1
|
Xu H, Li W, Xue K, Zhang H, Li H, Yu H, Hu L, Gu Y, Li H, Sun X, Liu Q, Wang D. ADAR1-regulated miR-142-3p/RIG-I axis suppresses antitumor immunity in nasopharyngeal carcinoma. Noncoding RNA Res 2025; 10:116-129. [PMID: 39351449 PMCID: PMC11439846 DOI: 10.1016/j.ncrna.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
Following the initial treatment of nasopharyngeal carcinoma (NPC), tumor progression often portends an adverse prognosis for these patients. MicroRNAs (miRNAs) have emerged as critical regulators of tumor immunity, yet their intricate mechanisms in NPC remain elusive. Through comprehensive miRNA sequencing, tumor tissue microarrays and tissue samples analysis, we identified miR-142-3p as a significantly upregulated miRNA that is strongly associated with poor prognosis in recurrent NPC patients. To elucidate the underlying molecular mechanism, we employed RNA sequencing, coupled with cellular and tissue assays, to identify the downstream targets and associated signaling pathways of miR-142-3p. Our findings revealed two potential targets, CFL2 and WASL, which are directly targeted by miR-142-3p. Functionally, overexpressing CFL2 or WASL significantly reversed the malignant phenotypes induced by miR-142-3p both in vitro and in vivo. Furthermore, signaling pathway analysis revealed that miR-142-3p repressed the RIG-I-mediated immune defense response in NPC by inhibiting the nuclear translocation of IRF3, IRF7 and p65. Moreover, we discovered that ADAR1 physically interacted with Dicer and promoted the formation of mature miR-142-3p in a dose-dependent manner. Collectively, ADAR1-mediated miR-142-3p processing promotes tumor progression and suppresses antitumor immunity, indicating that miR-142-3p may serve as a promising prognostic biomarker and therapeutic target for NPC patients.
Collapse
Affiliation(s)
- Haoyuan Xu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Wanpeng Li
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Kai Xue
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Huankang Zhang
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Han Li
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Haoran Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Li Hu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Yurong Gu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Houyong Li
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Xicai Sun
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Quan Liu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| | - Dehui Wang
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, China
| |
Collapse
|
2
|
Kim M, Kubelick KP, Vanderlaan D, Qin D, Lee J, Jhunjhunwala A, Cadena M, Nikolai RJ, Kim J, Emelianov SY. Coupling Gold Nanospheres into Nanochain Constructs for High-Contrast, Longitudinal Photoacoustic Imaging. NANO LETTERS 2024; 24:7202-7210. [PMID: 38747634 PMCID: PMC11194844 DOI: 10.1021/acs.nanolett.4c00992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 06/14/2024]
Abstract
Structural parameters play a crucial role in determining the electromagnetic and thermal responses of gold nanoconstructs (GNCs) at near-infrared (NIR) wavelengths. Therefore, developing GNCs for reliable, high-contrast photoacoustic imaging has been focused on adjusting structural parameters to achieve robust NIR light absorption with photostability. In this study, we introduce an efficient photoacoustic imaging contrast agent: gold sphere chains (GSCs) consisting of plasmonically coupled gold nanospheres. The chain geometry results in enhanced photoacoustic signal generation originating from outstanding photothermal characteristics compared to traditional gold contrast agents, such as gold nanorods. Furthermore, the GSCs produce consistent photoacoustic signals at laser fluences within the limits set by the American National Standards Institute. The exceptional photoacoustic response of GSCs allows for high-contrast photoacoustic imaging over multiple imaging sessions. Finally, we demonstrate the utility of our GSCs for molecular photoacoustic cancer imaging, both in vitro and in vivo, through the integration of a tumor-targeting moiety.
Collapse
Affiliation(s)
- Myeongsoo Kim
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- Petit
Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Kelsey P. Kubelick
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Don Vanderlaan
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - David Qin
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Jeungyoon Lee
- School
of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Anamik Jhunjhunwala
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Melissa Cadena
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Robert J. Nikolai
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Jinhwan Kim
- Department
of Biomedical Engineering, University of
California Davis, Davis, California 95616, United States
- Department
of Surgery, School of Medicine, University
of California Davis, Sacramento, California 95817, United States
| | - Stanislav Y. Emelianov
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- Petit
Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- School
of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
3
|
Gómez-Acebo I, Llorca J, Alonso-Molero J, Díaz-Martínez M, Pérez-Gómez B, Amiano P, Belmonte T, Molina AJ, Burgui R, Castaño-Vinyals G, Moreno V, Molina-Barceló A, Marcos-Gragera R, Kogevinas M, Pollán M, Dierssen-Sotos T. Circulating miRNAs signature on breast cancer: the MCC-Spain project. Eur J Med Res 2023; 28:480. [PMID: 37925534 PMCID: PMC10625260 DOI: 10.1186/s40001-023-01471-2] [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: 06/19/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
PURPOSE To build models combining circulating microRNAs (miRNAs) able to identify women with breast cancer as well as different types of breast cancer, when comparing with controls without breast cancer. METHOD miRNAs analysis was performed in two phases: screening phase, with a total n = 40 (10 controls and 30 BC cases) analyzed by Next Generation Sequencing, and validation phase, which included 131 controls and 269 cases. For this second phase, the miRNAs were selected combining the screening phase results and a revision of the literature. They were quantified using RT-PCR. Models were built using logistic regression with LASSO penalization. RESULTS The model for all cases included seven miRNAs (miR-423-3p, miR-139-5p, miR-324-5p, miR-1299, miR-101-3p, miR-186-5p and miR-29a-3p); which had an area under the ROC curve of 0.73. The model for cases diagnosed via screening only took in one miRNA (miR-101-3p); the area under the ROC curve was 0.63. The model for disease-free cases in the follow-up had five miRNAs (miR-101-3p, miR-186-5p, miR-423-3p, miR-142-3p and miR-1299) and the area under the ROC curve was 0.73. Finally, the model for cases with active disease in the follow-up contained six miRNAs (miR-101-3p, miR-423-3p, miR-139-5p, miR-1307-3p, miR-331-3p and miR-21-3p) and its area under the ROC curve was 0.82. CONCLUSION We present four models involving eleven miRNAs to differentiate healthy controls from different types of BC cases. Our models scarcely overlap with those previously reported.
Collapse
Affiliation(s)
- Inés Gómez-Acebo
- Department of Preventive Medicine and Public Health, University of Cantabria, Santander, Spain.
- IDIVAL, Santander, Spain.
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain.
| | - Javier Llorca
- Department of Preventive Medicine and Public Health, University of Cantabria, Santander, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
| | - Jessica Alonso-Molero
- Department of Preventive Medicine and Public Health, University of Cantabria, Santander, Spain
- IDIVAL, Santander, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
| | - Marta Díaz-Martínez
- Department of Preventive Medicine and Public Health, University of Cantabria, Santander, Spain
| | - Beatriz Pérez-Gómez
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- National Centre for Epidemiology, Carlos III Institute of Health, Madrid, Spain
| | - Pilar Amiano
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- Sub Directorate for Public Health and Addictions of Gipuzkoa, Ministry of Health of the Basque Government, San Sebastian, Spain
- Epidemiology of Chronic and Communicable Diseases Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Thalía Belmonte
- IUOPA, University of Oviedo and ISPA (Health Research Institute of Asturias), Oviedo, Spain
| | - Antonio J Molina
- Grupo de Investigación en Interacción, Gen-Ambiente-Salud (GIIGAS), Instituto de Biomedicina (IBIOMED), Universidad de León, León, Spain
| | - Rosana Burgui
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- Institute of Public and Occupational Health of Navarre (ISPLN), 31003, Pamplona, Spain
| | - Gemma Castaño-Vinyals
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Víctor Moreno
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- Colorectal Cancer Group, ONCOBELL Program, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Oncology Data Analytics Program, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, Faculty of Medicine and health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona, Barcelona, Spain
| | - Ana Molina-Barceló
- Cancer and Public Health UnitFoundation for the Promotion of Health and Biomedical Research (FISABIO-Salud Pública) in the Valencia Region, Valencia, Spain
| | - Rafael Marcos-Gragera
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- Epidemiology Unit and Girona Cancer Registry, Oncology Coordination Plan, Department of Health, Autonomous Government of Catalonia, Catalan Institute of Oncology (ICO), Girona Biomedical Research Institute (IdiBGi), Girona, Spain
| | - Manolis Kogevinas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Marina Pollán
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
- National Centre for Epidemiology, Carlos III Institute of Health, Madrid, Spain
| | - Trinidad Dierssen-Sotos
- Department of Preventive Medicine and Public Health, University of Cantabria, Santander, Spain
- IDIVAL, Santander, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Jiang Y, Liu Y, Zhang Y, Ouyang J, Feng Y, Li S, Wang J, Zhang C, Tan L, Zhong J, Zou L. MicroRNA-142-3P suppresses the progression of papillary thyroid carcinoma by targeting FN1 and inactivating FAK/ERK/PI3K signaling. Cell Signal 2023:110792. [PMID: 37406787 DOI: 10.1016/j.cellsig.2023.110792] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/25/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
OBJECTIVES miR-142-3P is a tumor suppressor in various malignant cancers. However, the function of miR-142-3P in papillary thyroid carcinoma (PTC) remains to be elucidated. The aim of this study was to explore the function and mechanism of miR-142-3P in PTC. METHODS Real Time Quantitative PCR (RT-qPCR) was used to assess the expression of miR-142-3P and Fibronectin 1 (FN1) in PTC. The correlation between FN1 and miR-142-3P expression was analyzed by Spearman's correlation analysis. Cell Counting Kit 8 (CCK8), 5-ethynyl-2'-deoxyuridine (EDU) assay, cell migration and invasion assay and wound healing measures evaluated the effect of miR-142-3P and FN1 on cell proliferation, migration and invasion. Dural Luciferase reported gene assay evaluated the interaction between miR-142-3P and 3' untranslated region (UTR) of FN1. The Epithelial-Mesenchymal-Transition (EMT) and apoptosis related marker genes were measured using western blot analysis (WB). RESULTS miR-142-3P was significantly decreased in both PTC specimens and relevant cell lines. Functionally, miR-142-3P inhibited cell proliferation, migration, invasion and EMT, and induced the cell apoptosis in PTC. In addition, miR-142-3P bound directly with 3' UTR of FN1 and negatively regulated the expression of FN1 in PTC. FN1 expression is elevated in PTC, and its aberrant high correlated with declines in recurrence-free survival (RFS). Moreover, FN1 promoted cell proliferation, migration, invasion and EMT, induced cell apoptosis in PTC cells. Depletion of FN1 rescues the effect of miR-142-3P inhibitor on cell proliferation, invasion, apoptosis and EMT via inactivating Focal Adhesion Kinase (FAK)/Extracellular Signal-Regulated Kinase (ERK) / Phosphoinostide 3-kinase (P13K) signaling. CONCLUSION miR-142-3P suppressed cell proliferation, migration, invasion and EMT through modulating FN1/FAK/ERK/PI3K signaling in PTC, suggesting it as a potential therapeutic target for PTC.
Collapse
Affiliation(s)
- Yufei Jiang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China; Aculty of Healty Science, University of Macau, Macau 999078, People's Republic of China
| | - Yarong Liu
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Yiyuan Zhang
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Jielin Ouyang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Yang Feng
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Shumei Li
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Jingjing Wang
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Chaojie Zhang
- Department of Papillary Thyroid Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Lihong Tan
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China.
| | - Jie Zhong
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China.
| | - Lianhong Zou
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China.
| |
Collapse
|
5
|
Zhang Y, Ma S, Zhang J, Lou L, Liu W, Gao C, Miao L, Sun F, Chen W, Cao X, Wei J. MicroRNA-142-3p promotes renal cell carcinoma progression by targeting RhoBTB3 to regulate HIF-1 signaling and GGT/GSH pathways. Sci Rep 2023; 13:5935. [PMID: 37045834 PMCID: PMC10097650 DOI: 10.1038/s41598-022-21447-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/27/2022] [Indexed: 04/14/2023] Open
Abstract
MicroRNAs play a critical regulatory role in different cancers, but their functions in renal cell carcinoma (RCC) have not been elucidated. Reportedly, miR-142-3p is involved in the tumorigenesis and the development of RCC in vitro and is clinically correlated with the poor prognosis of RCC patients. However, the molecular target of miR-142-3p and the underlying mechanism are unclear. In this study, we found that miR-142-3p was upregulated in RCC tumor tissues and downregulated in exosomes compared to normal tissues. The expression of miR-142-3p was inversely associated with the survival of patients with kidney renal clear cell carcinoma (KIRC). RhoBTB3 was reduced in RCC, and miR-142-3p plays an inverse function with RhoBTB3 in KIRC. The direct interaction between RhoBTB3 and miR-142-3p was demonstrated by a dual luciferase reporter assay. miR-142-3p promoted metastasis in the xenograft model, and the suppression of miR-142-3p upregulated RhoBTB3 protein expression and inhibited the mRNAs and proteins of HIF1A, VEGFA, and GGT1. Also, the miR-142-3p overexpression upregulated the mRNA of HIF1A, VEGFA, and GGT1. In conclusion, miR-142-3p functions as an oncogene in RCC, especially in KIRC, by targeting RhoBTB3 to regulate HIF-1 signaling and GGT/GSH pathways, which needs further exploration.
