1
|
Wang Y, Su Q, Tang H, Lin X, Yi Y, Tian Q, Luo Z, Fu M, Peng J, Zhang K. Circ-USP9X accelerates deep vein thrombosis after fracture by acting as a miR-148b-3p sponge and upregulates SRC kinase signaling inhibitor 1. Clinics (Sao Paulo) 2024; 79:100403. [PMID: 38878321 PMCID: PMC11226750 DOI: 10.1016/j.clinsp.2024.100403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/11/2024] [Accepted: 05/12/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVES This study aims to elucidate the role of circUSP9X (Circular RNA Ubiquitin Specific Peptidase 9 X-Linked) in the development of venous thrombosis in the lower extremities. METHODS An animal model of Deep Vein Thrombosis (DVT) and a hypoxic model of Human Umbilical Vein Endothelial Cells (HUVECs) treated with Cobalt (II) Chloride (CoCl2) were developed. The expression levels of circUSP9X, microRNA-148b-3p (miR-148b-3p), and SRC Kinase Signaling Inhibitor 1 (SRCIN1) were quantified using quantitative reverse transcription Polymerase Chain Reaction and Western blot analysis. Cell cytotoxicity, viability, apoptosis, and inflammation in HUVECs were assessed via Lactate Dehydrogenase (LDH) assay, MTT assay, flow cytometry, Enzyme-Linked Immunosorbent Assay, and Western blot, respectively. Hematoxylin and Eosin staining were employed for histopathological examination of the venous tissues in the animal model. The interaction between circUSP9X, miR-148b-3p, and SRCIN1 was further explored through dual-luciferase reporter assays and RNA Immunoprecipitation experiments. RESULTS The present findings reveal a significant upregulation of circUSP9X and SRCIN1 and a concurrent downregulation of miR-148b-3p in DVT cases. Knockdown of circUSP9X or overexpression of miR-148b-3p ameliorated CoCl2-induced apoptosis in HUVECs, reduced LDH release, enhanced cellular viability, and mitigated inflammation. Conversely, overexpression of circUSP9X intensified CoCl2's cytotoxic effects. The effects of manipulating circUSP9X expression were counteracted by the corresponding modulation of miR-148b-3p and SRCIN1 levels. Additionally, circUSP9X knockdown effectively inhibited the formation of DVT in the mouse model. A competitive binding mechanism of circUSP9X for miR-148b-3p, modulating SRCIN1 expression, was identified. CONCLUSION circUSP9X promotes the formation of DVT through the regulation of the miR-148b-3p/SRCIN1 axis.
Collapse
Affiliation(s)
- YongChao Wang
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - Qin Su
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - HaiRong Tang
- School of Nursing, Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - Xin Lin
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - YanHua Yi
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - Qiang Tian
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - ZhangFeng Luo
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - MeiChun Fu
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - JiaQi Peng
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China
| | - KeYun Zhang
- Department of Joint Sport Medicine, The First Affiliated Hospital of Hunan Medical College, Huaihua City, Hunan Province, PR China.
| |
Collapse
|
2
|
Centonze G, Natalini D, Grasso S, Morellato A, Salemme V, Piccolantonio A, D'Attanasio G, Savino A, Bianciotto OT, Fragomeni M, Scavuzzo A, Poncina M, Nigrelli F, De Gregorio M, Poli V, Arina P, Taverna D, Kopecka J, Dupont S, Turco E, Riganti C, Defilippi P. p140Cap modulates the mevalonate pathway decreasing cell migration and enhancing drug sensitivity in breast cancer cells. Cell Death Dis 2023; 14:849. [PMID: 38123597 PMCID: PMC10733353 DOI: 10.1038/s41419-023-06357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/09/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
p140Cap is an adaptor protein involved in assembling multi-protein complexes regulating several cellular processes. p140Cap acts as a tumor suppressor in breast cancer (BC) and neuroblastoma patients, where its expression correlates with a better prognosis. The role of p140Cap in tumor metabolism remains largely unknown. Here we study the role of p140Cap in the modulation of the mevalonate (MVA) pathway in BC cells. The MVA pathway is responsible for the biosynthesis of cholesterol and non-sterol isoprenoids and is often deregulated in cancer. We found that both in vitro and in vivo, p140Cap cells and tumors show an increased flux through the MVA pathway by positively regulating the pace-maker enzyme of the MVA pathway, the 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), via transcriptional and post-translational mechanisms. The higher cholesterol synthesis is paralleled with enhanced cholesterol efflux. Moreover, p140Cap promotes increased cholesterol localization in the plasma membrane and reduces lipid rafts-associated Rac1 signalling, impairing cell membrane fluidity and cell migration in a cholesterol-dependent manner. Finally, p140Cap BC cells exhibit decreased cell viability upon treatments with statins, alone or in combination with chemotherapeutic at low concentrations in a synergistic manner. Overall, our data highlight a new perspective point on tumor suppression in BC by establishing a previously uncharacterized role of the MVA pathway in p140Cap expressing tumors, thus paving the way to the use of p140Cap as a potent biomarker to stratify patients for better tuning therapeutic options.
Collapse
Affiliation(s)
- Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Silvia Grasso
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Giacomo D'Attanasio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Aurora Savino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Olga Teresa Bianciotto
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Matteo Fragomeni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Andrea Scavuzzo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Matteo Poncina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Francesca Nigrelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Mario De Gregorio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, WC1E 6BT, London, UK
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Italy; Molecular Biotechnology Center, Piazza Nizza 44, 10126, Torino, Italy
| | - Sirio Dupont
- Department of Molecular Medicine (DMM), University of Padova, Padua, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Italy; Molecular Biotechnology Center, Piazza Nizza 44, 10126, Torino, Italy.
