a deleterious variant is identified, prophylactic surgery could be recommended. Therefore, over the last few years, intensive research has focused on evaluating the functional consequences of CDH1 missense variants and in assessing E-cadherin pathogenicity. In that context, our group has contributed to better characterize CDH1 germline missense variants and is now considered a worldwide reference centre. In this review, we highlight the state of the art methodologies to categorize CDH1 variants, as neutral or deleterious. This information is subsequently integrated with clinical data for genetic counseling and management of CDH1 variant carriers.
Collapse
|
37
|
Kurihara H, Maruyama R, Ishiguro K, Kanno S, Yamamoto I, Ishigami K, Mitsuhashi K, Igarashi H, Ito M, Tanuma T, Sukawa Y, Okita K, Hasegawa T, Imai K, Yamamoto H, Shinomura Y, Nosho K. The relationship between EZH2 expression and microRNA-31 in colorectal cancer and the role in evolution of the serrated pathway. Oncotarget 2017; 7:12704-17. [PMID: 26871294 PMCID: PMC4914316 DOI: 10.18632/oncotarget.7260] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
Polycomb group protein enhancer of zeste homolog 2 (EZH2) is a methyltransferase that correlates with the regulation of invasion and metastasis and is overexpressed in human cancers such as colorectal cancer. MicroRNA-31 (miR-31) plays an oncogenic role and is associated with BRAF mutation and poor prognosis in colorectal cancer. EZH2 is functionally considered to suppress miR-31 expression in human cancers; however, no study has reported its relationship with colon cancer. We therefore evaluated EZH2 expression using immunohistochemistry and assessed miR-31 and epigenetic alterations using 301 colorectal carcinomas and 207 premalignant lesions. Functional analysis was performed to identify the association between EZH2 and miR-31 using cancer cell lines. In the current study, negative, weak, moderate, and strong EZH2 expressions were observed in 15%, 19%, 25%, and 41% of colorectal cancers, respectively. EZH2 was inversely associated with miR-31 (P < 0.0001), independent of clinicopathological and molecular features. In a multivariate stage-stratified analysis, high EZH2 expression was related to favorable prognosis (P = 0.0022). Regarding premalignant lesions, negative EZH2 expression was frequently detected in sessile serrated adenomas/polyps (SSA/Ps) (76%; P < 0.0001) compared with hyperplastic polyps, traditional serrated adenomas, and non-serrated adenomas (25–36%). Functional analysis demonstrated that the knockdown of EZH2 increased miR-31 expression. In conclusion, an inverse association was identified between EZH2 and miR-31 in colorectal cancers. Our data also showed that upregulation of EZH2 expression may be rare in SSA/Ps. These results suggest that EZH2 suppresses miR-31 in colorectal cancer and may correlate with differentiation and evolution of serrated pathway.
Collapse
Affiliation(s)
- Hiroyoshi Kurihara
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Reo Maruyama
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuya Ishiguro
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Kanno
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Itaru Yamamoto
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keisuke Ishigami
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kei Mitsuhashi
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hisayoshi Igarashi
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Miki Ito
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tokuma Tanuma
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasutaka Sukawa
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenji Okita
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tadashi Hasegawa
- Department of Surgical Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kohzoh Imai
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Yamamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | | | - Katsuhiko Nosho
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
38
|
de Freitas Junior JCM, Morgado-Díaz JA. The role of N-glycans in colorectal cancer progression: potential biomarkers and therapeutic applications. Oncotarget 2017; 7:19395-413. [PMID: 26539643 PMCID: PMC4991391 DOI: 10.18632/oncotarget.6283] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022] Open
Abstract
Changes in glycosylation, which is one of the most common protein post-translational modifications, are considered to be a hallmark of cancer. N-glycans can modulate cell migration, cell-cell adhesion, cell signaling, growth and metastasis. The colorectal cancer (CRC) is a leading cause of cancer-related mortality and the correlation between CRC progression and changes in the pattern of expression of N-glycans is being considered in the search for new biomarkers. Here, we review the role of N-glycans in CRC cell biology. The perspectives on emerging N-glycan-related anticancer therapies, along with new insights and challenges, are also discussed.