Collapse
Affiliation(s)
- Yijing Zhang
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Sha Ma
- Department of Hematopathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jun Zhang
- Department of Pulmonary and Critical Care Medicine, Yantaishan Hospital, Yantai, China
| | - Lu Lou
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Wanqi Liu
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Chao Gao
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Long Miao
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Fanghao Sun
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Wei Chen
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China
| | - Xiliang Cao
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China.
| | - Jin Wei
- Department of Urology, China University of Mining and Technology, Xuzhou No.1 People's Hospital, Xuzhou, China.
| |
Collapse
|
6
|
MicroRNAs: A Link between Mammary Gland Development and Breast Cancer. Int J Mol Sci 2022; 23:ijms232415978. [PMID: 36555616 PMCID: PMC9786715 DOI: 10.3390/ijms232415978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is among the most common cancers in women, second to skin cancer. Mammary gland development can influence breast cancer development in later life. Processes such as proliferation, invasion, and migration during mammary gland development can often mirror processes found in breast cancer. MicroRNAs (miRNAs), small, non-coding RNAs, can repress post-transcriptional RNA expression and can regulate up to 80% of all genes. Expression of miRNAs play a key role in mammary gland development, and aberrant expression can initiate or promote breast cancer. Here, we review the role of miRNAs in mammary development and breast cancer, and potential parallel roles. A total of 32 miRNAs were found to be expressed in both mammary gland development and breast cancer. These miRNAs are involved in proliferation, metastasis, invasion, and apoptosis in both processes. Some miRNAs were found to have contradictory roles, possibly due to their ability to target many genes at once. Investigation of miRNAs and their role in mammary gland development may inform about their role in breast cancer. In particular, by studying miRNA in development, mechanisms and potential targets for breast cancer treatment may be elucidated.
Collapse
|
7
|
Tan Z, Zhang Z, Yu K, Yang H, Liang H, Lu T, Ji Y, Chen J, He W, Chen Z, Mei Y, Shen XL. Integrin subunit alpha V is a potent prognostic biomarker associated with immune infiltration in lower-grade glioma. Front Neurol 2022; 13:964590. [PMID: 36388191 PMCID: PMC9642104 DOI: 10.3389/fneur.2022.964590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/15/2022] [Indexed: 09/30/2023] Open
Abstract
As a member of integrin receptor family, ITGAV (integrin subunit α V) is involved in a variety of cell biological processes and overexpressed in various cancers, which may be a potential prognostic factor. However, its prognostic value and potential function in lower-grade glioma (LGG) are still unclear, and in terms of immune infiltration, it has not been fully elucidated. Here, the expression preference, prognostic value, and clinical traits of ITGAV were investigated using The Cancer Genome Atlas database (n = 528) and the Chinese Glioma Genome Atlas dataset (n = 458). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses and gene set enrichment analysis (GSEA) were used to explore the biological function of ITGAV. Using R package "ssGSEA" analysis, it was found thatthe ITGAV mRNA expression level showed intense correlation with tumor immunity, such as tumor-infiltrating immune cells and multiple immune-related genes. In addition, ITGAV is associated with some immune checkpoints and immune checkpoint blockade (ICB) and response to chemotherapy. and the expression of ITGAV protein in LGG patients was verified via immunohistochemistry (IHC). ITGAV expression was higher in LGG tissues than in normal tissues (P < 0.001) and multifactor analysis showed that ITGAV mRNA expression was an independent prognostic factor for LGG overall survival (OS; hazard ratio = 2.113, 95% confidence interval = 1.393-3.204, P < 0.001). GSEA showed that ITGAV expression was correlated with Inflammatory response, complement response, KRAS signal, and interferon response. ssGSEA results showed a positive correlation between ITGAV expression and Th2 cell infiltration level. ITGAV mRNA was overexpressed in LGG, and high ITGAV mRNA levels were found to be associated with poor protein expression and poor OS. ITGAV is therefore a potential biomarker for the diagnosis and prognosis of LGG and may be a potential immunotherapy target.
Collapse
Affiliation(s)
- Zilong Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
- The Graduate Department, Jiangxi Medical College of Nanchang University Nanchang, Nanchang, China
| | - Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
- The Graduate Department, Jiangxi Medical College of Nanchang University Nanchang, Nanchang, China
| | - Kai Yu
- Department of Neurosurgery, People's Hospital of Wuhan University, Wuhan, China
| | - Huan Yang
- Department of Neurosurgery, Changde Hospital of Traditional Chinese Medicine, Changde, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianzhu Lu
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Yulong Ji
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Junjun Chen
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Wei He
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuran Mei
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-Li Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Bloomfield J, Sabbah M, Castela M, Mehats C, Uzan C, Canlorbe G. Clinical Value and Molecular Function of Circulating MicroRNAs in Endometrial Cancer Regulation: A Systematic Review. Cells 2022; 11:cells11111836. [PMID: 35681531 PMCID: PMC9180151 DOI: 10.3390/cells11111836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
This systematic review of literature highlights the different microRNAs circulating in the serum or plasma of endometrial cancer patients and their association with clinical and prognostic characteristics in endometrial cancer. This study also investigates the molecular functions of these circulating microRNAs. According to this systematic review, a total of 33 individual circulating miRs (-9, -15b, -20b-5p, -21, -27a, -29b, -30a-5p, -92a, -99a, -100, -135b, -141, -142-3p, -143-3p, -146a-5p, -150-5p, -151a-5p, -186, -195-5p, -199b, -200a, -203, -204, -205, -222, -223, -301b, -423-3p, -449, -484, -887-5p, -1228, and -1290) and 6 different panels of miRs (“miR-222/miR-223/miR-186/miR-204”, “miR-142-3p/miR-146a-5p/miR-151a-5p”, “miR-143-3p/miR-195-5p/miR-20b-5p/miR-204-5p/miR-423-3p/miR-484”, “mir-9/miR-1229”, “miR-9/miR-92a”, and “miR-99a/miR-199b”) had a significant expression variation in EC patients compared to healthy patients. Also, seven individual circulating miRs (-9, -21, -27a, -29b, -99a, -142-3p, and -449a) had a significant expression variation according to EC prognostic factors such as the histological type and grade, tumor size, FIGO stage, lymph node involvement, and survival rates. One panel of circulating miRs (“-200b/-200c/-203/-449a”) had a significant expression variation according to EC myometrial invasion. Further studies are needed to better understand their function and circulation.
Collapse
Affiliation(s)
- Joy Bloomfield
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Sorbonne University, INSERM UMR_S_938, 75020 Paris, France; (J.B.); (M.S.); (C.U.)
- Assistance Publique des Hôpitaux de Paris (AP-HP), Department of Gynecological and Breast Surgery and Oncology, Pitié-Salpêtrière University Hospital, 75013 Paris, France
| | - Michèle Sabbah
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Sorbonne University, INSERM UMR_S_938, 75020 Paris, France; (J.B.); (M.S.); (C.U.)
- Centre National de la Recherche Scientifique (CNRS), 75012 Paris, France
| | - Mathieu Castela
- Scarcell Therapeutics, 101 Rue de Sèvres, 75006 Paris, France;
| | - Céline Mehats
- U1016, CNRS, UMR8104, Institut Cochin, INSERM, Université de Paris, 75014 Paris, France;
| | - Catherine Uzan
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Sorbonne University, INSERM UMR_S_938, 75020 Paris, France; (J.B.); (M.S.); (C.U.)
- Assistance Publique des Hôpitaux de Paris (AP-HP), Department of Gynecological and Breast Surgery and Oncology, Pitié-Salpêtrière University Hospital, 75013 Paris, France
- Institut Universitaire de Cancérologie (IUC), 75020 Paris, France
| | - Geoffroy Canlorbe
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Sorbonne University, INSERM UMR_S_938, 75020 Paris, France; (J.B.); (M.S.); (C.U.)
- Assistance Publique des Hôpitaux de Paris (AP-HP), Department of Gynecological and Breast Surgery and Oncology, Pitié-Salpêtrière University Hospital, 75013 Paris, France
- Institut Universitaire de Cancérologie (IUC), 75020 Paris, France
- Correspondence:
| |
Collapse
|
9
|
Mazzolini J, Le Clerc S, Morisse G, Coulonges C, Zagury J, Sieger D. Wasl is crucial to maintain microglial core activities during glioblastoma initiation stages. Glia 2022; 70:1027-1051. [PMID: 35194846 PMCID: PMC9306864 DOI: 10.1002/glia.24154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
Abstract
Microglia actively promotes the growth of high-grade gliomas. Within the glioma microenvironment an amoeboid microglial morphology has been observed, however the underlying causes and the related impact on microglia functions and their tumor promoting activities is unclear. Using the advantages of the larval zebrafish model, we identified the underlying mechanism and show that microglial morphology and functions are already impaired during glioma initiation stages. The presence of pre-neoplastic HRasV12 expressing cells induces an amoeboid morphology of microglia, increases microglial numbers and decreases their motility and phagocytic activity. RNA sequencing analysis revealed lower expression levels of the actin nucleation promoting factor wasla in microglia. Importantly, a microglia specific rescue of wasla expression restores microglial morphology and functions. This results in increased phagocytosis of pre-neoplastic cells and slows down tumor progression. In conclusion, we identified a mechanism that de-activates core microglial functions within the emerging glioma microenvironment. Restoration of this mechanism might provide a way to impair glioma growth.
Collapse
Affiliation(s)
- Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Sigrid Le Clerc
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Gregoire Morisse
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Cédric Coulonges
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Jean‐François Zagury
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
10
|
Kahl I, Mense J, Finke C, Boller AL, Lorber C, Győrffy B, Greve B, Götte M, Espinoza-Sánchez NA. The cell cycle-related genes RHAMM, AURKA, TPX2, PLK1, and PLK4 are associated with the poor prognosis of breast cancer patients. J Cell Biochem 2022; 123:581-600. [PMID: 35014077 DOI: 10.1002/jcb.30205] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 01/02/2023]
Abstract
Breast cancer is the third most common type of cancer diagnosed. Cell cycle is a complex but highly organized and controlled process, in which normal cells sense mitogenic growth signals that instruct them to enter and progress through their cell cycle. This process culminates in cell division generating two daughter cells with identical amounts of genetic material. Uncontrolled proliferation is one of the hallmarks of cancer. In this study, we analyzed the expression of the cell cycle-related genes receptor for hyaluronan (HA)-mediated motility (RHAMM), AURKA, TPX2, PLK1, and PLK4 and correlated them with the prognosis in a collective of 3952 breast cancer patients. A high messenger RNA expression of all studied genes correlated with a poor prognosis. Stratifying the patients according to the expression of hormonal receptors, we found that in patients with estrogen and progesterone receptor-positive and human epithelial growth factor receptor 2-negative tumors, and Luminal A and Luminal B tumors, the expression of the five analyzed genes correlates with worse survival. qPCR analysis of a panel of breast cancer cell lines representative of major molecular subtypes indicated a predominant expression in the luminal subtype. In vitro experiments showed that radiation influences the expression of the five analyzed genes both in luminal and triple-negative model cell lines. Functional analysis of MDA-MB-231 cells showed that small interfering RNA knockdown of PLK4 and TPX2 and pharmacological inhibition of PLK1 had an impact on the cell cycle and colony formation. Looking for a potential upstream regulation by microRNAs, we observed a differential expression of RHAMM, AURKA, TPX2, PLK1, and PLK4 after transfecting the MDA-MB-231 cells with three different microRNAs. Survival analysis of miR-34c-5p, miR-375, and miR-142-3p showed a different impact on the prognosis of breast cancer patients. Our study suggests that RHAMM, AURKA, TPX2, PLK1, and PLK4 can be used as potential targets for treatment or as a prognostic value in breast cancer patients.
Collapse
Affiliation(s)
- Iris Kahl
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Julian Mense
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Christopher Finke
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Anna-Lena Boller
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Clara Lorber
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary.,Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| |
Collapse
|
11
|
Wang B, Ji D, Xing W, Li F, Huang Z, Zheng W, Xue J, Zhu Y, Yang X. miR-142-3p and HMGB1 Are Negatively Regulated in Proliferation, Apoptosis, Migration, and Autophagy of Cartilage Endplate Cells. Cartilage 2021; 13:592S-603S. [PMID: 33955243 PMCID: PMC8804737 DOI: 10.1177/19476035211012444] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cartilage endplate (CEP) degeneration plays a vital role in the pathological process of intervertebral disc degeneration. It has been previously reported that microRNAs may participate in the occurrence and development of intervertebral disc degeneration through regulating its target genes directly. The regulatory roles of miR-142-3p/HMGB1 in some orthopedic diseases have been determined successively, but there was no report about the degeneration of CEP. Therefore, we aimed to determine the regulation of miR-142-3p/HMGB1 or potential molecular mechanisms on proliferation, apoptosis, migration, and autophagy of CEP cells. METHODS The target gene of miR-142-3p was determined by double luciferase assay. We selected ATDC5 cell lines. CCK-8 method was used to detect cell proliferation. Real-time fluorescence quantitative polymerase chain reaction was used to determine gene expression levels, and western blot analysis was used to determine protein expression levels. We chose flow cytometry to measure cell apoptosis and cell cycle. RESULTS The result of luciferase detection showed that the target gene of miR-142-3p in CEP cells was HMGB1. Knockdown of the miR-142-3p inhibited the expression level of HMGB1, the proliferation and migration of CEP cells, but it promoted apoptosis of CEP cells. In addition, the detection results of the proteins related to apoptosis or autophagy showed that knockdown of miR-142-3p promoted apoptosis and autophagy. CONCLUSION The negative regulation of miR-142-3p/HMGB1 can affect the proliferation, apoptosis, migration, and autophagy of CEP cells. Our results provide a new idea for the targeted treatment of CEP degeneration by inhibiting the expression of HMGB1.