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
3
|
Xu WW, Liao L, Dai W, Zheng CC, Tan XP, He Y, Zhang QH, Huang ZH, Chen WY, Qin YR, Chen KS, He ML, Law S, Lung ML, He QY, Li B. Genome-wide CRISPR/Cas9 screening identifies a targetable MEST-PURA interaction in cancer metastasis. EBioMedicine 2023; 92:104587. [PMID: 37149929 DOI: 10.1016/j.ebiom.2023.104587] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Metastasis is one of the most lethal hallmarks of esophageal squamous cell carcinoma (ESCC), yet the mechanisms remain unclear due to a lack of reliable experimental models and systematic identification of key drivers. There is urgent need to develop useful therapies for this lethal disease. METHODS A genome-wide CRISPR/Cas9 screening, in combination with gene profiling of highly invasive and metastatic ESCC sublines, as well as PDX models, was performed to identify key regulators of cancer metastasis. The Gain- and loss-of-function experiments were taken to examine gene function. Protein interactome, RNA-seq, and whole genome methylation sequencing were used to investigate gene regulation and molecular mechanisms. Clinical significance was analyzed in tumor tissue microarray and TCGA databases. Homology modeling, modified ELISA, surface plasmon resonance and functional assays were performed to identify lead compound which targets MEST to suppress cancer metastasis. FINDINGS High MEST expression was associated with poor patient survival and promoted cancer invasion and metastasis in ESCC. Mechanistically, MEST activates SRCIN1/RASAL1-ERK-snail signaling by interacting with PURA. miR-449a was identified as a direct regulator of MEST, and hypermethylation of its promoter led to MEST upregulation, whereas systemically delivered miR-449a mimic could suppress tumor metastasis without overt toxicity. Furthermore, molecular docking and computational screening in a small-molecule library of 1,500,000 compounds and functional assays showed that G699-0288 targets the MEST-PURA interaction and significantly inhibits cancer metastasis. INTERPRETATION We identified the MEST-PURA-SRCIN1/RASAL1-ERK-snail signaling cascade as an important mechanism underlying cancer metastasis. Blockade of MEST-PURA interaction has therapeutic potential in management of cancer metastasis. FUNDING This work was supported by National Key Research and Development Program of China (2021YFC2501000, 2021YFC2501900, 2017YFA0505100); National Natural Science Foundation of China (31961160727, 82073196, 81973339, 81803551); NSFC/RGC Joint Research Scheme (N_HKU727/19); Natural Science Foundation of Guangdong Province (2021A1515011158, 2021A0505030035); Key Laboratory of Guangdong Higher Education Institutes of China (2021KSYS009).
Collapse
Affiliation(s)
- Wen Wen Xu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes and Key Laboratory of Protein Modification and Degradation, The Fifth Affiliated Hospital of Guangzhou Medical University and School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Long Liao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes and Key Laboratory of Protein Modification and Degradation, The Fifth Affiliated Hospital of Guangzhou Medical University and School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wei Dai
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Can-Can Zheng
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes and Key Laboratory of Protein Modification and Degradation, The Fifth Affiliated Hospital of Guangzhou Medical University and School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiang-Peng Tan
- Research Center of Cancer Diagnosis and Therapy, and Department of Clinical Oncology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yan He
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes and Key Laboratory of Protein Modification and Degradation, The Fifth Affiliated Hospital of Guangzhou Medical University and School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qi-Hua Zhang
- MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhi-Hao Huang
- MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wen-You Chen
- Department of Thoracic Surgery, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yan-Ru Qin
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Department of Clinical Oncology, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Kui-Sheng Chen
- Henan Province Key Laboratory of Tumor Pathology, Department of Pathology, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Simon Law
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Maria Li Lung
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Bin Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes and Key Laboratory of Protein Modification and Degradation, The Fifth Affiliated Hospital of Guangzhou Medical University and School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; MOE Key Laboratory of Tumor Molecular Biology, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| |
Collapse
|
4
|
Ginsenoside Rh2 suppresses colon cancer growth by targeting the miR-150-3p/SRCIN1/Wnt axis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:633-648. [PMID: 36916297 DOI: 10.3724/abbs.2023032] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Ginsenoside Rh2, which is extracted from ginseng, exerts antitumor activity. Recent studies suggest that Rh2 may suppress the growth of colon cancer (CC) in vitro. However, the underlying mechanism remains unclear. In this study, we identified the relative levels of miR-150-3p in CC tissues and cells by a comprehensive strategy of data mining, computational biology, and real-time reverse transcription PCR (qRT-PCR) experiments. The regulatory effects of miR-150-3p/SRCIN1 on the proliferative and invasive abilities of CC cells are evaluated by CCK-8, EdU, wound healing, and transwell assays. Cell cycle- and apoptosis-related protein levels are assessed by western blot analysis. An in vivo tumor formation assay was conducted to explore the effects of miR-150-3p on tumor growth. Furthermore, bioinformatics and dual luciferase reporter assays are applied to determine the functional binding of miRNA to mRNA of the target gene. Finally, the relationship between Rh2 and miR-150-3p was further verified in SW620 and HCT-116 cells. miR-150-3p is downregulated in CC tissues and cell lines. Functional assays indicate that the upregulation of miR-150-3p inhibits tumor growth both in vivo and in vitro. In addition, SRCIN1 is upregulated in CC and predicts a poor prognosis, and it is the direct target for miR-150-3p. Moreover, the miR-150-3p mimic decreases Topflash/Fopflash-dependent luciferase activity, resulting in the inhibition of Wnt pathway activity. Rh2 can suppress the growth of CC by increasing miR-150-3p expression. Rh2 alleviates the accelerating effect on Wnt pathway activity, cell proliferation/migration, and colony formation caused by miR-150-3p inhibition. Rh2 inhibits the miR-150-3p/SRCIN1/Wnt axis to suppress colon cancer growth.