Collapse
Affiliation(s)
| | - José Andrés Morgado-Díaz
- Cellular Biology Program, Structural Biology Group, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
39
|
Digital PCR identifies changes in CDH1 (E-cadherin) transcription pattern in intestinal-type gastric cancer. Oncotarget 2017; 8:18811-18820. [PMID: 27861150 PMCID: PMC5386649 DOI: 10.18632/oncotarget.13401] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/09/2016] [Indexed: 12/19/2022] Open
Abstract
E-cadherin is a cell-cell adhesion protein encoded by CDH1 tumor-suppressor gene. CDH1 inactivating mutations, leading to loss of protein expression, are common in gastric cancer of the diffuse histotype, while alternative mechanisms modulating E-cadherin expression characterize the more common intestinal histotype. These mechanisms are still poorly understood. CDH1 intron 2 has recently emerged as a cis-modulator of E-cadherin expression, encoding non-canonical transcripts. One in particular, CDH1a, proved to be expressed in gastric cancer cell lines, while being absent in the normal stomach. For the first time, we evaluated by digital PCR the expression of CDH1 and CDH1a transcripts in cancer and normal tissue samples from 32 patients with intestinal-type gastric cancer. We found a significant decrease in CDH1 expression in tumors compared to normal counterparts (P = 0.001), which was especially evident in 76% of cases. CDH1a was detected at extremely low levels in 47% of tumors, but not in normal mucosa. A trend was observed of having less CDH1 in tumors expressing CDH1atranscript. The majority of tumors with both a decrease in CDH1 and presence of CDH1a also showed a decrease in miR-101 expression levels. On the whole, the decrease of CDH1 transcript, corresponding to the canonical protein, and the presence of CDH1a, corresponding to an alternative isoform, are likely to perturb E-cadherin-mediated signaling and cell-cell adhesion, thus contributing to intestinal-type gastric carcinogenesis.
Collapse
|
40
|
Low plasma levels of miR-101 are associated with tumor progression in gastric cancer. Oncotarget 2017; 8:106538-106550. [PMID: 29290969 PMCID: PMC5739754 DOI: 10.18632/oncotarget.20860] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/15/2017] [Indexed: 12/16/2022] Open
Abstract
Background Several studies have identified the decreased expression of the tumor suppressor miR-101 in various cancers. In this study, we tested miR-101 as a potential therapeutic target and novel plasma biomarker for gastric cancer (GC). Results The miR-101 expression level was significantly lower in GC tissues (P = 0.0038) and GC cell lines (P = 0.0238) than in normal gastric mucosa. Both exosomal and plasma miR-101 were significantly downregulated in GC patients compared with healthy volunteers (P = 0.0281 and P < 0.0001, respectively). Low miR-101 plasma level was significantly associated with advanced T factor, advanced disease stage, and peritoneal metastasis and predicted poor prognosis in GC patients (P = 0.0368; hazard ratio, 3.079; 95% confidence interval: 1.06–11.08). Overexpression of miR-101 in GC cells induced apoptosis by inhibiting MCL1 and suppressed cell migration and invasion by regulating ZEB1. Conclusions Depletion of the tumor suppressor miRNA-101 in plasma is related to tumor progression and poor outcomes. Low plasma miR-101 may be a biomarker for GC, and its restoration might be a novel anticancer treatment strategy.
Collapse
|
41
|
Guo Y, Qi Y, Guo A, Du C, Zhang R, Chu X. miR-564 is downregulated in gastric carcinoma and targets E2F3. Oncol Lett 2017; 13:4155-4160. [PMID: 28588702 DOI: 10.3892/ol.2017.5964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/27/2017] [Indexed: 12/17/2022] Open
Abstract
Numerous aberrantly expressed microRNAs (miRNAs/miRs) have been identified in gastric cancer (GC); however, only a fraction of these have been functionally investigated and novel deregulated miRNAs in GC remain to be explored. Through examining two public miRNA expression profile datasets, the present study identified aberrantly expressed miRNAs in GC. One of these miRNA, miR-564, was identified to be downregulated in GC, which was validated in tissue samples from patients with GC by reverse transcription-quantitative polymerase chain reaction analysis. Targets of miR-564 were then predicted bioinformatically, including transcription factor E2F3 (E2F3), which was identified to be functionally enriched in several cancer signaling pathways. Furthermore, overexpression of miR-564 decreased the activity of a luciferase reporter carrying the 3'-untranslated region of E2F3, in addition to the mRNA and protein level of E2F3, indicating that miR-564 directly targets E2F3. These data suggest that by targeting E2F3, miR-564 may act as a tumor suppressor gene in gastric carcinogenesis.