Collapse
Affiliation(s)
- Bo Wang
- School of Graduate, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Demin Ji
- School of Graduate, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wenhua Xing
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Feng Li
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Zhi Huang
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wenkai Zheng
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jianmin Xue
- School of Graduate, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yong Zhu
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xuejun Yang
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
12
|
Molecular Characterization of Membrane Steroid Receptors in Hormone-Sensitive Cancers. Cells 2021; 10:cells10112999. [PMID: 34831222 PMCID: PMC8616056 DOI: 10.3390/cells10112999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer is one of the most common causes of death worldwide, and its development is a result of the complex interaction of genetic factors, environmental cues, and aging. Hormone-sensitive cancers depend on the action of one or more hormones for their development and progression. Sex steroids and corticosteroids can regulate different physiological functions, including metabolism, growth, and proliferation, through their interaction with specific nuclear receptors, that can transcriptionally regulate target genes via their genomic actions. Therefore, interference with hormones’ activities, e.g., deregulation of their production and downstream pathways or the exposition to exogenous hormone-active substances such as endocrine-disrupting chemicals (EDCs), can affect the regulation of their correlated pathways and trigger the neoplastic transformation. Although nuclear receptors account for most hormone-related biologic effects and their slow genomic responses are well-studied, less-known membrane receptors are emerging for their ability to mediate steroid hormones effects through the activation of rapid non-genomic responses also involved in the development of hormone-sensitive cancers. This review aims to collect pre-clinical and clinical data on these extranuclear receptors not only to draw attention to their emerging role in cancer development and progression but also to highlight their dual role as tumor microenvironment players and potential candidate drug targets.
Collapse
|
13
|
Yoon S, Yang H, Ryu HM, Lee E, Jo Y, Seo S, Kim D, Lee CH, Kim W, Jung KH, Park SR, Choi EK, Kim SW, Park KS, Lee DH. Integrin αvβ3 Induces HSP90 Inhibitor Resistance via FAK Activation in KRAS-Mutant Non-Small Cell Lung Cancer. Cancer Res Treat 2021; 54:767-781. [PMID: 34607394 PMCID: PMC9296920 DOI: 10.4143/crt.2021.651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/29/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose HSP90 remains an important cancer target because of its involvement in multiple oncogenic protein pathways and biologic processes. Although many HSP90 inhibitors have been tested in the treatment of KRAS-mutant non-small cell lung cancer (NSCLC), most, including AUY922, have failed due to toxic effects and resistance generation, even though a modest efficacy has been observed for these drugs in clinical trials. In our present study, we investigated the novel mechanism of resistance to AUY922 to explore possible avenues of overcoming and want to provide some insights that may assist with the future development of successful next-generation HSP90 inhibitors. Materials and Methods We established two AUY922-resistant KRAS-mutated NSCLC cells and conducted RNA sequencing to identify novel resistance biomarker. Results We identified novel two resistance biomarkers. We observed that both integrin Av (ITGAv) and β3 (ITGB3) induce AUY922-resistance via focal adhesion kinase (FAK) activation, as well as an epithelial-mesenchymal transition (EMT), in both in vitro and in vivo xenograft model. mRNAs of both ITGAv and ITGB3 were also found to be elevated in a patient who had shown acquired resistance in a clinical trial of AUY922. ITGAv was induced by miR-142 downregulation, and ITGB3 was increased by miR-150 downregulation during the development of AUY922-resistance. Therefore, miR-150 and miR-142 overexpression effectively inhibited ITGAvB3-dependent FAK activation, restoring sensitivity to AUY922. Conclusion The synergistic co-targeting of FAK and HSP90 attenuated the growth of ITGAvB3-induced AUY922-resistant KRAS-mutated NSCLC cells in vitro and in vivo, suggesting that this combination may overcome acquired AUY922-resistance in KRAS-mutant NSCLC.
Collapse
Affiliation(s)
- Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hannah Yang
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Hyun-Min Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Eunjin Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yujin Jo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seyoung Seo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Deokhoon Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chang Hoon Lee
- Bio & Drug Discovery Division, Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Wanlim Kim
- Department of Orthopaedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sook Ryun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Kyung Choi
- Center for Advancing Cancer Therapeutics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-We Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kang-Seo Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Dae Ho Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Circ_0008673 regulates breast cancer malignancy by miR-153-3p/CFL2 axis. Arch Gynecol Obstet 2021; 305:223-232. [PMID: 34324029 DOI: 10.1007/s00404-021-06149-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Breast cancer is an aggressive tumor, which poses a heavy burden to human health. Circular RNAs have been involved in the pathogenesis of breast cancer. This study aims to investigate whether circ_0008673 mediates breast cancer malignant progression by microRNA-153-3p (miR-153-3p)/cofilin 2 (CFL2) pathway. METHODS The RNA levels of circ_0008673, miR-153-3p and CFL2 were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The protein expression of CFL2, E-cadherin and N-cadherin was determined by western blot analysis. Cell proliferation was demonstrated through cell counting kit-8 and cell colony-formation assays. Cell apoptosis was detected by flow cytometry analysis. Cell migratory and invasive capacities were determined by transwell assay. The associated relationship between miR-153-3p and circ_0008673 or CFL2 was predicted by online databases, and testified by dual-luciferase reporter and RNA immunoprecipitation assays. In vivo assay was employed to demonstrate the effects of circ_0008673 silencing on tumor formation in vivo. RESULTS Circ_0008673 and CFL2 expressions were upregulated, while miR-153-3p expression was downregulated in breast cancer tissues and cells compared with adjacent normal breast tissues and cells, respectively. Circ_0008673 overexpression promoted cell proliferation, migration and invasion, and repressed cell apoptosis, while circ_0008673 silencing had opposite effects. Additionally, circ_0008673 served as a sponge of miR-153-3p. And circ_0008673 was proved to regulate breast cancer cell malignancy by sponging miR-153-3p. MiR-153-3p was found to modulate breast cancer cell carcinogenesis via targeting CFL2. Furthermore, circ_0008673 silencing repressed tumor growth in vivo. CONCLUSION Circ_0008673 promoted breast cancer progression by upregulating CFL2 expression through sponging miR-153-3p. This study provides a theoretical basis for researching circRNA-directed treatment of breast cancer.
Collapse
|
15
|
de Sousa E, Lérias JR, Beltran A, Paraschoudi G, Condeço C, Kamiki J, António PA, Figueiredo N, Carvalho C, Castillo-Martin M, Wang Z, Ligeiro D, Rao M, Maeurer M. Targeting Neoepitopes to Treat Solid Malignancies: Immunosurgery. Front Immunol 2021; 12:592031. [PMID: 34335558 PMCID: PMC8320363 DOI: 10.3389/fimmu.2021.592031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 05/07/2021] [Indexed: 12/26/2022] Open
Abstract
Successful outcome of immune checkpoint blockade in patients with solid cancers is in part associated with a high tumor mutational burden (TMB) and the recognition of private neoantigens by T-cells. The quality and quantity of target recognition is determined by the repertoire of ‘neoepitope’-specific T-cell receptors (TCRs) in tumor-infiltrating lymphocytes (TIL), or peripheral T-cells. Interferon gamma (IFN-γ), produced by T-cells and other immune cells, is essential for controlling proliferation of transformed cells, induction of apoptosis and enhancing human leukocyte antigen (HLA) expression, thereby increasing immunogenicity of cancer cells. TCR αβ-dependent therapies should account for tumor heterogeneity and availability of the TCR repertoire capable of reacting to neoepitopes and functional HLA pathways. Immunogenic epitopes in the tumor-stroma may also be targeted to achieve tumor-containment by changing the immune-contexture in the tumor microenvironment (TME). Non protein-coding regions of the tumor-cell genome may also contain many aberrantly expressed, non-mutated tumor-associated antigens (TAAs) capable of eliciting productive anti-tumor immune responses. Whole-exome sequencing (WES) and/or RNA sequencing (RNA-Seq) of cancer tissue, combined with several layers of bioinformatic analysis is commonly used to predict possible neoepitopes present in clinical samples. At the ImmunoSurgery Unit of the Champalimaud Centre for the Unknown (CCU), a pipeline combining several tools is used for predicting private mutations from WES and RNA-Seq data followed by the construction of synthetic peptides tailored for immunological response assessment reflecting the patient’s tumor mutations, guided by MHC typing. Subsequent immunoassays allow the detection of differential IFN-γ production patterns associated with (intra-tumoral) spatiotemporal differences in TIL or peripheral T-cells versus TIL. These bioinformatics tools, in addition to histopathological assessment, immunological readouts from functional bioassays and deep T-cell ‘adaptome’ analyses, are expected to advance discovery and development of next-generation personalized precision medicine strategies to improve clinical outcomes in cancer in the context of i) anti-tumor vaccination strategies, ii) gauging mutation-reactive T-cell responses in biological therapies and iii) expansion of tumor-reactive T-cells for the cellular treatment of patients with cancer.
Collapse
Affiliation(s)
- Eric de Sousa
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana R Lérias
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Antonio Beltran
- Department of Pathology, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Carolina Condeço
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jéssica Kamiki
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Nuno Figueiredo
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | - Carlos Carvalho
- Digestive Unit, Champalimaud Clinical Centre, Lisbon, Portugal
| | | | - Zhe Wang
- Jiangsu Industrial Technology Research Institute (JITRI), Applied Adaptome Immunology Institute, Nanjing, China
| | - Dário Ligeiro
- Lisbon Centre for Blood and Transplantation, Instituto Português do Sangue e Transplantação (IPST), Lisbon, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,I Medical Clinic, Johannes Gutenberg University of Mainz, Mainz, Germany
| |
Collapse
|
16
|
MiR-142 suppresses progression of gastric carcinoma via directly targeting LRP8. Clin Res Hepatol Gastroenterol 2021; 45:101520. [PMID: 33268037 DOI: 10.1016/j.clinre.2020.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/26/2020] [Accepted: 08/04/2020] [Indexed: 02/04/2023]
Abstract
Recently, the role of miRNA-142 (miR-142) in tumor development has attracted extensive attention. The aim of this study was to investigate the impact of miR-142 and its potential target low-density lipoprotein receptor (LDLR)-related protein 8 (LRP8) on the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of gastric carcinoma (GC). Gene and protein expressions were detected using RT-qPCR and Western blotting, respectively. The biological behaviors of GC cell lines were determined by CCK-8, flow cytometry and Transwell assays, respectively. The interaction between miR-142 and LRP8 was confirmed with dual luciferase reporter assay. Xenograft nude mouse model was used to observe tumor growth. Here, miR-142 expression was markedly reduced in GC tissues and cells, and was negatively correlated with lymph node metastasis and poor prognosis in patients with GC. Stable miR-142 overexpression was sufficient to inhibit cell proliferation, migration and invasion in vitro and reduce tumor growth in vivo, accompanied by increased expression of the epithelial marker and reduced levels of mesenchymal markers. Mechanistically, the 3'-untranslated regions (3'-UTR) of LRP8 was a direct target of miR-142. Restoration of LRP8 attenuated the inhibitory effect of miR-142 on GC cells, whereas inhibition of LRP8 caused the opposite outcomes. In conclusion, our findings suggest that miR-142 plays a significant role in suppressing progression of GC by targeting LRP8, and miR-142 may be useful for the development of novel targeted therapies.