Collapse
|
5
|
miR-657 Targets SRCIN1 via the Slug Pathway to Promote NSCLC Tumor Growth and EMT Induction. DISEASE MARKERS 2022; 2022:4842454. [PMID: 36033827 PMCID: PMC9402383 DOI: 10.1155/2022/4842454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/06/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022]
Abstract
Background MicroRNA- (miR-) 657 has been shown to regulate immunological and inflammatory activity, and it has also been defined to be dysregulated in both non-small-cell lung cancer (NSCLC) and hepatocellular carcinoma. The mechanistic role whereby miR-657 influences NSCLC progression, however, has yet to be clarified. Methods miR-657 and SRCIN1 expression levels were assessed via qPCR in the cell lines and tissues of NSCLC. Besides, correlations between the levels of miR-657 and NSCLC patient pathological characteristics were examined, and the Kaplan-Meier approach was employed for the evaluation of the prognostic utility of miR-657 in these patients. Moreover, the Pearson correlation analyses and dual-luciferase reporter assessments were used for detecting interactive relationships between miR-657 and SRCIN1. In addition, CCK-8, EdU, and Transwell assessments were employed for the appraisal of the ability of miR-657/SRCIN1 to regulate NSCLC cell proliferation and invasion. Western blotting was employed for the assessment of the levels of NSCLC cell proteins associated with the epithelial-mesenchymal transition (EMT) that were influenced by miR-657. The nude mice xenograft tumor model is established to observe the effect of miR-657 on NSCLC growth in vivo. Results NSCLC patient tissues and cell lines exhibited upregulated miR-657 expression that was closely related to tumor differentiation, lymphoid metastasis, and TNM stage. High levels of miR-657 were predictive of a poorer NSCLC patient prognosis, and overexpressing miR-657 resulted in the more rapid growth of NCI-H1650 and A549 cells, with a concomitant increase in their invasion. In addition, miR-657 overexpression raised the levels of Slug, N-cadherin, and Vimentin in these two cell lines while promoting E-cadherin downregulation. Dual-luciferase reporter assays confirmed that miR-657 was capable of binding to the SRCIN1 gene, and SRCIN1 expression levels were negatively associated with those of miR-657, indicating that it acts as a negative regulator of this gene. Knocking down SRCIN1 was capable to reverse the influences of miR-657 inhibitor treatment on NSCLC cell behavior. Finally, in vivo studies showed that miR-657 promoted NSCLC cell growth. Conclusion The obtained findings illuminate that miR-657 can promote the growth of tumors and the induction of the EMT in NSCLC cells by targeting SRCIN1 expression and modulating Slug pathway activation, highlighting this pathway as a promising therapeutic target in cases suffering from NSCLC.
Collapse
|
6
|
Huang T, Meng F, Huang H, Wang L, Wang L, Liu Y, Liu Y, Wang J, Li W, Zhang J, Liu Y. GALNT8 suppresses breast cancer cell metastasis potential by regulating EGFR O-GalNAcylation. Biochem Biophys Res Commun 2022; 601:16-23. [PMID: 35220009 DOI: 10.1016/j.bbrc.2022.02.072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/18/2022] [Indexed: 12/26/2022]
Abstract
Breast cancer represents the most lethal malignancy that threatens the health of females. Metastasis is the fatal hallmark of breast cancer, and current effective therapeutic targets of metastasis are still lacking. Aberrant O-GalNAcylation, which is attributed to alteration of polypeptide N-acetylgalactosaminyl transferases (GALNTs), has been implicated in cancer metastasis. However, GALNTs that drive metastasis in breast cancer and their underlying mechanisms are largely unclear. In the present study, a negative correlation between GALNT8 and the prognosis of breast cancer patients was observed in multiple groups of Gene Expression Omnibus (GEO) datasets. We then constructed a stable GALNT8 knockdown MCF7 cell line and performed transcriptome analysis using RNA sequencing, which revealed that the expression of multiple migration-related genes was changed. GALNT8 was identified as a regulator of epithelial-mesenchymal transition (EMT) markers, including E-cadherin, N-cadherin, ZO-1 and vimentin. Moreover, loss- and gain-of-function GALNT8 assays demonstrated that this glycosyltransferase inhibited the metastatic potential of breast cancer cells. Interestingly, the O-GalNAcylation of EGFR, which is the key factor related to the metastasis cascade, was impacted by GALNT8. Furthermore, our results suggested that the GALNT8-mediated O-GalNAcylation led to the suppression of the EGFR signaling pathway and metastatic potential in breast cancer cells. These results suggested that GALNT8 acts as a tumor suppressor, represses tumor metastasis and inhibits the EMT process through the EGFR signaling pathway. This finding may provide insight into the mechanism by which aberrant O-glycosylation modulates breast cancer metastasis.
Collapse
Affiliation(s)
- Tianmiao Huang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Fanxu Meng
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Huang Huang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Liping Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Lingyan Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Yangzhi Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Yajie Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Jie Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Wenli Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China
| | - Jianing Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China.
| | - Yubo Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 122406, China.
| |
Collapse
|
7
|
Centonze G, Natalini D, Salemme V, Costamagna A, Cabodi S, Defilippi P. p130Cas/ BCAR1 and p140Cap/ SRCIN1 Adaptors: The Yin Yang in Breast Cancer? Front Cell Dev Biol 2021; 9:729093. [PMID: 34708040 PMCID: PMC8542790 DOI: 10.3389/fcell.2021.729093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
p130Cas/BCAR1 is an adaptor protein devoid of any enzymatic or transcriptional activity, whose modular structure with various binding motifs, allows the formation of multi-protein signaling complexes. This results in the induction and/or maintenance of signaling pathways with pleiotropic effects on cell motility, cell adhesion, cytoskeleton remodeling, invasion, survival, and proliferation. Deregulation of p130Cas/BCAR1 adaptor protein has been extensively demonstrated in a variety of human cancers in which overexpression of p130Cas/BCAR1 correlates with increased malignancy. p140Cap (p130Cas associated protein), encoded by the SRCIN1 gene, has been discovered by affinity chromatography and mass spectrometry analysis of putative interactors of p130Cas. It came out that p140Cap associates with p130Cas not directly but through its interaction with the Src Kinase. p140Cap is highly expressed in neurons and to a lesser extent in epithelial tissues such as the mammary gland. Strikingly, in vivo and in vitro analysis identified its tumor suppressive role in breast cancer and in neuroblastoma, showing an inverse correlation between p140Cap expression in tumors and tumor progression. In this review, a synopsis of 15 years of research on the role of p130Cas/BCAR1 and p140Cap/SRCIN1 in breast cancer will be presented.