Collapse
Affiliation(s)
- Yong Guo
- Department of Pathology, No. 161 Hospital of the People's Liberation Army, Wuhan, Hubei 430010, P.R. China
| | - Yong Qi
- Outpatient Department, The People's Liberation Army Naval University of Engineering, Wuhan, Hubei 430033, P.R. China
| | - Aitao Guo
- Department of Pathology, The General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| | - Chengxiong Du
- Department of General Surgery, No. 161 Hospital of the People's Liberation Army, Wuhan, Hubei 430010, P.R. China
| | - Rong Zhang
- Clinical Laboratory, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| | - Xiaoyong Chu
- Medical Clinic of Military Economy Academy of the People's Liberation Army, Wuhan, Hubei 430035, P.R. China
| |
Collapse
|
42
|
Kong P, Zhu X, Geng Q, Xia L, Sun X, Chen Y, Li W, Zhou Z, Zhan Y, Xu D. The microRNA-423-3p-Bim Axis Promotes Cancer Progression and Activates Oncogenic Autophagy in Gastric Cancer. Mol Ther 2017; 25:1027-1037. [PMID: 28254439 DOI: 10.1016/j.ymthe.2017.01.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/04/2017] [Accepted: 01/14/2017] [Indexed: 12/19/2022] Open
Abstract
Human serum microRNAs (miRNAs) have been shown to serve as disease fingerprints for predicting survival of cancer patients. However, the roles of specific miRNAs involved in gastric cancer (GC) are largely unknown. In this study, miRNA profiling was performed on sera obtained from six patients in good- and poor-survival groups. Expression of miR-423-3p was validated by quantitative RT-PCR in another 67 GC serum samples and paired normal and cancerous gastric tissues. Luciferase reporter assays were used to identify the target gene Bcl-2-interacting mediator of cell death (Bim). As a result, between the good-survival and poor-survival groups, the expression of nine serum miRNAs was altered more than two-fold. Among these, miR-423-3p was significantly increased in the poor-survival group, and its overexpression in GC tissues predicted poor survival in 119 patients with GC. miR-423-3p was found to promote cell proliferation, migration, and invasion in cell lines and animal models. Mechanistically, knockdown of the autophagy-related gene (Atg) 7 rescued the GC-promoting effect of miR-423-3p. In conclusion, miR-423-3p activates oncogenic and Beclin-1-dependent autophagy and promotes GC progression by reducing the expression of Bim. The newly identified miR-423-3p-Bim axis might be a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Pengfei Kong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China; Department of the VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China
| | - Qirong Geng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Hematology Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Liangping Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of the VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Xiaowei Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Yingbo Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Wei Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Zhiwei Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Youqing Zhan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China
| | - Dazhi Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510006, China; Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510006, China.
| |
Collapse
|
43
|
Wang JL, Ling ZQ. Progress in research of gastric signet ring cell carcinoma. Shijie Huaren Xiaohua Zazhi 2017; 25:358-363. [DOI: 10.11569/wcjd.v25.i4.358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric signet ring cell carcinoma is a special type of gastric cancer. In recent years the incidence of gastric cancer has decreased, but the incidence of gastric signet ring cell carcinoma is still rising. Although a large number of studies reported the clinicopathologic features and oncogenesis of gastric signet ring cell carcinoma, the results are inconsistent. This article mainly discusses the clinicopathologic features, prognosis, and molecular characteristics of oncogenesis of gastric signet ring cell carcinoma to provide a basis and strategy for individualized treatment of this malignancy.