Collapse
|
17
|
Hou J, Chen C, Hu Y, Gong Q, Gan L, Xu Y. Identify Function of WASL in Prognosis of Cervical Cancer Based on Omics Data. Front Cell Dev Biol 2021; 9:670890. [PMID: 34222242 PMCID: PMC8248809 DOI: 10.3389/fcell.2021.670890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To clarify the clinical relevance of WASP like actin nucleation promoting factor (WASL) in patients with cervical cancer and associated mechanisms. Methods and Materials We obtained high prediction accuracy and determined the correlation between the expression of WASL and the clinical characteristics of cervical cancer patients. Differentially expressed genes (DEGs) were identified using microarray. Gene ontology (GO) enrichment analysis and gene set enrichment analysis (GSEA) were performed to determine potentially relevant mechanisms related to the prognostication ability of WASL expression. Results Chi-square test and multivariable logistic regression analysis suggested that lower expression of WASL was associated with lower pathological stage (chi-square test: p = 0.022, chi-square = 9.613; logistic regression: OR = 0.869, 95% CI: 0.756-0.991, p = 0.041). Patients in the WASL high expression group have worse overall survival (OS) [hazard ratio (HR): 0.555, 95% CI: 0.348-0.884, log-rank p = 0.012] and recurrence-free survival (RFS) (HR = 0.449, 95% CI: 0.215-0.934, log-rank p = 0.028) compared with those in the WASL low expression group. Univariate and multivariable Cox proportional hazards regression model suggested that WASL expression was an independent prognostic factor for predicting OS and RFS in cervical cancer. DEGs were mostly enriched GO terms related to DNA replication or the proliferation of tumor cells. The results of GSEA suggested samples in the WASL knockdown group were enriched in glycolysis, TNF-α signaling via NFkB, mTORC1 signaling, and Wnt/β-catenin signaling. Conclusions WASL expression was associated with the pathological stage, and it might be an independent prognostication factor in patients with cervical cancer. Knockdown of WASL might be correlated with biological processes such as glycolysis, TNFα signaling, mTOR signaling, and Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Jinxuan Hou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Yingying Hu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Qing Gong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gan
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yu Xu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Li P, Butcher NJ, Minchin RF. Effect arylamine N-acetyltransferase 1 on morphology, adhesion, migration, and invasion of MDA-MB-231 cells: role of matrix metalloproteinases and integrin αV. Cell Adh Migr 2021; 14:1-11. [PMID: 31910058 PMCID: PMC6961680 DOI: 10.1080/19336918.2019.1710015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Reducted arylamine N-acetyltransferase (NAT1) in breast cancers is associated with poor patient survival. NAT1 has also been associated with changes in cancer cell survival and invasion both invitro and invivo. Here, we report the effects of NAT1 in cancer cell invasion by addressing its role in adherence, migration, and invasion in vitro. The NAT1 gene was deleted in MDA-MB-231, HT-29 and HeLa cells using CRISPR/Cas9 gene editing. Loss of NAT1 increased adherence to collagen in all three cell-lines but migration was unaffected. NAT1 deletion decreased invasion and induced changes to cell morphology. These effects were independent of matrix metalloproteinases but were related to integrin ITGαV expression. The data suggest NAT1 is important in adhesion and invasion through integrin expression.
Collapse
Affiliation(s)
- Pengcheng Li
- School of Biomedical Sciences, University of Queensland, St Lucia, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, University of Queensland, St Lucia, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, University of Queensland, St Lucia, Australia
| |
Collapse
|
19
|
Mc Cormack B, Maenhoudt N, Fincke V, Stejskalova A, Greve B, Kiesel L, Meresman GF, Vankelecom H, Götte M, Barañao RI. The ellagic acid metabolites urolithin A and B differentially affect growth, adhesion, motility, and invasion of endometriotic cells in vitro. Hum Reprod 2021; 36:1501-1519. [PMID: 33748857 DOI: 10.1093/humrep/deab053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION What are the effects of plant-derived antioxidant compounds urolithin A (UA) and B (UB) on the growth and pathogenetic properties of an in vitro endometriosis model? SUMMARY ANSWER Both urolithins showed inhibitory effects on cell behavior related to the development of endometriosis by differentially affecting growth, adhesion, motility, and invasion of endometriotic cells in vitro. WHAT IS KNOWN ALREADY Endometriosis is one of the most common benign gynecological diseases in women of reproductive age and is defined by the presence of endometrial tissue outside the uterine cavity. As current pharmacological therapies are associated with side effects interfering with fertility, we aimed at finding alternative therapeutics using natural compounds that can be administered for prolonged periods with a favorable side effects profile. STUDY DESIGN, SIZE, DURATION In vitro cultures of primary endometriotic stromal cells from 6 patients subjected to laparoscopy for benign pathologies with histologically confirmed endometriosis; and immortalized endometrial stromal (St-T1b) and endometriotic epithelial cells (12Z) were utilized to assess the effects of UA and UB on endometriotic cell properties. Results were validated in three-dimensional (3D) in vitro co-culture spheroids of 12Z and primary endometriotic stroma cells of one patient, and organoids from 3 independent donors with endometriosis. PARTICIPANTS/MATERIALS, SETTING, METHODS The effects on cell growth were measured by non-radioactive colorimetric assay to measure cellular metabolic activity as an indicator of cell viability (MTT assay) and flow cytometric cell cycle assay on primary cultures, St-T1b, and 12Z. Apoptosis analyses, the impact on in vitro adhesion, migration, and invasion were evaluated in the cell lines. Moreover, Real-Time Quantitative Reverse Transcription polymerase chain reaction (RT-qPCR) assays were performed on primary cultures, St- T1b and 12Z to evaluate a plausible mechanistic contribution by factors related to proteolysis (matrix metalloproteinase 2, 3 and 9 -MMP2, MMP3, MMP9-, and tissue inhibitor of metalloproteinases -TIMP-1-), cytoskeletal regulators (Ras-related C3 botulinum toxin substrate 1 -RAC1-, Rho-associated coiled-coil containing protein kinase 2 -ROCK2-), and cell adhesion molecules (Syndecan 1 -SDC1-, Integrin alpha V-ITGAV-). Finally, the urolithins effects were evaluated on spheroids and organoids by formation, viability, and drug screen assays. MAIN RESULTS AND THE ROLE OF CHANCE 40 µM UA and 20 µM UB produced a significant decrease in cell proliferation in the primary endometriotic cell cultures (P < 0.001 and P < 0.01, respectively) and in the St-T1b cell line (P < 0.001 and P < 0.05, respectively). In St-T1b, UA exhibited a mean half-maximum inhibitory concentration (IC50) of 39.88 µM, while UB exhibited a mean IC50 of 79.92 µM. Both 40 µM UA and 20 µM UB produced an increase in cells in the S phase of the cell cycle (P < 0.01 and P < 0.05, respectively). The same concentration of UA also increased the percentage of apoptotic ST-t1b cells (P < 0.05), while both urolithins decreased cell migration after 24 h (P < 0.001 both). Only the addition of 5 µM UB decreased the number of St-T1b adherent cells. TIMP-1 expression was upregulated in response to treating the cells with 40 µM UA (P < 0.05). Regarding the 12Z endometriotic cell line, only 40 µM UA decreased proliferation (P < 0.01); while both 40 µM UA and 20 µM UB produced an increase in cells in the G2/M phase (P < 0.05 and P < 0.01, respectively). In this cell line, UA exhibited a mean IC50 of 40.46 µM, while UB exhibited a mean IC50 of 54.79 µM. UB decreased cell migration (P < 0.05), and decreased the number of adherent cells (P < 0.05). Both 40 µM UA and 20 µM UB significantly decreased the cellular invasion of these cells; and several genes were altered when treating the cells with 40 µM UA and 10 µM UB. The expression of MMP2 was downregulated by UA (P < 0.001), and expression of MMP3 (UA P < 0.001 and UB P < 0.05) and MMP9 (P < 0.05, both) were downregulated by both urolithins. Moreover, UA significantly downregulated ROCK2 (P < 0.05), whereas UB treatment was associated with RAC1 downregulation (P < 0.05). Finally, the matrix adhesion receptors and signaling (co)receptors SDC1 and ITGAV were downregulated upon treatment with either UA or UB (P < 0.01 and P < 0.05, respectively in both cases). Regarding the effects of urolithins on 3D models, we have seen that they significantly decrease the viability of endometriosis spheroids (80 µM UA and UB: P < 0.05 both) as well as affecting their area (40 µM UA: P < 0.05, and 80 µM UA: P < 0.01) and integrity (40 µM UA and UB: P < 0.05, 80 µM UA and UB: P < 0.01). On the other hand, UA and UB significantly inhibited organoid development/outgrowth (40 and 80 µM UA: P < 0.0001 both; 40 µM UB: P < ns-0.05-0.001, and 80 µM UB: P < 0.01-0.001-0.001), and all organoid lines show urolithins sensitivity resulting in decreasing viability (UA exhibited a mean IC50 of 33.93 µM, while UB exhibited a mean IC50 of 52.60 µM). LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This study was performed on in vitro endometriosis models. WIDER IMPLICATIONS OF THE FINDINGS These in vitro results provide new insights into the pathogenetic pathways affected by these compounds and mark their use as a potential new therapeutic strategy for the treatment of endometriosis. STUDY FUNDING/COMPETING INTEREST(S) This study was funded EU MSCA-RISE-2015 project MOMENDO (691058). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Barbara Mc Cormack
- Instituto de Biología y Medicina Experimental (IBYME)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - N Maenhoudt
- Laboratory of Tissue Plasticity in Health and Disease, Stem Cell and Developmental Biology Cluster, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - V Fincke
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - A Stejskalova
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - B Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - L Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - G F Meresman
- Instituto de Biología y Medicina Experimental (IBYME)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - H Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Stem Cell and Developmental Biology Cluster, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - M Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - R I Barañao
- Instituto de Biología y Medicina Experimental (IBYME)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
20
|
Qiao EQ, Yang HJ, Zhang XP. Screening of miRNAs associated with lymph node metastasis in Her-2-positive breast cancer and their relationship with prognosis. J Zhejiang Univ Sci B 2021; 21:495-508. [PMID: 32478495 DOI: 10.1631/jzus.b1900584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The aim of this study was to identify some biomarkers for predicting lymph node metastasis and prognosis of human epidermal growth factor receptor 2 (Her-2)-positive breast cancer (BC). We analyzed correlations between microRNAs (miRNAs) and the prognosis of patients with BC based on data collected from The Cancer Genome Atlas (TCGA) database. The expression levels of miR-455, miR-143, and miR-99a were measured in clinical samples of Her-2-positive BC patients with different degrees of lymph node metastasis. We investigated the impacts of overexpressed miR-455 on the proliferation and invasiveness of MDA-MB-453 cells and measured its effects on the expression of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) by quantitative real-time polymerase chain reaction (qRT-PCR). The expression of miR-455 was significantly and positively correlated to the prognosis and overall survival (OS) of the BC (P=0.028), according to TCGA information. The expression level of miR-455 was positively correlated with OS and relapse-free survival (RFS) of patients with Her-2-positive BC, and was negatively correlated with the number of metastatic lymph nodes (P<0.05). Transwell assay suggested that MDA-MB-453 cells became much less invasive (P<0.01) after being transfected with miR-455 mimics. During the qRT-PCR, the expression level of MALAT1 declined significantly after transfection (P<0.01). Overexpressed miR-455 significantly inhibited the proliferation and migration of MDA-MB-453 cells and the expression of MALAT1. We conclude that miR-455 may be a useful potential biomarker for forecasting lymph node metastasis and the prognosis of Her-2-positive BC patients. miR-455 may play an important role in lymph node metastasis of BC by interacting with MALAT1.
Collapse
Affiliation(s)
- En-Qi Qiao
- Department of Breast Surgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Hong-Jian Yang
- Department of Breast Surgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Xi-Ping Zhang
- Department of Breast Surgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310022, China
| |
Collapse
|
21
|
Hinman JD, Ngo KJ, Kim D, Chen C, Abraham CR, Ghanbari M, Ikram MA, Kushner SA, Kawaguchi R, Coppola G, Goth K, Bellusci S, Hernandez I, Kosik KS, Fogel BL. miR-142-3p regulates cortical oligodendrocyte gene co-expression networks associated with tauopathy. Hum Mol Genet 2021; 30:103-118. [PMID: 33555315 PMCID: PMC8496370 DOI: 10.1093/hmg/ddaa252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/13/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023] Open
Abstract
Oligodendrocytes exist in a heterogenous state and are implicated in multiple neuropsychiatric diseases including dementia. Cortical oligodendrocytes are a glial population uniquely positioned to play a key role in neurodegeneration by synchronizing circuit connectivity but molecular pathways specific to this role are lacking. We utilized oligodendrocyte-specific translating ribosome affinity purification and RNA-seq (TRAP-seq) to transcriptionally profile adult mature oligodendrocytes from different regions of the central nervous system. Weighted gene co-expression network analysis reveals distinct region-specific gene networks. Two of these mature myelinating oligodendrocyte gene networks uniquely define cortical oligodendrocytes and differentially regulate cortical myelination (M8) and synaptic signaling (M4). These two cortical oligodendrocyte gene networks are enriched for genes associated with dementia including MAPT and include multiple gene targets of the regulatory microRNA, miR-142-3p. Using a combination of TRAP-qPCR, miR-142-3p overexpression in vitro, and miR-142-null mice, we show that miR-142-3p negatively regulates cortical myelination. In rTg4510 tau-overexpressing mice, cortical myelination is compromised, and tau-mediated neurodegeneration is associated with gene co-expression networks that recapitulate both the M8 and M4 cortical oligodendrocyte gene networks identified from normal cortex. We further demonstrate overlapping gene networks in mature oligodendrocytes present in normal cortex, rTg4510 and miR-142-null mice, and existing datasets from human tauopathies to provide evidence for a critical role of miR-142-3p-regulated cortical myelination and oligodendrocyte-mediated synaptic signaling in neurodegeneration.