Collapse
Affiliation(s)
- Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Andrea Costamagna
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| |
Collapse
|
8
|
Sun J, Li H, Lv C, Draz E, Liu Y, Lin Z, Hu W, Mo K, Lin J, Xu W, Wang S. Trps1 targets Ccnd1 to regulate mouse Leydig cell proliferation. Andrology 2021; 9:1923-1933. [PMID: 34185441 DOI: 10.1111/andr.13072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND The tricho-rhino-phalangeal syndrome-1 gene (Trps1) is an atypical GATA family member. Although current studies of Trps1 mainly focus on tumors, whether Trps1 plays a role in the male reproductive system remains unknown. OBJECTIVES The purpose of this study was to elucidate the function of Trps1 in Leydig cells, indicating its regulatory mechanism on the cell cycle. METHODS Gene-silencing technology, RNA-seq, RT-qPCR, and western blotting were used to evaluate the function of Trps1 in mouse primary Leydig cells and MLTC-1 cells. In addition, ChIP-base sets and ChIP-qPCR were employed to further assess the regulatory mechanism of Trps1 in MLTC-1 cells. RESULTS Knockdown of Trps1 in Leydig cells significantly suppressed phosphorylation of Src and Akt and expression of Ccnd1, which was accompanied by impairment of cell proliferative ability. Trps1 may affect the cell cycle through the Src/Akt/Ccnd1 signaling pathway. In addition, Trps1 may bind to the promoter of Srcin1 to regulate its transcription, thus influencing Src phosphorylation levels and the proliferation of Leydig cells. DISCUSSION AND CONCLUSION Src increases in Leydig cells during pubertal development, suggesting its functional involvement in differentiated adult Leydig cells. Inhibition of the Src/Akt pathway would reduce Ccnd1 expression. In the present study, we found that Trps1 may regulate the phosphorylation level of Src and Akt through Srcin1, targeting Ccnd1 to influence mouse Leydig cell proliferation. These findings shed light on the regulation of Trps1 on cell proliferation and differentiation of mouse Leydig cells.
Collapse
Affiliation(s)
- Jiandong Sun
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Hua Li
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China.,Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P. R. China
| | - Chengyu Lv
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Eman Draz
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China.,Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P. R. China
| | - Yue Liu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China.,Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P. R. China
| | - Zihang Lin
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Weitao Hu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Kaien Mo
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Jianmin Lin
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Weiwei Xu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China
| | - Shie Wang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, P. R. China.,Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P. R. China
| |
Collapse
|
9
|
Winiarczyk D, Winiarczyk M, Winiarczyk S, Michalak K, Adaszek Ł. Proteomic Analysis of Tear Film Obtained from Diabetic Dogs. Animals (Basel) 2020; 10:ani10122416. [PMID: 33348610 PMCID: PMC7766195 DOI: 10.3390/ani10122416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Canine diabetes is a serious disease, which can lead to severe complications, eventually even death. Currently, all the diagnostic procedures are the invasive ones, with blood collection remaining as a golden standard for both initial diagnosis, and later follow-up. Tears can be obtained in a non-invasive manner, which makes them a perfect candidate for a screening tool in canine diabetes. In this study we aimed to analyze the protein composition of the tears collected from the healthy animals and compared it to the diabetic group. There are significant differences between these two groups, and we believe that the identified proteins hold promise as a potential diagnostic tool, which can be later on used both in clinical practice, and for better understanding of the disease. Abstract Canine diabetes mellitus is a significant health burden, followed with numerous systemic complications, including diabetic cataracts and retinopathy, leading to blindness. Diabetes should be considered as a disease damaging all the body organs, including gastrointestinal tract, through a complex combination of vascular and metabolic pathologies, leading to impaired gut function. Tear film can be obtained in a non-invasive way, which makes it a feasible biomarker source. In this study we compared proteomic changes ongoing in tear film of diabetic dogs. The study group consisted of 15 diabetic dogs, and 13 dogs served as a control group. After obtaining tear film with Schirmer strips, we performed 2-dimensional electrophoresis, followed by Delta2D software analysis, which allowed to select statistically significant differentially expressed proteins. After their identification with MALDI-TOF (matrix assisted laser desorption and ionisation time of flight) spectrometry we found one up-regulated protein in tear film of diabetic dogs—SRC kinase signaling inhibitor 1 (SRCIN1). Eight proteins were down-regulated: phosphatidylinositol-4 kinase type 2 alpha (PI4KIIα), Pro-melanin concentrating hormone (Pro-MCH), Flotillin-1, Protein mono-ADP ribosyltransferase, GRIP and coiled coil domain containing protein 2, tetratricopeptide repeat protein 36, serpin, and Prelamin A/C. Identified proteins were analyzed by Panther Gene Ontology software, and their possible connections with diabetic etiopathology were discussed. We believe that this is the first study to target tear film proteome in canine diabetes. We believe that combined with traditional examination, the tear film proteomic analysis can be a new source of biomarkers both for clinical practice, and experimental research.
Collapse
Affiliation(s)
- Dagmara Winiarczyk
- Department of Internal Diseases of Small Animals, University of Life Sciences of Lublin, 20-950 Lublin, Poland;
| | - Mateusz Winiarczyk
- Department of Vitreoretinal Surgery, Medical University of Lublin, 20-950 Lublin, Poland;
| | - Stanisław Winiarczyk
- Department of Epizootiology, University of Life Sciences of Lublin, 20-950 Lublin, Poland; (S.W.); (K.M.)
| | - Katarzyna Michalak
- Department of Epizootiology, University of Life Sciences of Lublin, 20-950 Lublin, Poland; (S.W.); (K.M.)
| | - Łukasz Adaszek
- Department of Epizootiology, University of Life Sciences of Lublin, 20-950 Lublin, Poland; (S.W.); (K.M.)
- Correspondence:
| |
Collapse
|
10
|
Salemme V, Angelini C, Chapelle J, Centonze G, Natalini D, Morellato A, Taverna D, Turco E, Ala U, Defilippi P. The p140Cap adaptor protein as a molecular hub to block cancer aggressiveness. Cell Mol Life Sci 2020; 78:1355-1367. [PMID: 33079227 PMCID: PMC7904710 DOI: 10.1007/s00018-020-03666-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/03/2020] [Accepted: 10/05/2020] [Indexed: 01/03/2023]
Abstract
The p140Cap adaptor protein is a scaffold molecule encoded by the SRCIN1 gene, which is physiologically expressed in several epithelial tissues and in the neurons. However, p140Cap is also strongly expressed in a significant subset of cancers including breast cancer and neuroblastoma. Notably, cancer patients with high p140Cap expression in their primary tumors have a lower probability of developing a distant event and ERBB2-positive breast cancer sufferers show better survival. In neuroblastoma patients, SRCIN1 mRNA levels represent an independent risk factor, which is inversely correlated to disease aggressiveness. Consistent with clinical data, SRCIN1 gain or loss of function mouse models demonstrated that p140Cap may affect tumor growth and metastasis formation by controlling the signaling pathways involved in tumorigenesis and metastatic features. This study reviews data showing the relevance of SRCIN1/p140Cap in cancer patients, the impact of SRCIN1 status on p140Cap expression, the specific mechanisms through which p140Cap can limit cancer progression, the molecular functions regulated by p140Cap, along with the p140Cap interactome, to unveil its key role for patient stratification in clinics.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Jennifer Chapelle
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, Università degli Studi di Torino, Largo Paolo Braccini 2, 10095, Grugliasco, TO, Italy.