Collapse
|
44
|
Zhang JX, Xu Y, Gao Y, Chen C, Zheng ZS, Yun M, Weng HW, Xie D, Ye S. Decreased expression of miR-939 contributes to chemoresistance and metastasis of gastric cancer via dysregulation of SLC34A2 and Raf/MEK/ERK pathway. Mol Cancer 2017; 16:18. [PMID: 28114937 PMCID: PMC5259972 DOI: 10.1186/s12943-017-0586-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 01/09/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The development of chemoresistance and metastasis are the leading causes of death for gastric cancer (GC) patients, however, the molecular mechanisms involved remain unclear. Dysregulation of miRNAs is associated with a variety of disease, including GC. Recently, microarray profiling analysis revealed that miR-939 was dysregulated in human GC samples, but the role of miR-939 in GC has not been intensively investigated. METHODS In the present study, we firstly examined the expression pattern of miR-939 in two independent cohorts of clinical GC samples: one cohort of 112 GC patients with stage I-III disease who underwent surgery followed by adjuvant chemotherapy; and another cohort of 110 GC patients with stage IV disease who received palliative chemotherapy. A series of in vivo and in vitro assays were then performed to investigate the function of miR-939 in GC. RESULTS We detected that reduced expression of miR-939 was associated with chemoresistance and increased risk of tumor recurrence in GC patients. Further function study demonstrated that overexpression of miR-939 suppressed GC cell growth, and enhanced 5-fluorouracil-induced chemosensitivity by compromising cellular growth and inducing apoptosis in vitro and in vivo. Moreover, miR-939 repressed the migration and invasion of GC cells in vitro, and diminished the occurrence of lung metastasis in vivo. We further identified solute carrier family 34 member 2 (SLC34A2) was a novel target of miR-939. Mechanistically, we elucidated that miR-939 exerted its function mainly through inhibiting SLC34A2/Raf/MEK/ERK pathway, which is activated in GC. Multivariate analysis identified miR-939, SLC34A2, and their combination as independent indicators for poor prognosis and tumor recurrence in GC patients. CONCLUSION Our data indicate that miR-939 acts as a tumor suppressor miRNA in GC, and miR-939/SLC34A2 axis represents a novel therapeutic strategy for future GC treatment.
Collapse
Affiliation(s)
- Jia-Xing Zhang
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China ,0000 0001 2360 039Xgrid.12981.33The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Yi Xu
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China
| | - Ying Gao
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China
| | - Cui Chen
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China
| | - Zhou-San Zheng
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China
| | - Miao Yun
- 0000 0001 2360 039Xgrid.12981.33The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Hui-Wen Weng
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China
| | - Dan Xie
- 0000 0001 2360 039Xgrid.12981.33The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, 510060 Guangzhou, People’s Republic of China ,0000 0001 2360 039Xgrid.12981.33Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Sheng Ye
- 0000 0001 2360 039Xgrid.12981.33Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan road II, 510080 Guangzhou, People’s Republic of China
| |
Collapse
|
45
|
Azarnezhad A, Mehdipour P. Cancer Genetics at a Glance: The Comprehensive Insights. CANCER GENETICS AND PSYCHOTHERAPY 2017:79-389. [DOI: 10.1007/978-3-319-64550-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
46
|
microRNAs regulate TAL1 expression in T-cell acute lymphoblastic leukemia. Oncotarget 2016; 7:8268-81. [PMID: 26882564 PMCID: PMC4884991 DOI: 10.18632/oncotarget.6987] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
The transcription factor TAL1 is a proto-oncogene whose aberrant expression in committed T-cell precursors is associated with the development of T-cell acute lymphoblastic leukemia (T-ALL). The mechanisms leading to aberrant activation of TAL1 in T-ALL patients who lack chromosomal rearrangements involving the TAL1 locus remain largely unknown. We hypothesized that TAL1 levels decrease during normal T-cell development at least in part due to miRNA-dependent silencing, in which case TAL1 over-expression in some T-ALL cases could be the consequence of deregulated miRNA expression. By performing computational prediction of miRNAs that bind to the human TAL1 mRNA we compiled a list of miRNAs that are candidates to regulate TAL1. Using a luciferase reporter system and mutagenesis assays we confirmed the miRNA-TAL1 mRNA interactions and selected candidate miRNAs: miR-101, miR-520d-5p, miR-140-5p, miR-448 and miR-485-5p. Over-expression of these microRNAs in different T-ALL cell lines consistently resulted in the down-regulation of TAL1 protein. In accordance, inhibition of miR-101 and miR-520d-5p promoted TAL1 protein expression. Importantly, we found that miR-101, miR-140-5p, miR-448 and miR-485-5p were down-regulated in T-ALL patient specimens and T-ALL cell lines. Our results show for the first time the existence of epigenetic regulation of TAL1 by specific miRNAs which may contribute, at least in part, to the ectopic expression of TAL1 in some T-ALL cases.