Collapse
Affiliation(s)
- Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Kathie J Ngo
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Deborah Kim
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Cidi Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118 USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad 13131 - 99137, Iran
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, The Netherlands
| | - Riki Kawaguchi
- Informatics Center for Neurogenetics and Neurogenomics, Semel Institute, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Giovanni Coppola
- Informatics Center for Neurogenetics and Neurogenomics, Semel Institute, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Kerstin Goth
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, 35392 Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, 35392 Giessen, Germany
- Department of Pulmonary and Critical Care Medicine, Key Laboratory of Interventional Pulmonology of Zhejiang Province, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Israel Hernandez
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Kenneth S Kosik
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Brent L Fogel
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
22
|
Nair MG, Somashekaraiah VM, Ramamurthy V, Prabhu JS, Sridhar TS. miRNAs: Critical mediators of breast cancer metastatic programming. Exp Cell Res 2021; 401:112518. [PMID: 33607102 DOI: 10.1016/j.yexcr.2021.112518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
MicroRNA mediated aberrant gene regulation has been implicated in several diseases including cancer. Recent research has highlighted the role of epigenetic modulation of the complex process of breast cancer metastasis by miRNAs. miRNAs play a crucial role in the process of metastatic evolution by facilitating alterations in the phenotype of tumor cells and the tumor microenvironment that promote this process. They act as critical determinants of the multi-step progression starting from carcinogenesis all the way to organotropism. In this review, we focus on the current understanding of the compelling role of miRNAs in breast cancer metastasis.
Collapse
Affiliation(s)
- Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India.
| | | | - Vishakha Ramamurthy
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| | - T S Sridhar
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
| |
Collapse
|
23
|
Liu P, Cao F, Sui J, Hong Y, Liu Q, Gao X, Gong H, Hao L, Lou Z, Zhang W. MicroRNA-142-3p Inhibits Tumorigenesis of Colorectal Cancer via Suppressing the Activation of Wnt Signaling by Directly Targeting to β-Catenin. Front Oncol 2021; 10:552944. [PMID: 33643894 PMCID: PMC7905391 DOI: 10.3389/fonc.2020.552944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Altered expression profile of microRNAs (miRNAs) was reported to be associated with colorectal cancer (CRC). The aims of this study are to identify the changed miRNAs in the plasma of CRC patients and explore the underlying mechanism of these miRNAs during tumorigenesis. METHODS Plasma miRNA expression profiles were compared between healthy people and CRC patients. MiRNA expression was measured using quantitative real-time PCR. Colony formation and MTT assays were used to test cell proliferation. Luciferase assay, immunohistochemistry and Western blotting were employed to explore the molecular mechanism. RESULTS MiR-142-3p level was found as the most significantly repressed miRNA in CRC patients. Overexpression of miR-142-3p dramatically repressed colony formation and cell proliferation of both HT29 and HCT116 cells while inhibition of miR-142-3p promoted those of the cells. Interestingly, overexpression of miR-142-3p reduced the level and nuclear accumulation of β-catenin. We further observed that miR-142-3p remarkably inhibited the transcriptional activity of β-catenin gene (CTNNB1). However, mutations in the predicted binding sites blocked this inhibition, suggesting that miR-142-3p may directly bind to the mRNA of β-catenin. CONCLUSION In conclusion, we identified miR-142-3p exerts its function as a tumor suppressor through blocking the activation of Wnt signaling by directly targeting to CTNNB1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Wei Zhang
- Colorectal Surgery Department, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Lin X, Wu Z, Hu H, Luo ML, Song E. Non-coding RNAs rewire cancer metabolism networks. Semin Cancer Biol 2021; 75:116-126. [PMID: 33421618 DOI: 10.1016/j.semcancer.2020.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Non-coding RNAs (ncRNAs) are functional RNAs with limited or no protein-coding ability. These interact with their target molecules and participate in the precise regulation of disease development. Metabolic reprogramming is a hallmark in cancer, and is considered essential in meeting increased macromolecular biosynthesis and energy generation of tumors. Recent studies have revealed the involvement of ncRNAs in several metabolic regulations of cancer through direct modulation of metabolic enzyme activities or participation of metabolism-related signaling pathways. Elucidation of how ncRNAs regulate metabolic reprogramming of cancers has opened up a novel intention to understand the mechanism of metabolic rewiring and also the opportunities of utilizing ncRNA-based therapeutics for targeting the metabolism in cancer treatment.
Collapse
Affiliation(s)
- Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Affiliated Hospital, Sun Yat-sen University, Shantou 515031, People's Republic of China; Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Zhiyong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Affiliated Hospital, Sun Yat-sen University, Shantou 515031, People's Republic of China
| | - Hai Hu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China.
| | - Man-Li Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China; Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, People's Republic of China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China; Fountain-Valley Institute for Life Sciences, 4th Floor, Building D, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Huangpu District, Guangzhou, People's Republic of China.
| |
Collapse
|
25
|
Interplay between p53 and non-coding RNAs in the regulation of EMT in breast cancer. Cell Death Dis 2021; 12:17. [PMID: 33414456 PMCID: PMC7791039 DOI: 10.1038/s41419-020-03327-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
The epithelial-mesenchymal transition (EMT) plays a pivotal role in the differentiation of vertebrates and is critically important in tumorigenesis. Using this evolutionarily conserved mechanism, cancer cells become drug-resistant and acquire the ability to escape the cytotoxic effect of anti-cancer drugs. In addition, these cells gain invasive features and increased mobility thereby promoting metastases. In this respect, the process of EMT is critical for dissemination of solid tumors including breast cancer. It has been shown that miRNAs are instrumental for the regulation of EMT, where they play both positive and negative roles often as a part of a feed-back loop. Recent studies have highlighted a novel association of p53 and EMT where the mutation status of p53 is critically important for the outcome of this process. Interestingly, p53 has been shown to mediate its effects via the miRNA-dependent mechanism that targets master-regulators of EMT, such as Zeb1/2, Snail, Slug, and Twist1. This regulation often involves interactions of miRNAs with lncRNAs. In this review, we present a detailed overview of miRNA/lncRNA-dependent mechanisms that control interplay between p53 and master-regulators of EMT and their importance for breast cancer.
Collapse
|
26
|
Grzywa TM, Klicka K, Włodarski PK. Regulators at Every Step-How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020; 12:E3709. [PMID: 33321819 PMCID: PMC7763175 DOI: 10.3390/cancers12123709] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial-mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
| |
Collapse
|
27
|
Valverde A, Nares S, Naqvi AR. Impaired cell migration and structural defects in myeloid cells overexpressing miR-30b and miR-142-3p. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194628. [PMID: 32979559 DOI: 10.1016/j.bbagrm.2020.194628] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/27/2022]
Abstract
Macrophages (MΦ) and dendritic cells (DC) play a fundamental role in shaping immune responses by sensing a plethora of Pathogen Associated Molecular Patterns (PAMPs), phagocytosis and antigen presentation to T lymphocytes. These important biological processes require efficient cell movement and an intact cellular morphology for dynamic interaction. The role of microRNAs (miRs) in this regard, however, is not well understood. In the present study, we show that miR-30b and miR-142-3p regulate migration and morphology of MΦ and DC. Transient overexpression of miR-30b and miR-142-3p attenuates migration and these cells display unique morphological deformities observed under electron microscopy. In addition, miR-142-3p overexpression in MΦ impaired phagocytosis of FITC-conjugated latex beads using live microscopy imaging. Interestingly, live cell imaging and F-actin staining revealed marked changes in the cell polarity and actin polymerization status, respectively. To identify miR-142-3p regulated pathways, we profiled global transcriptome changes in miR-142-3p or control mimic transfected DC. Expression of several genes were differentially altered by miR-142-3p and were associated with pathways related to cell movement, cell adhesion, and cytoskeletal rearrangement. Bioinformatics analysis identified a significant subset of downregulated genes with one or more predicted miR-142-3p binding sites in their 3'UTR strongly suggesting direct post-transcriptional impact of these miRNAs on multiple transcripts. Using dual luciferase assays, novel miR-142-3p binding sites were validated for three genes (Vinculin, Dab2 and Skap2) directly associated with cytoskeletal rearrangement and cell movement. In summary, our results show that miR-30b and miR-142-3p are regulators of myeloid cell cytoskeletal homeostasis and morphology.
Collapse
Affiliation(s)
- Araceli Valverde
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, United States of America
| | - Salvador Nares
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, United States of America
| | - Afsar Raza Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, United States of America.
| |
Collapse
|
28
|
Chao X, Jia Y, Feng X, Wang G, Wang X, Shi H, Zhao F, Jiang C. A Case-Control Study of ADCY9 Gene Polymorphisms and the Risk of Hepatocellular Carcinoma in the Chinese Han Population. Front Oncol 2020; 10:1450. [PMID: 32983975 PMCID: PMC7477943 DOI: 10.3389/fonc.2020.01450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/08/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Adenylyl cyclase type 9 (ADCY9) modulates signal transduction by producing the second messenger cyclic AMP. It has been reported that ADCY9 gene polymorphisms were associated with cancer development. The aim of this study was to investigate whether ADCY9 gene polymorphisms could contribute to the susceptibility of hepatocellular carcinoma (HCC) in the Chinese Han population. Methods: In the present study, five single-nucleotide polymorphisms (SNPs) in ADCY9 were genotyped using Agena MassARRAY platform in 876 subjects from China. Logistic regression was used to assess the effects of SNPs on HCC risk. Associations were also evaluated for HCC risk stratified by age and gender. False discovery rate (FDR) was used to correct multiple testing. Results: After adjusting for age and gender, we found a significant relationship between heterozygous genotypes of rs2531995 and HCC risk (OR = 1.34, 95% CI = 1.01–1.77, p = 0.045). ADCY9 rs2230742 had a strong relationship with lower risk of HCC in allele (p = 0.006), co-dominant (p = 0.023), dominant (p = 0.010), and additive (p = 0.006) models. Stratified analysis showed that rs879620 increased HCC risk and rs2230742 was associated with lower risk of HCC in the individuals aged 55 or younger, rs2531992 significantly decreased HCC risk in the elder group (age > 55). For women, rs2230742 and rs2230741 were significantly associated with HCC risk in multiple models (p < 0.05). FDR analysis showed that rs2230742 could protect individuals from HCC risk in the allele model (FDR-p = 0.030). In addition, haplotype analysis indicated that Crs879620Ars2230742Ars2230741 haplotype was a protective factor for HCC (OR = 0.67, 95% CI = 0.50–0.89, p = 0.007, FDR-p = 0.028). Conclusion: Our findings suggest that ADCY9 gene polymorphisms are associated with HCC risk in the Chinese Han population.
Collapse
Affiliation(s)
- Xu Chao
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China.,The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yong Jia
- The Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xuesong Feng
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Guoquan Wang
- The College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaoping Wang
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hailong Shi
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Fei Zhao
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chao Jiang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
29
|
Börschel CS, Stejskalova A, Schäfer SD, Kiesel L, Götte M. miR-142-3p Reduces the Size, Migration, and Contractility of Endometrial and Endometriotic Stromal Cells by Targeting Integrin- and Rho GTPase-Related Pathways That Regulate Cytoskeletal Function. Biomedicines 2020; 8:biomedicines8080291. [PMID: 32824678 PMCID: PMC7460043 DOI: 10.3390/biomedicines8080291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Downregulated microRNA-142-3p signaling contributes to the pathogenesis of endometriosis, an invasive disease where the lining of the uterus grows at ectopic locations, by yet incompletely understood mechanisms. Using bioinformatics and in vitro assays, this study identifies cytoskeletal regulation and integrin signaling as two relevant categories of miR-142-3p targets. qPCR revealed that miR-142-3p upregulation in St-T1b cells downregulates Rho-associated protein kinase 2 (ROCK2), cofilin 2 (CFL2), Ras-related C3 botulinum toxin substrate 1 (RAC1), neural Wiskott-Aldrich syndrome protein (WASL), and integrin α-V (ITGAV). qPCR and Western-blotting showed miR-142-3p effect on WASL and ITGAV was significant also in primary endometriotic stroma cells. Luciferase reporter assays in ST-T1b cells then confirmed direct regulation of ITGAV and WASL. On the functional side, miR-142-3p upregulation significantly reduced ST-T1b cell size, the size of vinculin plaques, migration through fibronectin-coated transwell filters, and the ability of ST-T1b and primary endometriotic stroma cells to contract collagen I gels. These results suggest that miR-142-3p has a strong mechanoregulatory effect on endometrial stroma cells and its external administration reduces the invasive endometrial phenotype. Within the limits of an in vitro investigation, our study provides new mechanistic insights into the pathogenesis of endometriosis and provides a perspective for the development of miR-142-3p based drugs for inhibiting invasive growth of endometriotic cells.
Collapse
Affiliation(s)
- Christin S. Börschel
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany; (C.S.B.); (S.D.S.); (L.K.)
- Department of Cardiology, University Heart and Vascular Centre Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anna Stejskalova
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany; (C.S.B.); (S.D.S.); (L.K.)
- Correspondence: (A.S.); (M.G.); Tel.: +49-251-835-6117 (M.G.)
| | - Sebastian D. Schäfer
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany; (C.S.B.); (S.D.S.); (L.K.)
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany; (C.S.B.); (S.D.S.); (L.K.)
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany; (C.S.B.); (S.D.S.); (L.K.)