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Science, Università degli Studi di Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
11
|
Jin W. Regulation of Src Family Kinases during Colorectal Cancer Development and Its Clinical Implications. Cancers (Basel) 2020; 12:cancers12051339. [PMID: 32456226 PMCID: PMC7281431 DOI: 10.3390/cancers12051339] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022] Open
Abstract
Src family kinases (SFKs) are non-receptor kinases that play a critical role in the pathogenesis of colorectal cancer (CRC). The expression and activity of SFKs are upregulated in patients with CRC. Activation of SFKs promotes CRC cell proliferation, metastases to other organs and chemoresistance, as well as the formation of cancer stem cells (CSCs). The enhanced expression level of Src is associated with decreased survival in patients with CRC. Src-mediated regulation of CRC progression involves various membrane receptors, modulators, and suppressors, which regulate Src activation and its downstream targets through various mechanisms. This review provides an overview of the current understanding of the correlations between Src and CRC progression, with a special focus on cancer cell proliferation, invasion, metastasis and chemoresistance, and formation of CSCs. Additionally, this review discusses preclinical and clinical strategies to improve the therapeutic efficacy of drugs targeting Src for treating patients with CRC.
Collapse
Affiliation(s)
- Wook Jin
- Laboratory of Molecular Disease and Cell Regulation, Department of Biochemistry, School of Medicine, Gachon University, Incheon 406-840, Korea
| |
Collapse
|
12
|
Russo I, Gavello D, Menna E, Vandael D, Veglia C, Morello N, Corradini I, Focchi E, Alfieri A, Angelini C, Bianchi FT, Morellato A, Marcantoni A, Sassoè-Pognetto M, Ottaviani MM, Yekhlef L, Giustetto M, Taverna S, Carabelli V, Matteoli M, Carbone E, Turco E, Defilippi P. p140Cap Regulates GABAergic Synaptogenesis and Development of Hippocampal Inhibitory Circuits. Cereb Cortex 2020; 29:91-105. [PMID: 29161354 DOI: 10.1093/cercor/bhx306] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/23/2017] [Indexed: 01/19/2023] Open
Abstract
The neuronal scaffold protein p140Cap was investigated during hippocampal network formation. p140Cap is present in presynaptic GABAergic terminals and its genetic depletion results in a marked alteration of inhibitory synaptic activity. p140Cap-/- cultured neurons display higher frequency of miniature inhibitory postsynaptic currents (mIPSCs) with no changes of their mean amplitude. Consistent with a potential presynaptic alteration of basal GABA release, p140Cap-/- neurons exhibit a larger synaptic vesicle readily releasable pool, without any variation of single GABAA receptor unitary currents and number of postsynaptic channels. Furthermore, p140Cap-/- neurons show a premature and enhanced network synchronization and appear more susceptible to 4-aminopyridine-induced seizures in vitro and to kainate-induced seizures in vivo. The hippocampus of p140Cap-/- mice showed a significant increase in the number of both inhibitory synapses and of parvalbumin- and somatostatin-expressing interneurons. Specific deletion of p140Cap in forebrain interneurons resulted in increased susceptibility to in vitro epileptic events and increased inhibitory synaptogenesis, comparable to those observed in p140Cap-/- mice. Altogether, our data demonstrate that p140Cap finely tunes inhibitory synaptogenesis and GABAergic neurotransmission, thus regulating the establishment and maintenance of the proper hippocampal excitatory/inhibitory balance.
Collapse
Affiliation(s)
- Isabella Russo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Daniela Gavello
- Department of Drug Science, University of Torino, Torino, Italy.,NIS Centre of Excellence, Torino, Italy
| | - Elisabetta Menna
- Institute of Neuroscience, CNR, Milano, Italy.,Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | - David Vandael
- Department of Drug Science, University of Torino, Torino, Italy.,NIS Centre of Excellence, Torino, Italy
| | - Carola Veglia
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Noemi Morello
- Department of Neuroscience, University of Torino, Torino, Italy
| | - Irene Corradini
- Institute of Neuroscience, CNR, Milano, Italy.,Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | | | - Annalisa Alfieri
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federico Tommaso Bianchi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Andrea Marcantoni
- Department of Drug Science, University of Torino, Torino, Italy.,NIS Centre of Excellence, Torino, Italy
| | - Marco Sassoè-Pognetto
- Department of Neuroscience, University of Torino, Torino, Italy.,National Institute of Neuroscience-Italy, Torino, Italy
| | | | - Latefa Yekhlef
- Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Maurizio Giustetto
- Department of Neuroscience, University of Torino, Torino, Italy.,National Institute of Neuroscience-Italy, Torino, Italy
| | - Stefano Taverna
- Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Valentina Carabelli
- Department of Drug Science, University of Torino, Torino, Italy.,NIS Centre of Excellence, Torino, Italy
| | - Michela Matteoli
- Institute of Neuroscience, CNR, Milano, Italy.,Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | - Emilio Carbone
- Department of Drug Science, University of Torino, Torino, Italy.,NIS Centre of Excellence, Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
13
|
Dong J, Li B, Lin D, Lu D, Liu C, Lu X, Tang X, Li L, Zhu D, Liu J, Qiu X, Tian L, Zhou Q. LincRNA00494 Suppresses Non-small Cell Lung Cancer Cell Proliferation by Regulating SRCIN1 Expression as a ceRNA. Front Oncol 2020; 10:79. [PMID: 32117734 PMCID: PMC7016015 DOI: 10.3389/fonc.2020.00079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 02/05/2023] Open
Abstract
Background: Lung cancer is the most common malignant tumor worldwide. Accumulating results have shown that long non-coding RNAs (lncRNAs) play a key role in tumorigenesis. Patients and Methods: A total of 163 tumor tissues were collected from non-small cell lung cancer (NSCLC) patients from West China Hospital of Sichuan University. LincRNA00494 is a novel lncRNA, and its expression and biological effect in NSCLC were reported in this study. NSCLC cell lines were used in this study. Results: LincRNA00494 is mainly distributed in the cytoplasm. LincRNA00494 was downregulated in the tumor tissues compared with the adjacent non-tumor tissues. LincRNA00494 expression was positively correlated with SRCIN1 expression (R = 0.57, P < 0.05). Silencing of LincRNA00494 in the cell lines substantially decreased SRCIN1 expression at the mRNA and protein levels, whereas overexpression of LincRNA00494 enhanced the SRCIN1 levels. miR-150-3p significantly decreased the luciferase signals of LincRNA00494 and SRCIN1 reporters. After transfection with miR-150-3p mimics and miR-150-3p inhibitor, overexpression of LincRNA00494 decreased the proliferation of the H358 (36%) and H1299 (29%) cell lines compared with that of the control cells, as shown by CCK-8 assays, whereas silencing LincRNA00494 promoted the proliferation of the H358 (47%) and H1299 (35%) cells. Tumor growth from LincRNA00494-overexpressing xenografts was significantly decreased; additionally, LincRNA00494 silencing substantially increased tumor growth compared with that of the control cells. Conclusions: Functional experiments revealed that LincRNA00494 inhibited NSCLC cell proliferation, which might be related to the suppression of SRCIN1, a tumor suppressor gene, by acting as a decoy for miR-150-3p. The data showed that LincRNA00494 might have antineoplastic effects during NSCLC tumorigenesis through its role as a ceRNA.