Collapse
|
47
|
Chen DL, Ju HQ, Lu YX, Chen LZ, Zeng ZL, Zhang DS, Luo HY, Wang F, Qiu MZ, Wang DS, Xu DZ, Zhou ZW, Pelicano H, Huang P, Xie D, Wang FH, Li YH, Xu RH. Long non-coding RNA XIST regulates gastric cancer progression by acting as a molecular sponge of miR-101 to modulate EZH2 expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:142. [PMID: 27620004 PMCID: PMC5020507 DOI: 10.1186/s13046-016-0420-1] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/07/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have emerged as critical regulators of tumor progression. However, the role and molecular mechanism of lncRNA XIST in gastric cancer is still unknown. METHODS Real-time PCR analysis was performed to measure the expression levels of lncRNA XIST in gastric cancer tissues and cell lines, the correlation between lncRNA XIST expression and clinicopathological characteristics and prognosis was analyzed in gastric cancer patients. The biological function of lncRNA XIST on gastric cancer cells were determined both in vitro and in vivo. The regulating relationship between lncRNA XIST and miR-101 was investigated in gastric cancer cells. RESULTS lncRNA XIST was significantly up-regulated in gastric cancer tissues and cell lines. Overexpression of lncRNA XIST was markedly associated with larger tumor size, lymph node invasion, distant metastasis and TNM stage in gastric cancer patients. Functionally, knockdown of lncRNA XIST exerted tumor-suppressive effects by inhibiting cell proliferation, migration and invasion in vitro and tumor growth and metastasis in vivo. Furthermore, an inverse relationship between lncRNA XIST and miR-101 was found. Polycomb group protein enhancer of zeste homolog 2 (EZH2), a direct target of miR-101, could mediated the biological effects that lncRNA XIST exerted. CONCLUSIONS lncRNA XIST is up-regulated and is associated with aggressive tumor phenotypes and patient survival in gastric cancer, and the newly identified lncRNA XIST/miR-101/EZH2 axis could be a potential biomarkers or therapeutic targets for gastric cancer patients.
Collapse
Affiliation(s)
- Dong-Liang Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Yun-Xin Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Le-Zong Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Zhao-Lei Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Dong-Sheng Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Hui-Yan Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Feng Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Miao-Zhen Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - De-Shen Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Da-Zhi Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Zhi-Wei Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Helene Pelicano
- University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Peng Huang
- University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Feng-Hua Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Yu-Hong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Medical Oncology, Sun Yat-sen University Cancer Center, No. 651 Dong Feng East Road, Guangzhou, 510060, China.
| |
Collapse
|
48
|
Zhang X, Peng Y, Jin Z, Huang W, Cheng Y, Liu Y, Feng X, Yang M, Huang Y, Zhao Z, Wang L, Wei Y, Fan X, Zheng D, Meltzer SJ. Integrated miRNA profiling and bioinformatics analyses reveal potential causative miRNAs in gastric adenocarcinoma. Oncotarget 2016; 6:32878-89. [PMID: 26460735 PMCID: PMC4741736 DOI: 10.18632/oncotarget.5419] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/25/2015] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related deaths throughout China and worldwide. The discovery of microRNAs (miRNAs) has provided a new opportunity for developing diagnostic biomarkers and effective therapeutic targets in GC. By performing microarray analyses of benign and malignant gastric epithelial cell lines (HFE145, NCI-N87, MKN28, RF1, KATO III and RF48), 16 significantly dysregulated miRNAs were found. 11 of these were validated by real-time qRT-PCR. Based on miRWalk online database scans, 703 potential mRNA targets of the 16 miRNAs were identified. Bioinformatic analyses suggested that these dysregulated miRNAs and their predicted targets were principally involved in tumor pathogenesis, MAPK signaling, and apoptosis. Finally, miRNA-gene network analyses identified miRNA-125b as a crucial miRNA in GC development. Taken together, these results develop a comprehensive expression and functional profile of differentially expressed miRNAs related to gastric oncogenesis. This profile may serve as a potential tool for biomarker and therapeutic target identification in GC patients.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Shenzhen Key Laboratory of Translational Medicine of Tumor, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Yin Peng
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Department of Pathology, Wuhan University School of Basic Medical Sciences, Hubei, People's Republic of China
| | - Zhe Jin