- Correspondence: (A.S.); (M.G.); Tel.: +49-251-835-6117 (M.G.)
| |
Collapse
|
30
|
Najminejad H, Farhadihosseinabadi B, Dabaghian M, Dezhkam A, Rigi Yousofabadi E, Najminejad R, Abdollahpour-Alitappeh M, Karimi MH, Bagheri N, Mahi-Birjand M, Ghasemi N, Mazaheri M, Kalantar SM, Seifalian A, Sheikhha MH. Key Regulatory miRNAs and their Interplay with Mechanosensing and Mechanotransduction Signaling Pathways in Breast Cancer Progression. Mol Cancer Res 2020; 18:1113-1128. [PMID: 32430354 DOI: 10.1158/1541-7786.mcr-19-1229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/14/2020] [Accepted: 05/15/2020] [Indexed: 11/16/2022]
Abstract
According to the WHO, breast cancer is the most common cancer in women worldwide. Identification of underlying mechanisms in breast cancer progression is the main concerns of researches. The mechanical forces within the tumor microenvironment, in addition to biochemical stimuli such as different growth factors and cytokines, activate signaling cascades, resulting in various changes in cancer cell physiology. Cancer cell proliferation, invasiveness, migration, and, even, resistance to cancer therapeutic agents are changed due to activation of mechanotransduction signaling. The mechanotransduction signaling is frequently dysregulated in breast cancer, indicating its important role in cancer cell features. So far, a variety of experimental investigations have been conducted to determine the main regulators of the mechanotransduction signaling. Currently, the role of miRNAs has been well-defined in the cancer process through advances in molecular-based approaches. miRNAs are small groups of RNAs (∼22 nucleotides) that contribute to various biological events in cells. The central role of miRNAs in the regulation of various mediators involved in the mechanotransduction signaling has been well clarified over the last decade. Unbalanced expression of miRNAs is associated with different pathologic conditions. Overexpression and downregulation of certain miRNAs were found to be along with dysregulation of mechanotransduction signaling effectors. This study aimed to critically review the role of miRNAs in the regulation of mediators involved in the mechanosensing pathways and clarify how the cross-talk between miRNAs and their targets affect the cell behavior and physiology of breast cancer cells.
Collapse
Affiliation(s)
- Hamid Najminejad
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Behrouz Farhadihosseinabadi
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center (NRC), Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Dabaghian
- Research and Development Department, Razi Vaccine and serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Asiyeh Dezhkam
- Department of Midwifery, School of Nursing and Midwifery, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Reza Najminejad
- Department of Internal Medicine, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | | | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Motahareh Mahi-Birjand
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nasrin Ghasemi
- Abortion Research Centre, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahta Mazaheri
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mehdi Kalantar
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (Ltd), The London BioScience Innovation Centre, London, United Kingdom.
| | - Mohammad Hasan Sheikhha
- Genetics and Biotechnology Lab, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
31
|
Biber G, Ben-Shmuel A, Sabag B, Barda-Saad M. Actin regulators in cancer progression and metastases: From structure and function to cytoskeletal dynamics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:131-196. [PMID: 33066873 DOI: 10.1016/bs.ircmb.2020.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cytoskeleton is a central factor contributing to various hallmarks of cancer. In recent years, there has been increasing evidence demonstrating the involvement of actin regulatory proteins in malignancy, and their dysregulation was shown to predict poor clinical prognosis. Although enhanced cytoskeletal activity is often associated with cancer progression, the expression of several inducers of actin polymerization is remarkably reduced in certain malignancies, and it is not completely clear how these changes promote tumorigenesis and metastases. The complexities involved in cytoskeletal induction of cancer progression therefore pose considerable difficulties for therapeutic intervention; it is not always clear which cytoskeletal regulator should be targeted in order to impede cancer progression, and whether this targeting may inadvertently enhance alternative invasive pathways which can aggravate tumor growth. The entire constellation of cytoskeletal machineries in eukaryotic cells are numerous and complex; the system is comprised of and regulated by hundreds of proteins, which could not be covered in a single review. Therefore, we will focus here on the actin cytoskeleton, which encompasses the biological machinery behind most of the key cellular functions altered in cancer, with specific emphasis on actin nucleating factors and nucleation-promoting factors. Finally, we discuss current therapeutic strategies for cancer which aim to target the cytoskeleton.
Collapse
Affiliation(s)
- G Biber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - A Ben-Shmuel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - B Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - M Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
32
|
Knockdown of Musashi RNA Binding Proteins Decreases Radioresistance but Enhances Cell Motility and Invasion in Triple-Negative Breast Cancer. Int J Mol Sci 2020; 21:ijms21062169. [PMID: 32245259 PMCID: PMC7139790 DOI: 10.3390/ijms21062169] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 12/28/2022] Open
Abstract
The therapeutic potential of Musashi (MSI) RNA-binding proteins, important stemness-associated gene expression regulators, remains insufficiently understood in breast cancer. This study identifies the interplay between MSI protein expression, stem cell characteristics, radioresistance, cell invasiveness and migration. MSI-1, MSI-2 and Notch pathway elements were investigated via quantitative polymerase chain reaction (qPCR) in 19 triple-negative breast cancer samples. Measurements were repeated in MDA-MB-231 cells after MSI-1 and -2 siRNA-mediated double knockdown, with further experiments performed after MSI silencing. Flow cytometry helped quantify expression of CD44 and leukemia inhibitory factor receptor (LIFR), changes in apoptosis and cell cycle progression. Proliferation and irradiation-induced effects were assessed using colony formation assays. Radiation-related proteins were investigated via Western blots. Finally, cell invasion assays and digital holographic microscopy for cell migration were performed. MSI proteins showed strong correlations with Notch pathway elements. MSI knockdown resulted in reduction of stem cell marker expression, cell cycle progression and proliferation, while increasing apoptosis. Cells were radiosensitized as radioresistance-conferring proteins were downregulated. However, MSI-silencing-mediated LIFR downregulation resulted in enhanced cell invasion and migration. We conclude that, while MSI knockdown results in several therapeutically desirable consequences, enhanced invasion and migration need to be counteracted before knockdown advantages can be fully exploited.
Collapse
|
33
|
Ha J, Park C, Park C, Park S. IMIPMF: Inferring miRNA-disease interactions using probabilistic matrix factorization. J Biomed Inform 2020; 102:103358. [DOI: 10.1016/j.jbi.2019.103358] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/11/2019] [Accepted: 12/12/2019] [Indexed: 12/09/2022]
|
34
|
Dastmalchi N, Safaralizadeh R, Baradaran B, Hosseinpourfeizi M, Baghbanzadeh A. An update review of deregulated tumor suppressive microRNAs and their contribution in various molecular subtypes of breast cancer. Gene 2019; 729:144301. [PMID: 31884105 DOI: 10.1016/j.gene.2019.144301] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Breast cancer (BC) is histologically classified into hormone-receptor+ (ER+, PR + ), human epidermal growth factor receptor-2+ (Her2 + ), and triple-negative breast cancer (TNBC) types. The important contribution of tumor-suppressive (TS) microRNAs (miRs) in BC development and treatment have been well-acknowledged in the literature. OBJECTIVE The present review focused on the contribution of recently examined TS miRs in the progression and treatment of various histological subtypes of BC. RESULTS In summary, various miRs have tumor-suppressive roles in BC, so that their aberrant expression leads to the abnormality in the cellular processes such as enhanced cell growth, decreased apoptosis, cell migration and metastasis, and decreased sensitivity to chemotherapy through deregulated expression of oncogene targets of TS miRs. CONCLUSION TS miRs could be regarded as a proper molecular target for target therapy of BC. However, further in vitro and in vivo investigations are required to confirm the exact molecular functions of TS miRs in BC cells to offer more efficient targeted therapies.
Collapse
Affiliation(s)
- Narges Dastmalchi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Sinclair D, Hooker G. Sparse inverse covariance estimation for high-throughput microRNA sequencing data in the Poisson log-normal graphical model. J STAT COMPUT SIM 2019. [DOI: 10.1080/00949655.2019.1657116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- David Sinclair
- Department of Statistical Science, Cornell University, Ithaca, NY, USA
| | - Giles Hooker
- Department of Statistical Science, Cornell University, Ithaca, NY, USA
| |
Collapse
|
36
|
Xu T, He BS, Pan B, Pan YQ, Sun HL, Liu XX, Xu XN, Chen XX, Zeng KX, Xu M, Wang SK. MiR-142-3p functions as a tumor suppressor by targeting RAC1/PAK1 pathway in breast cancer. J Cell Physiol 2019; 235:4928-4940. [PMID: 31674013 DOI: 10.1002/jcp.29372] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/07/2019] [Indexed: 12/24/2022]
Abstract
MicroRNA-142-3p (miR-142-3p) was previously investigated in various cancers, whereas, it's role in breast cancer (BC) remains far from understood. In this study, we found that miR-142-3p was markedly decreased both in cell lines and BC tumor tissues. Elevated miR-142-3p expression suppressed growth and metastasis of BC cell lines via gain-of-function assay in vitro and in vivo. Mechanistically, miR-142-3p could regulate the ras-related C3 botulinum toxin substrate 1 (RAC1) expression in protein level, which simultaneously suppressed the epithelial-to-mesenchymal transition related protein levels and the activity of PAK1 phosphorylation, respectively. In addition, rescue experiments revealed RAC1 overexpression could reverse tumor-suppressive role of miR-142-3p. Our results showed miR-142-3p could function as a tumor suppressor via targeting RAC1/PAK1 pathway in BC, suggesting a potent therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bang-Shun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bei Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Qin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hui-Ling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang-Xiang Liu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xue-Ni Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Medical College, Southeast University, Nanjing, China
| | - Xiao-Xiang Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Medical College, Southeast University, Nanjing, China
| | - Kai-Xuan Zeng
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Medical College, Southeast University, Nanjing, China
| | - Mu Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shu-Kui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
MicroRNAs Contribute to Breast Cancer Invasiveness. Cells 2019; 8:cells8111361. [PMID: 31683635 PMCID: PMC6912645 DOI: 10.3390/cells8111361] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer statistics in 2018 highlight an 8.6 million incidence in female cancers, and 4.2 million cancer deaths globally. Moreover, breast cancer is the most frequent malignancy in females and twenty percent of these develop metastasis. This provides only a small chance for successful therapy, and identification of new molecular markers for the diagnosis and prognostic prediction of metastatic disease and development of innovative therapeutic molecules are therefore urgently required. Differentially expressed microRNAs (miRNAs) in cancers cause multiple changes in the expression of the tumorigenesis-promoting genes which have mostly been investigated in breast cancers. Herein, we summarize recent data on breast cancer-specific miRNA expression profiles and their participation in regulating invasive processes, in association with changes in cytoskeletal structure, cell-cell adhesion junctions, cancer cell-extracellular matrix interactions, tumor microenvironments, epithelial-to-mesenchymal transitions and cancer cell stem abilities. We then focused on the epigenetic regulation of individual miRNAs and their modified interactions with other regulatory genes, and reviewed the function of miRNA isoforms and exosome-mediated miRNA transfer in cancer invasiveness. Although research into miRNA’s function in cancer is still ongoing, results herein contribute to improved metastatic cancer management.
Collapse
|
38
|
Fan Y, Mu J, Huang M, Imani S, Wang Y, Lin S, Fan J, Wen Q. Epigenetic identification of ADCY4 as a biomarker for breast cancer: an integrated analysis of adenylate cyclases. Epigenomics 2019; 11:1561-1579. [PMID: 31584294 DOI: 10.2217/epi-2019-0207] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To explore the role of adenylyl cyclase isoforms and its epigenetics in cancer. Materials & methods: Adenylyl cyclase expression profiles, epigenetic alterations, prognostic value and molecular networks were assessed by use of public omics datasets. Results: ADCY4 was significantly downregulated in breast cancer. This downregulation was associated with promoter hypermethylation. High ADCY4 expression was correlated with better survival of patients with breast cancer and its different intrinsic subtypes and tumor stages. ADCY4 was shown to be strongly associated with G protein coupled receptors and the downstream cAMP signaling pathway, which was also significantly enriched in newly identified lysophosphatidic acid receptor 4 and glucagon-like peptide-1. Conclusion: ADCY4 may be used as an epigenetic biomarker for breast cancer, as well as a possible target for therapy.