Collapse
Affiliation(s)
- Jingsi Dong
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bingjie Li
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Lin
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Lu
- Department of Otorhinolaryngology, Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | - Xingbing Lu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojun Tang
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Li
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Daxing Zhu
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiewei Liu
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoming Qiu
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Long Tian
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qinghua Zhou
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Chapelle J, Sorokina O, McLean C, Salemme V, Alfieri A, Angelini C, Morellato A, Adrait A, Menna E, Matteoli M, Couté Y, Ala U, Turco E, Defilippi P, Armstrong JD. Dissecting the Shared and Context-Dependent Pathways Mediated by the p140Cap Adaptor Protein in Cancer and in Neurons. Front Cell Dev Biol 2019; 7:222. [PMID: 31681758 PMCID: PMC6803390 DOI: 10.3389/fcell.2019.00222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/19/2019] [Indexed: 12/26/2022] Open
Abstract
The p140Cap adaptor protein is a scaffold molecule physiologically expressed in few epithelial tissues, such as the mammary gland, and in differentiated neurons. While the role of p140Cap in mammary gland epithelia is not still understood, we already know that a significant subset of breast cancers express p140Cap. In the subgroup of ERBB2-amplified breast cancers, a high p140Cap status predicts a significantly lower probability of developing a distant event and a clear difference in survival. p140Cap is causal in dampening ERBB2-positive tumor cell progression, impairing tumor onset and growth, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. Since only a few p140Cap interacting proteins have been identified in breast cancer and the molecular complexes and pathways underlying the cancer function of p140Cap are largely unknown, we generated a p140Cap interactome from ERBB2-positive breast cancer cells, identifying cancer specific components and those shared with the synaptic interactome. We identified 373 interacting proteins in cancer cells, including those with functions relevant to cell adhesion, protein homeostasis, regulation of cell cycle and apoptosis, which are frequently deregulated in cancer. Within the interactome, we identified 15 communities (clusters) with topology-functional relationships. In neurons, where p140Cap is key in regulating synaptogenesis, synaptic transmission and synaptic plasticity, it establishes an extensive interactome with proteins that cluster to sub complexes located in the postsynaptic density. p140Cap interactors converge on key synaptic processes, including synaptic transmission, actin cytoskeleton remodeling and cell-cell junction organization. Comparing the breast cancer to the synaptic interactome, we found 39 overlapping proteins, a relatively small overlap. However, cell adhesion and remodeling of actin cytoskeleton clearly emerge as common terms in the shared subset. Thus, the functional signature of the two interactomes is primarily determined by organ/tissue and functional specificity, while the overlap provides a list of shared functional terms, which might be linked to both cancer and neurological functions.
Collapse
Affiliation(s)
- Jennifer Chapelle
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Oksana Sorokina
- Simons Initiative for the Developing Brain, School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| | - Colin McLean
- Simons Initiative for the Developing Brain, School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Annalisa Alfieri
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Annie Adrait
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, BGE, Grenoble, France
| | - Elisabetta Menna
- Institute of Neuroscience, CNR, Milan, Italy
- Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | - Michela Matteoli
- Institute of Neuroscience, CNR, Milan, Italy
- Istituto Clinico Humanitas, IRCCS, Rozzano, Italy
| | - Yohann Couté
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, BGE, Grenoble, France
| | - Ugo Ala
- Department of Veterinary Sciences, Università degli Studi di Torino, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Turin, Italy
| | - J. Douglas Armstrong
- Simons Initiative for the Developing Brain, School of Informatics, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Grasso S, Cangelosi D, Chapelle J, Alzona M, Centonze G, Lamolinara A, Salemme V, Angelini C, Morellato A, Saglietto A, Bianchi FT, Cabodi S, Salaroglio IC, Fusella F, Ognibene M, Iezzi M, Pezzolo A, Poli V, Di Cunto F, Eva A, Riganti C, Varesio L, Turco E, Defilippi P. The SRCIN1/p140Cap adaptor protein negatively regulates the aggressiveness of neuroblastoma. Cell Death Differ 2019; 27:790-807. [PMID: 31285546 PMCID: PMC7205889 DOI: 10.1038/s41418-019-0386-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/21/2019] [Accepted: 06/21/2019] [Indexed: 01/01/2023] Open
Abstract
Neuroblastoma is the most common extra-cranial pediatric solid tumor, responsible for 13–15% of pediatric cancer death. Its intrinsic heterogeneity makes it difficult to target for successful therapy. The adaptor protein p140Cap/SRCIN1 negatively regulates tumor cell features and limits breast cancer progression. This study wish to assess if p140Cap is a key biological determinant of neuroblastoma outcome. RNAseq profiles of a large cohort of neuroblastoma patients show that SRCIN1 mRNA levels are an independent risk factor inversely correlated to disease aggressiveness. In high-risk patients, CGH+SNP microarray analysis of primary neuroblastoma identifies SRCIN1 as frequently altered by hemizygous deletion, copy-neutral loss of heterozygosity, or disruption. Functional experiments show that p140Cap negatively regulates Src and STAT3 signaling, affects anchorage-independent growth and migration, in vivo tumor growth and spontaneous lung metastasis formation. p140Cap also increases sensitivity of neuroblastoma cells to doxorubicin and etoposide treatment, as well as to a combined treatment with chemotherapy drugs and Src inhibitors. Our functional findings point to a causal role of p140Cap in curbing the aggressiveness of neuroblastoma, due to its ability to impinge on specific molecular pathways, and to sensitize cells to therapeutic treatment. This study provides the first evidence that the SRCIN1/p140Cap adaptor protein is a key player in neuroblastoma as a new independent prognostic marker for patient outcome and treatment. Altogether, these data highlight the potential clinical impact of SRCIN1/p140Cap expression in neuroblastoma tumors, in terms of reducing cytotoxic effects of chemotherapy, one of the main issues for pediatric tumor treatment.