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Shenzhen Key Laboratory of Micromolecule Innovatal Drugs, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Shenzhen Key Laboratory of Translational Medicine of Tumor, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, People's Republic of China
| | - Weiling Huang
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Yulan Cheng
- Department of Medicine/GI Division, Johns Hopkins University and Sidney Kimmel Cancer Center, Baltimore, MD, USA
| | - Yudan Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xianling Feng
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Mengting Yang
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Yong Huang
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Zhenfu Zhao
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Liang Wang
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Shenzhen Key Laboratory of Translational Medicine of Tumor, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Yanjie Wei
- Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong, People's Republic of China
| | - Xinmin Fan
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Duo Zheng
- Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China.,Shenzhen Key Laboratory of Translational Medicine of Tumor, The Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Stephen J Meltzer
- Department of Medicine/GI Division, Johns Hopkins University and Sidney Kimmel Cancer Center, Baltimore, MD, USA
| |
Collapse
|
49
|
Zhou P, Jiang N, Zhang GX, Sun Q. MiR-203 inhibits tumor invasion and metastasis in gastric cancer by ATM. Acta Biochim Biophys Sin (Shanghai) 2016; 48:696-703. [PMID: 27542403 DOI: 10.1093/abbs/gmw063] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/25/2016] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer is one of the most common malignancies in the world. A number of miRNAs are aberrantly expressed during the progression of gastric cancer. In this study, we aimed to investigate the role of miR-203 in the invasion and metastasis of gastric cancer and the potential mechanism of the effect of miR-203 on the tumor progression of gastric cancer. Our results showed that miR-203 was significantly downregulated in gastric cancer tissues and cells, while ataxia telangiectasia mutated kinase (ATM) was upregulated in gastric cancer tissues and cells and was directly regulated by miR-203. Ectopic overexpression of miR-203 inhibited the colony formation, migration, and invasion of gastric cancer cells. In addition, miR-203 overexpression significantly suppressed the protein level of Snail and obviously promoted the protein level of E-cadherin in gastric cancer cells. ATM knockdown phenocopied the effect of miR-203 overexpression. These results suggested that miR-203 suppressed the migration and invasion of gastric cancer through regulating the level of ATM-mediated-Snail and E-cadherin. MiR-203 might be a novel therapeutic strategy for the treatment of gastric cancer.
Collapse
|
50
|
Beauchemin M, Smith A, Yin VP. Dynamic microRNA-101a and Fosab expression controls zebrafish heart regeneration. Development 2016; 142:4026-37. [PMID: 26628091 DOI: 10.1242/dev.126649] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in the Western world owing to the limited regenerative capacity of the mammalian cardiovascular system. In lieu of new muscle synthesis, the human heart replaces necrotic tissue with deposition of a noncontractile scar. By contrast, the adult zebrafish is endowed with a remarkable regenerative capacity, capable of de novo cardiomyocyte (CM) creation and scar tissue removal when challenged with an acute injury. In these studies, we examined the contributions of the dynamically regulated microRNA miR-101a during adult zebrafish heart regeneration. We demonstrate that miR-101a expression is rapidly depleted within 3 days post-amputation (dpa) but is highly upregulated by 7-14 dpa, before returning to uninjured levels at the completion of the regenerative process. Employing heat-inducible transgenic strains and antisense oligonucleotides, we demonstrate that decreases in miR-101a levels at the onset of cardiac injury enhanced CM proliferation. Interestingly, prolonged suppression of miR-101a activity stimulates new muscle synthesis but with defects in scar tissue clearance. Upregulation of miR-101a expression between 7 and 14 dpa is essential to stimulate removal of the scar. Through a series of studies, we identified the proto-oncogene fosab (cfos) as a potent miR-101a target gene, stimulator of CM proliferation, and inhibitor of scar tissue removal. Importantly, combinatorial depletion of fosab and miR-101a activity rescued defects in scar tissue clearance mediated by miR-101a inhibition alone. In summation, our studies indicate that the precise temporal modulation of the miR-101a/fosab genetic axis is crucial for coordinating CM proliferation and scar tissue removal during zebrafish heart regeneration.
Collapse
Affiliation(s)
- Megan Beauchemin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, USA Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Ashley Smith
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, USA
| | - Viravuth P Yin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, USA Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| |
Collapse
|