Collapse
Affiliation(s)
- Yu Fan
- Oncology Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| | - Junhao Mu
- Chongqing Key Laboratory of Molecular Oncology & Epigenetics, The First Affiliated Hospital of Chongqing Medical University, 400010 Chongqing, PR China
| | - Mingquan Huang
- Breast Surgery Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| | - Saber Imani
- Oncology Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| | - Yu Wang
- Health Examination Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| | - Sheng Lin
- Oncology Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| | - Juan Fan
- Oncology Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| | - Qinglian Wen
- Oncology Department, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, PR China
| |
Collapse
|
39
|
Mansoori B, Mohammadi A, Gjerstorff MF, Shirjang S, Asadzadeh Z, Khaze V, Holmskov U, Kazemi T, Duijf PHG, Baradaran B. miR-142-3p is a tumor suppressor that inhibits estrogen receptor expression in ER-positive breast cancer. J Cell Physiol 2019; 234:16043-16053. [PMID: 30741415 DOI: 10.1002/jcp.28263] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
Estrogen receptors (ERs) are involved in the development of many types of malignant tumors, in particular, breast cancer. Among others, ERs affect cell growth, proliferation, and differentiation. The microRNA (miRNA) miR-142-3p has been shown to inhibit carcinogenesis by regulating various cellular processes, including cell cycle progression, cell migration, apoptosis, and invasion. It does so via targeting molecules involved in a range of signaling pathways. We surgically collected 20 ER-positive breast cancer samples, each with matched adjacent normal breast tissue, and measured the expression of miR-142-3p via quantitative real-time polymerase chain reaction (qRT-PCR). Bioinformatics methods, luciferase reporter assay, qRT-PCR, and western blot analysis were used to assess whether miR-142-3p could target ESR1, which encodes the estrogen receptor, in ER-positive breast cancer cells and patient samples. We also restored miRNA expression and performed cell viability, cytotoxicity, and colony formation assays. Western blot analysis and qRT-PCR were used to study the expression of apoptosis and stemness markers. We found that miR-142-3p is downregulated in ER-positive breast cancers. Restoration of miR-142-3p expression in ER-positive breast cancer cells reduced cell viability, induced apoptosis via the intrinsic pathway and decreased both colony formation and the expression of stem cell markers. Bioinformatic analysis predicted miR-142-3p could bind to 3'-untranslated region ESR1 messenger RNA (mRNA). Consistently, we demonstrated that miR-142-3p reduced luciferase activity in ER-positive breast cancer cells, and decreased ESR1 expression in both mRNA and protein levels. The results revealed miR-142-3p and ESR1 expression correlated negatively in ER-positive breast cancer samples. The results suggest miR-142-3p acts as a tumor suppressor via multiple mechanisms. Thus, restoration of miR-142-3p expression, for example, via miRNA replacement therapy, may represent an effective strategy for the treatment of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Ma L, Li Z, Li W, Ai J, Chen X. MicroRNA-142-3p suppresses endometriosis by regulating KLF9-mediated autophagy in vitro and in vivo. RNA Biol 2019; 16:1733-1748. [PMID: 31425004 DOI: 10.1080/15476286.2019.1657352] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The detailed pathogenesis of endometriosis remains largely unclear despite decades of research. Recent studies have demonstrated that miRNAs plays an important role in endometriosis. The expression of miR-142-3p was decreased in ectopic endometrial tissues, while KLF9 and VEGFA expression levels were increased. Overexpression of miR-142-3p or knockdown of KLF9 significantly suppressed CRL-7566 cell proliferation and metastasis, induced cell apoptosis, and decreased both cell autophagy and vascularization. Additionally, KLF9 was confirmed to be a direct target of miR-142-3p and to directly bind to the promoter of the VEGFA gene, regulating its expression. Finally, intraperitoneal injection of miR-142-3p lentivirus significantly attenuated ectopic endometriotic lesions in vivo.miR-142-3p directly targeted KLF9, regulated VEGFA expression, and was protective against the growth of ectopic endometriotic lesions. Therefore, the miR-142-3p/KLF9/VEGFA signalling pathway may be a potential target in endometriosis treatment.
Collapse
Affiliation(s)
- Lin Ma
- Reproductive Medicine center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zaiyi Li
- Reproductive Medicine center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weihao Li
- Reproductive Medicine center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jing Ai
- Reproductive Medicine center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaoxuan Chen
- Reproductive Medicine center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
41
|
Ghanbari M, Munshi ST, Ma B, Lendemeijer B, Bansal S, Adams HH, Wang W, Goth K, Slump DE, den Hout MC, IJcken WF, Bellusci S, Pan Q, Erkeland SJ, Vrij FM, Kushner SA, Ikram MA. A functional variant in the miR‐142 promoter modulating its expression and conferring risk of Alzheimer disease. Hum Mutat 2019; 40:2131-2145. [DOI: 10.1002/humu.23872] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/13/2019] [Accepted: 07/11/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Genetics, School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Shashini T. Munshi
- Department of Psychiatry, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Buyun Ma
- Department of Gastroenterology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Bas Lendemeijer
- Department of Psychiatry, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Sakshi Bansal
- Department of Psychiatry, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Hieab H. Adams
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Wenshi Wang
- Department of Gastroenterology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Kerstin Goth
- Department of Lung Matrix Remodeling, Excellence Cluster Cardio‐Pulmonary System (ECCPS) University Justus Liebig Giessen Giessen Germany
| | - Denise E. Slump
- Department of Psychiatry, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Mirjam C.G.N. den Hout
- Center for Biomics, Department of Cell Biology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Wilfred F.J. IJcken
- Center for Biomics, Department of Cell Biology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Saverio Bellusci
- Department of Lung Matrix Remodeling, Excellence Cluster Cardio‐Pulmonary System (ECCPS) University Justus Liebig Giessen Giessen Germany
| | - Qiuwei Pan
- Department of Gastroenterology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Stefan J. Erkeland
- Department of Immunology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Femke M.S. Vrij
- Department of Psychiatry, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Steven A. Kushner
- Department of Psychiatry, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| |
Collapse
|
42
|
Su Q, Liu Y, Lv XW, Ye ZL, Sun YH, Kong BH, Qin ZB. Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. J Mol Cell Cardiol 2019; 133:12-25. [PMID: 31145943 DOI: 10.1016/j.yjmcc.2019.05.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play a central role in regulating heart diseases. In the present study, we examined the effects of lncRNA taurine up-regulated gene 1 (TUG1) in ischemia/reperfusion (I/R)- or hydrogen peroxide-challenged cardiomyocytes, with specific focus on autophagy-induced cell apoptosis. METHODS The expressions of miR-142-3p and TUG1 in H2O2-challenged cardiomyocytes and I/R-injured heart tissue were measured by RT-qPCR. Cell death was measured by trypan blue staining assay. Cell apoptosis was determined by Annexin V/PI staining and TUNEL assay. Autophagy was examined by quantifying cells or tissues containing LC3+ autophagic vacuoles by immunofluorescence, or by measuring the expressions of autophagy-related biomarkers by Western blot. The direct interaction between miR-142-3p and TUG1, high mobility group box 1 protein (HMGB1), or Ras-related C3 botulinum toxin substrate 1 (Rac1) was examined using luciferase reporter assay. The significance of miR-142-3p and TUG1 on cell apoptosis or autophagy was examined using both gain-of-function and loss-of-function approaches. The importance of HMGB1 or Rac1 was assessed using siRNA-mediated gene silencing. RESULTS miR-142-3p was down-regulated, while TUG1 up-regulated in H2O2-challenged cardiomyocytes in vitro and I/R-injured heart tissues in vivo. Functionally, inhibition of TUG1 and overexpression of miR-142-3p inhibited cell apoptosis and autophagy in cardiomyocytes. The function of TUG1 were achieved by sponging miR-142-3p and releasing the suppression of the putative targets of miR-142-3p, HMGB1 and Rac1. Both HMGB1 and Rac1 essentially mediated cell apoptosis and autophagy induced by TUG1. CONCLUSIONS TUG1, by targeting miR-142-3p and up-regulating HMGB1 and Rac1, plays a central role in stimulating autophagic cell apoptosis in ischemia/hypoxia-challenged cardiomyocytes. Down-regulating TUG1 or up-regulating miR-142-3p may ameliorate myocardial injury and protect against acute myocardial infarction.
Collapse
Affiliation(s)
- Qiang Su
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, PR China.
| | - Yang Liu
- Department of Cardiology, The Second People's Hospital of Nanning City, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530031, PR China
| | - Xiang-Wei Lv
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, PR China
| | - Zi-Liang Ye
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, PR China
| | - Yu-Han Sun
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China
| | - Bing-Hui Kong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China
| | - Zhen-Bai Qin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China
| |
Collapse
|
43
|
Wang H, Chen H, Jiang Z, Lin Y, Wang X, Xiang J, Peng J. Integrin subunit alpha V promotes growth, migration, and invasion of gastric cancer cells. Pathol Res Pract 2019; 215:152531. [PMID: 31320250 DOI: 10.1016/j.prp.2019.152531] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/12/2019] [Accepted: 07/05/2019] [Indexed: 12/15/2022]
Abstract
Integrin subunit alpha V (ITGAV), a member of integrin family of extracellular matrix receptors, is involved in many types of cancer. In this study, the expression levels, clinical features and prognosis of ITGAV in gastric cancer (GC) patients were investigated, and the functional roles of ITGAV were also investigated. Cell Counting Kit-8 (CCK-8) assay was performed to examine the proliferation of GC cells. Transwell assays and wound-healing assays were conducted to explore the effect of ITGAV expression on GC cell migration and invasion. We found that ITGAV was overexpressed in both GC tissues and GC cells. ITGAV expression was positively correlated with lymph node metastasis and TNM stage of GC. High expression of ITGAV was associated with shorter overall survival (OS) and disease-free survival (DFS). Interestingly, the downregulation of ITGAV resulted in suppression of proliferation, migration, and invasion in GC cells. In conclusion, ITGAV is overexpressed in gastric cancer and is associated with poorer prognostic outcomes. ITGAV may serve as an important prognostic marker for GC staging and progression.
Collapse
Affiliation(s)
- Huashe Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China
| | - Honglei Chen
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China; Department of Gastrointestinal Endoscopy, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China
| | - Zhipeng Jiang
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China
| | - Yijia Lin
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China
| | - Xinyou Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China
| | - Jun Xiang
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China
| | - Junsheng Peng
- Department of Gastrointestinal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, PR China.
| |
Collapse
|
44
|
Jin C, Xiao L, Zhou Z, Zhu Y, Tian G, Ren S. MiR-142-3p suppresses the proliferation, migration and invasion through inhibition of NR2F6 in lung adenocarcinoma. Hum Cell 2019; 32:437-446. [PMID: 31168689 DOI: 10.1007/s13577-019-00258-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/06/2019] [Indexed: 12/09/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide and lung adenocarcinoma is its main type. MicroRNAs are small, non-coding and single-strand RNAs that regulate gene expression in human cancers. The aim of our study is to investigate the underlying molecular mechanism of miR-142-3p in NSCLC. The expression of miR-142-3p in lung adenocarcinoma tissues and cells was detected by RT-qPCR. Next, cell proliferation, migration, invasion and apoptosis were examined by CCK-8, scratch assay, transwell assay and flow cytometry in A549 and HCC827 cells, respectively. Then, the target of miR-142-3p was predicted by targetscanHuman 7.2 and confirmed using dual-luciferase reporter assay. Additionally, RT-qPCR and western blot were used to detect the expression of NR2F6, MMP2, MMP9 and caspase-3. The results showed that miR-142-3p expression was significantly decreased in tumor tissues and cells. Overexpression of miR-142-3p inhibited the proliferation, migration, invasion and promoted cell apoptosis in vitro, while knockdown of miR-142-3p had reversed function. Furthermore, NR2F6 was identified as a direct target of miR-142-3p, which was negatively correlated with miR-142-3p expression. Finally, miR-142-3p overexpression suppressed the expression of NR2F6, MMP2 and MMP9, but improved caspase-3 expression, while miR-142-3p knockdown got the opposite expression results. Suppressing MMP2 and MMP9 activities inhibited cell invasion. In summary, these findings indicated that miR-142-3p inhibits lung adenocarcinoma cell proliferation, migration and invasion, and enhances cell apoptosis by targeting NR2F6, suggesting that miR-142-3p may be a novel therapeutic target for lung adenocarcinoma treatment.
Collapse
Affiliation(s)
- Chang'e Jin
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Liang Xiao
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Zeqiang Zhou
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Yan Zhu
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Geng Tian
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Shuhua Ren
- Department of Thoracic Surgery, Tangshan Gongren Hospital, No. 27, Wenhua Road, Tangshan, 063000, Hebei, China.
| |
Collapse
|
45
|
Peng D, Dong J, Zhao Y, Peng X, Tang J, Chen X, Wang L, Hu DN, Reinach PS, Qu J, Yan D. miR-142-3p suppresses uveal melanoma by targeting CDC25C, TGFβR1, GNAQ, WASL, and RAC1. Cancer Manag Res 2019; 11:4729-4742. [PMID: 31213897 PMCID: PMC6541795 DOI: 10.2147/cmar.s206461] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/18/2019] [Indexed: 02/03/2023] Open
Abstract
Purpose: Uveal melanoma (UM) is the most frequent metastatic ocular tumor in adults. Therapeutic intervention remains ineffective since none of the novel procedures used to treat this disease increased survival rates. To deal with this limitation, additional studies are required to clarify its pathogenesis. The current study focused on describing how epigenetic modulation by miR-142-3p affects changes in some cellular functions underlying UM pathogenesis. Methods and results: Microarray analysis identified 374 miRNAs which were differentially expressed between UM cells and uveal melanocytes. miR-142-3p was one of the 10 most downregulated miRNAs. Quantitative RT-PCR analysis confirmed that miR-142-3p expression levels were significantly decreased in both UM cell lines and clinical specimens. The results of the MTS, clone formation, scratch wound, transwell assays, and in vivo biofluorescence imaging showed that miR-142-3p overexpression significantly inhibited cell proliferation, migration, and invasiveness. Nevertheless, miR-142-3p did not affect cell apoptotic activity or sensitivity to doxorubicin. Cell cycle and EdU analysis showed that miR-142-3p overexpression induced G1/G2 cell cycle arrest and reduced DNA synthesis in UM cells. Microarray analysis showed that miR-142-3p mainly regulates the TGFβ signaling pathway, and those in which MAPK and PI3K-Akt are constituents. Functional interactions between miR-142-3p and CDC25C, TGFβR1, GNAQ, WASL, and RAC1 target genes were confirmed based on the results of the luciferase reporter assay and Western blot analysis. CDC25C or RAC1 downregulation is in agreement with cell cycle arrest and DNA synthesis disorder induction, while downregulation of TGFβR1, GNAQ, WASL, or RAC1 accounts for declines in cell migration. Conclusion: miR-143-3p is a potential therapeutic target to treat UM since overriding its declines in expression that occur in this disease reversed the pathogenesis of this disease. Such insight reveals novel biomarker for decreasing UM vitality and for improved tracking of tumor progression.