Collapse
Affiliation(s)
- Silvia Grasso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Davide Cangelosi
- Laboratory of Molecular Biology, Giannina Gaslini Institute, 16147, Genova, Italy
| | - Jennifer Chapelle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Melissa Alzona
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Alessia Lamolinara
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Andrea Saglietto
- Cardiology Division, Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Federico Tommaso Bianchi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy.,Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano (TO), Italy
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Iris Chiara Salaroglio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy.,Department of Oncology, University of Torino, 10126, Torino, Italy
| | - Federica Fusella
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Marzia Ognibene
- Laboratorio Cellule Staminali Post Natali e Terapie Cellulari, Istituto Giannina Gaslini, Genova, Italy
| | - Manuela Iezzi
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, Italy
| | - Annalisa Pezzolo
- Laboratorio Cellule Staminali Post Natali e Terapie Cellulari, Istituto Giannina Gaslini, Genova, Italy
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043, Orbassano (TO), Italy
| | - Alessandra Eva
- Laboratory of Molecular Biology, Giannina Gaslini Institute, 16147, Genova, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126, Torino, Italy
| | - Luigi Varesio
- Laboratory of Molecular Biology, Giannina Gaslini Institute, 16147, Genova, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126, Torino, Italy.
| |
Collapse
|
16
|
Wu W, He L, Huang Y, Hou L, Zhang W, Zhang L, Wu C. MicroRNA-510 Plays Oncogenic Roles in Non-Small Cell Lung Cancer by Directly Targeting SRC Kinase Signaling Inhibitor 1. Oncol Res 2019; 27:879-887. [PMID: 30982489 PMCID: PMC7848405 DOI: 10.3727/096504018x15451308507747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
An increasing number of studies have demonstrated that microRNAs (miRNAs) may play key roles in various cancer carcinogenesis and progression, including non-small cell lung cancer (NSCLC). However, the expressions, roles, and mechanisms of miR-510 in NSCLC have, up to now, been largely undefined. In vivo assay showed that miR-510 was upregulated in NSCLC tissues compared with that in adjacent nontumor lung tissues. miR-510 expression was significantly correlated with TNM stage and lymph node metastasis. In vitro assay indicated that expressions of miR-510 were also increased in NSCLC cell lines. Downregulation of miR-510 suppressed NSCLC cell proliferation and invasion in vitro. We identified SRC kinase signaling inhibitor 1 (SRCIN1) as a direct target gene of miR-510 in NSCLC. Expression of SRCIN1 was downregulated in lung cancer cells and negatively correlated with miR-510 expression in tumor tissues. Downregulation of SRCIN1, leading to inhibition of miR-510 expression, reversed cell proliferation and invasion in NSCLC cells. These results showed that miR-510 acted as an oncogenic miRNA in NSCLC, partly by targeting SRCIN1, suggesting that miR-510 can be a potential approach for the treatment of patients with malignant lung cancer.
Collapse
Affiliation(s)
- Wei Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Linyan He
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Yan Huang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Wei Zhang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Liping Zhang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
17
|
Madarampalli B, Watts GFM, Panipinto PM, Nguygen HN, Brenner MB, Noss EH. Interactions between cadherin-11 and platelet-derived growth factor receptor-alpha signaling link cell adhesion and proliferation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1516-1524. [PMID: 30876808 DOI: 10.1016/j.bbadis.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 02/25/2019] [Accepted: 03/11/2019] [Indexed: 12/26/2022]
Abstract
Cadherins are homophilic cell-to-cell adhesion molecules that help cells respond to environmental changes. Newly formed cadherin junctions are associated with increased cell phosphorylation, but the pathways driving this signaling response are largely unknown. Since cadherins have no intrinsic signaling activity, this phosphorylation must occur through interactions with other signaling molecules. We previously reported that cadherin-11 engagement activates joint synovial fibroblasts, promoting inflammatory and degradative pathways important in rheumatoid arthritis (RA) pathogenesis. Our objective in this study was to discover interacting partners that mediate cadherin-11 signaling. Protein array screening showed that cadherin-11 extracellular binding domains linked to an Fc domain (cad11Fc) induced platelet-derived growth factor (PDGFR)-α phosphorylation in synovial fibroblasts and glioblastoma cells. PDGFRs are growth factor receptor tyrosine kinases that promote cell proliferation, survival, and migration in mesodermally derived cells. Increased PDGFR activity is implicated in RA pathology and associates with poor prognosis in several cancers, including sarcoma and glioblastoma. PDGFRα activation by cadherin-11 signaling promoted fibroblast proliferation, a signaling pathway independent from cadherin-11-stimulated IL-6 or matrix metalloproteinase (MMP)-3 release. PDGFRα phosphorylation mediated most of the cad11Fc-induced phosphatidyl-3-kinase (PI3K)/Akt activation, but only part of the mitogen-activated protein kinase (MAPK) response. PDGFRα-dependent signaling did not require cell cadherin-11 expression. Rather, cad11Fc immunoprecipitated PDGFRα, indicating a direct interaction between cadherin-11 and PDGFRα extracellular domains. This study is the first to report an interaction between cadherin-11 and PDGFRα and adds to our growing understanding that cadherin-growth factor receptor interactions help balance the interplay between tissue growth and adhesion.