Collapse
Affiliation(s)
- Dewei Peng
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Jing Dong
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Yunping Zhao
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaomei Peng
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Jingjing Tang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaoyan Chen
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Lihua Wang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Dan-Ning Hu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China.,Tissue Culture Center, New York Eye and Ear Infirmary, New York Medical College, New York, NY, USA
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Jia Qu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Dongsheng Yan
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
46
|
Chen M, Zhang Y, Li A, Li Z, Liu W, Chen Z. Bipartite Heterogeneous Network Method Based on Co-neighbor for MiRNA-Disease Association Prediction. Front Genet 2019; 10:385. [PMID: 31080459 PMCID: PMC6497741 DOI: 10.3389/fgene.2019.00385] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/10/2019] [Indexed: 12/22/2022] Open
Abstract
In recent years, miRNA variation and dysregulation have been found to be closely related to human tumors, and identifying miRNA-disease associations is helpful for understanding the mechanisms of disease or tumor development and is greatly significant for the prognosis, diagnosis, and treatment of human diseases. This article proposes a Bipartite Heterogeneous network link prediction method based on co-neighbor to predict miRNA-disease association (BHCN). According to the structural characteristics of the bipartite network, the concept of bipartite network co-neighbors is proposed, and the co-neighbors were used to represent the probability of association between disease and miRNA. To predict the isolated diseases and the new miRNA based on the association probability expressed by co-neighbors, we utilized the similarity between disease nodes and the similarity between miRNA nodes in heterogeneous networks to represent the association probability between disease and miRNA. The model's predictive performance was evaluated by the leave-one-out cross validation (LOOCV) on different datasets. The AUC value of BHCN on the gold benchmark dataset was 0.7973, and the AUC obtained on the prediction dataset was 0.9349, which was better than that of the classic global algorithm. In this case study, we conducted predictive studies on breast neoplasms and colon neoplasms. Most of the top 50 predicted results were confirmed by three databases, namely, HMDD, miR2disease, and dbDEMC, with accuracy rates of 96 and 82%. In addition, BHCN can be used for predicting isolated diseases (without any known associated diseases) and new miRNAs (without any known associated miRNAs). In the isolated disease case study, the top 50 of breast neoplasm and colon neoplasm potentials associated with miRNAs predicted an accuracy of 100 and 96%, respectively, thereby demonstrating the favorable predictive power of BHCN for potentially relevant miRNAs.
Collapse
Affiliation(s)
- Min Chen
- School of Computer Science and Technology, Hunan Institute of Technology, Hengyang, China
| | - Yi Zhang
- School of Information Science and Engineering, Guilin University of Technology, Guilin, China
| | - Ang Li
- School of Computer Science and Technology, Hunan Institute of Technology, Hengyang, China
| | - Zejun Li
- School of Computer Science and Technology, Hunan Institute of Technology, Hengyang, China
| | - Wenhua Liu
- School of Computer Science and Technology, Hunan Institute of Technology, Hengyang, China
| | - Zheng Chen
- School of Computer Science and Technology, Hunan Institute of Technology, Hengyang, China
| |
Collapse
|
47
|
Liu J, Tian W, Zhang W, Jia Y, Yang X, Wang Y, Zhang J. MicroRNA-142-3p/MALAT1 inhibits lung cancer progression through repressing β-catenin expression. Biomed Pharmacother 2019; 114:108847. [PMID: 30970294 DOI: 10.1016/j.biopha.2019.108847] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/29/2022] Open
Abstract
MALAT1 is well documented to be highly expressed in non-small cell lung cancer (NSCLC) and its overexpression closely associates the malignant phenotype of NSCLC cells and poor prognosis of NSCLC patients. MALAT1 is also identified to enhance β-catenin expression and under the negative regulation of miR-142-3p. However, the role of miR-142-3p/MALAT1/β-catenin in the occurrence and development of NSCLC remains unclear. The objective of this study was to explore it. The results showed that miR-142-3p expression was reduced in NSCLC tissues, while β-catenin and MALAT1 expression levels were elevated. MTT, transwell chamber, flow cytometry assays demonstrated that up-regulation of miR-142-3p with mimic transfection significantly inhibited the proliferation, migration and promoted the apoptosis of NSCLC H1299 cells, and induced a G0/G1 phase arrest and S phase reduction. Besides, miR-142-3p negatively decreased MALAT1 expression as detected by RT-PCR and luciferase reporter assays. Moreover, up-regulation of miR-142-3p decreased β-catenin expression through down-regulating MALAT1 in H1299 cells. And in vivo experiment showed that miR-142-3p up-regulation, as well as the knockdown of either β-catenin or MALAT1 significantly reduced the tumorigenesis of NSCLC cells. Taken together, our study makes clear that miR-142-3p functions as a tumor suppressor in NSCLC progression through inhibiting MALAT1/β-catenin signaling.
Collapse
Affiliation(s)
- Jingyi Liu
- Department of Cancer Biotherapy, Shanxi Academy of Medical Sciences (Shanxi Dayi Hospital), Taiyuan 030032, China.
| | - Weiwei Tian
- Department of Endoscopy, Shanxi Province Cancer Hospital, Taiyuan 030013, China
| | - Wenbin Zhang
- Department of Hematology, Shanxi Academy of Medical Sciences (Shanxi Dayi Hospital), Taiyuan 030032, China
| | - Yuan Jia
- Department of Cancer Biotherapy, Shanxi Academy of Medical Sciences (Shanxi Dayi Hospital), Taiyuan 030032, China
| | - Xiaoling Yang
- Department of Cancer Biotherapy, Shanxi Academy of Medical Sciences (Shanxi Dayi Hospital), Taiyuan 030032, China
| | - Yanli Wang
- Department of Cancer Biotherapy, Shanxi Academy of Medical Sciences (Shanxi Dayi Hospital), Taiyuan 030032, China
| | - Junping Zhang
- Department of Cancer Biotherapy, Shanxi Academy of Medical Sciences (Shanxi Dayi Hospital), Taiyuan 030032, China
| |
Collapse
|
48
|
Ha J, Park C, Park S. PMAMCA: prediction of microRNA-disease association utilizing a matrix completion approach. BMC SYSTEMS BIOLOGY 2019; 13:33. [PMID: 30894171 PMCID: PMC6425656 DOI: 10.1186/s12918-019-0700-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 01/29/2019] [Indexed: 01/29/2023]
Abstract
Background Numerous experimental results have indicated that microRNAs (miRNAs) play a vital role in biological processes, as well as outbreaks of diseases at the molecular level. Despite their important role in biological processes, knowledge regarding specific functions of miRNAs in the development of human diseases is very limited. While attempting to solve this problem, many computational approaches have been proposed and attracted significant attention. However, most previous approaches suffer from the common problem of being inapplicable to new diseases without any known miRNA-disease associations. Results This paper proposes a novel method for inferring disease-miRNA associations utilizing a machine learning technique called matrix factorization, which is widely used in recommendation systems. In recommendation systems, the goal is to predict rating scores that a user might assign to specific items. By replacing users with miRNAs and items with diseases, we can efficiently predict miRNA-disease associations without seed miRNAs. As a result, our proposed model, called prediction of microRNA-disease association utilizing a matrix completion approach, achieves excellent performance compared to previous approaches with a reliable AUC value of 0.882 by implementing five-fold cross validation. Conclusions To the best of our knowledge, the proposed method applies the matrix completion technique to infer miRNA-disease associations and overcome the seed-miRNA problem negatively affects existing computational models. Electronic supplementary material The online version of this article (10.1186/s12918-019-0700-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jihwan Ha
- Department of Computer Science, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul, South Korea
| | - Chihyun Park
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, 9211 Euclid Ave., Cleveland, OH, 44106, USA
| | - Sanghyun Park
- Department of Computer Science, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul, South Korea.
| |
Collapse
|
49
|
Liu E, Liu Z, Zhou Y, Chen M, Wang L, Li J. MicroRNA‑142‑3p inhibits trophoblast cell migration and invasion by disrupting the TGF‑β1/Smad3 signaling pathway. Mol Med Rep 2019; 19:3775-3782. [PMID: 30864732 DOI: 10.3892/mmr.2019.9997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 07/02/2018] [Indexed: 11/06/2022] Open
Abstract
Insufficient invasion of trophoblasts is known to be associated with preeclampsia (PE) development. Recently, microRNAs (miRNAs) have been reported to serve important roles in the pathogenesis of PE. However, little is known regarding the regulation of trophoblastic invasion by miRNAs. The aim of the present study was to explore the role of miRNAs in trophoblastic invasion and the underlying molecular mechanism. Using a miRNA microarray, miRNAs putatively involved in the pathophysiology of PE were examined between normal and preeclamptic placentas. Validation analysis of miR‑142‑3p level in placenta specimens was performed using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Then, the regulation of miR‑142‑3p on trophoblast cells migration and invasion was evaluated using wound healing and transwell migration assays. Furthermore, the target gene of miR‑142‑3p and the downstream signaling pathway were also investigated. Microarray analysis and RT‑qPCR revealed that miR‑142‑3p was significantly upregulated in placenta specimens from patients with PE. Its overexpression inhibited trophoblast cell invasion and migration, whereas its knockdown enhanced trophoblast cell invasion and migration. In addition, overexpression of miR‑142‑3p inhibited the mRNA expression and the activities of matrix metalloproteinase‑2 (MMP2) and MMP9, which are closely associated with cell invasion and migration, while inhibition of miR‑142‑3p had the opposite result. Subsequent analyses demonstrated that transforming growth factor‑β1 (TGF‑β1) was a direct and functional target of miR‑142‑3p. Notably, the knockdown of TGF‑β1 effectively reversed the enhancement of miR‑142‑3p inhibitor on trophoblast cell invasion and migration. Finally, the present study confirmed that miR‑142‑3p inhibitor enhanced cell invasion and migration by reactivating the TGF‑β1/Smad3 signaling pathway. Taken together, the results of the present study suggest that miR‑142‑3p may serve an important role in human placental development by suppressing trophoblast cell invasion and migration through disruption of the TGF‑β1/smad3 signaling pathway, suggesting that knockdown of miR‑142‑3p may provide a novel therapy for PE.
Collapse
Affiliation(s)
- Enling Liu
- Department of Obstetrics and Gynecology, Tangshan Worker Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Zheng Liu
- Department of Rheumatology and Immunology, Tianjin General Hospital, Tianjin Medical University, Tianjin 300052, P.R. China
| | - Yuxiu Zhou
- Department of Rheumatology and Immunology, Tangshan Worker Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Mei Chen
- Department of Obstetrics and Gynecology, Tangshan Worker Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Liqun Wang
- Department of Obstetrics and Gynecology, Tangshan Worker Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Jun Li
- Department of Obstetrics and Gynecology, Tangshan Worker Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
50
|
Wu YS, Lin H, Chen D, Yi Z, Zeng B, Jiang Y, Ren G. A four-miRNA signature as a novel biomarker for predicting survival in endometrial cancer. Gene 2019; 697:86-93. [PMID: 30779946 DOI: 10.1016/j.gene.2019.01.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 01/07/2019] [Accepted: 01/22/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The microRNAs (miRNAs) have been validated as prognostic markers in many cancers. The aim of this study was to identify new miRNA prognostic biomarkers in endometrial cancer (EC) and to develop an expression-based miRNA signature to provide survival risk prediction for EC patients. METHODS From TCGA database, the miRNA datasets of EC and clinical information were downloaded in April 2018. Using univariate and multivariate Cox regression analyses identify prognostic factors. Using area under the curve (AUC) of receiver operating characteristic (ROC) curve assess the sensitivity and specificity of prognostic model. RESULTS 530 patients were randomly divided into training set and testing set. Among 561 differentially expressed miRNAs, 4 miRNAs (miR-4758, miR-876, miR-142, miR-190b) were demonstrated to be predictive biomarkers of overall survival (OS) for EC patients in training set. Based on the risk score of 4-miRNA model, patients in the training set were divided into high-risk and low-risk groups with significantly different OS. This 4-miRNA model was validated in testing and entire set. The AUC for the ROC curves in the entire set was 0.704. Meanwhile, multivariate Cox regression combined with other traditional clinical parameters indicated that the 4-miRNA model can be used as an independent OS prognostic factor. Functional enrichment analysis revealed that these miRNAs are involved in biological processes and pathways that are closely related to cancer. CONCLUSION A robust 4-miRNA signature as an independent prognostic factor for OS in EC patients was established.
Collapse
Affiliation(s)
- Yu-Shen Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Huapeng Lin
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, PR China
| | - Duke Chen
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ziying Yi
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Beilei Zeng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yicheng Jiang
- Department of Oncology, The People's Hospital of Chongqing Hechuan, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China; Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|