Collapse
Affiliation(s)
- Bhanupriya Madarampalli
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98019, USA.
| | - Gerald F M Watts
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Paul M Panipinto
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98019, USA.
| | - Hung N Nguygen
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Michael B Brenner
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| | - Erika H Noss
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98019, USA; Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Lu Q, Guo Z, Qian H. Role of microRNA-150-5p/SRCIN1 axis in the progression of breast cancer. Exp Ther Med 2019; 17:2221-2229. [PMID: 30867707 PMCID: PMC6396020 DOI: 10.3892/etm.2019.7206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022] Open
Abstract
In China, breast cancer is the most commonly occurring cancer in women. MicroRNAs (miRs) are a group of endogenous small non-coding RNAs, which serve a role in many biological processes through the regulation of target genes. In the current study, miR-150-5p expression was significantly up-regulated in breast cancer tissues and cell lines. To investigate the cellular function and underlying molecular mechanism of miR-150-5p in breast cancer, TargetScan7.2 was used to identify miR-150-5p target genes. SRC kinase signaling inhibitor 1 (SRCIN1) was identified as a direct target gene of miR-150-5p and the current study demonstrated that SRCIN1 was negatively regulated by miR-150-5p in breast cancer cells. Furthermore, SRCIN1 expression was significantly down-regulated in breast cancer tissues and cell lines. Taken together, these results demonstrated that there was a negative association between miR-150-5p and SRCIN1 in breast cancer. The CCK-8 and Transwell assays were used to examine breast cancer cell viability, invasion and migration ability. The current study demonstrated that over-expression of miR-150-5p enhanced breast cancer cell proliferation, invasion and migration. In addition, miR-150-5p over-expression increased the expression of mesenchymal cell markers (vimentin, N-cadherin and β-catenin) and decreased the expression of epithelial cell markers (E-cadherin and zonula occludens-1). By contrast, miR-150-5p knockdown inhibited breast cancer cell viability, invasion and migration. Additionally, miR-150-5p knockdown decreased the expression of mesenchymal cell markers and increased the expression of epithelial cell markers. Taken together, these results suggest that the miR-150-5p/SRCIN1 axis may be a potential target in the treatment of breast cancer.
Collapse
Affiliation(s)
- Qingfu Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Zhaoji Guo
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Haixin Qian
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
19
|
Zhu J, Han S. miR-150-5p promotes the proliferation and epithelial-mesenchymal transition of cervical carcinoma cells via targeting SRCIN1. Pathol Res Pract 2019; 215:738-747. [PMID: 30679084 DOI: 10.1016/j.prp.2019.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/14/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022]
Abstract
Cervical carcinoma is one of the most universal cancers among women. Recent researches have reported that microRNA-150-5p (miR-150-5p) is up-regulated in diverse carcinomas containing cervical carcinoma. The purpose of this study was to further investigate the potential role of miR-150-5p in the progress of cervical carcinoma cells including proliferation and epithelial-mesenchymal transition (EMT).The ability of miR-150-5p to promote carcinogenesis was analyzed using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot assays, respectively. Bioinformatics analyses predicted and identified whether SRC kinase signaling inhibitor 1 (SRCIN1) was served as a potential target of miR-150-5p. C-33A and HeLa cells were utilized to determine the function of miR-150-5p through targeting SRCIN1. Among the aberrantly expressed miRNAs, miR-150-5p was significantly revealed differential expression in cervical carcinoma cell lines and was closely relevant to cell growth regulation. Furthermore, we found that SRCIN1 overexpression could obviously inhibit the proliferation and EMT of cervical cancer cells triggered by miR-150-5p mimics as well as accelerated the apoptosis of cervical carcinoma cells. In conclusion, our data demonstrated that miR-150-5p could promote the proliferation and EMT of cervical carcinoma cells via targeting SRCIN1. Thus, miR-150-5p may hold a promise as a prognostic biomarker and potential therapeutic target for cervical carcinoma.
Collapse
Affiliation(s)
- Jinming Zhu
- Department of Oncology, Affiliated Zhongshan Hospital, Dalian University, Dalian, China
| | - Shichao Han
- Department of Gynecology, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
20
|
Murine and Human Mammary Cancer Cell Lines: Functional Tests. Methods Mol Biol 2018. [PMID: 29959713 DOI: 10.1007/978-1-4939-8600-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The biological characterization of mammary cancer cells is a prerequisite that helps the scientist understand some aspect of tumor biology. Once isolated from the tumor, cells are subjected to multiple tests that dissect their ability to growth, migrate, degrade the surrounding stroma, produce 3-dimensional structures and differentiate. Targeted inhibitors, when added to these tests, are used to unravel how specific growth factors, receptors, and intracellular translational pathways promote the ability of mammary tumor cells to achieve their biological behavior. Herein we describe a set of techniques used to put in focus the biological capacities in mammary cancer cells. When the characterization of a biological trait (e.g., proliferation) is assessable by multiple assays, we will limit the description to only one technique, possibly the easier to manage and that requires minimal laboratory equipment.
Collapse
|
21
|
Chen R, Liao JY, Huang J, Chen WL, Ma XJ, Luo XD. [ARTICLE WITHDRAWN] Downregulation of SRC Kinase Signaling Inhibitor 1 (SRCIN1) Expression by MicroRNA-32 Promotes Proliferation and Epithelial-Mesenchymal Transition in Human Liver Cancer Cells. Oncol Res 2018; 26:573-579. [PMID: 28550679 PMCID: PMC7844739 DOI: 10.3727/096504017x14954923820137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
THIS ARTICLE WAS WITHDRAWN BY THE PUBLISHER IN NOVEMBER 2020.
Collapse
Affiliation(s)
- Ren Chen
- Department of Infectious Diseases, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, P.R. China
| | - Jin-Yao Liao
- Department of Infectious Diseases, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, P.R. China
| | - Jing Huang
- Department of Infectious Diseases, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, P.R. China
| | - Wen-Li Chen
- Department of Infectious Diseases, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, P.R. China
| | - Xiao-Jun Ma
- Department of Infectious Diseases, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, P.R. China
| | - Xiao-Dan Luo
- Department of Infectious Diseases, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, P.R. China
| |
Collapse
|
22
|
Sun W, Wang X, Li J, You C, Lu P, Feng H, Kong Y, Zhang H, Liu Y, Jiao R, Chen X, Ba Y. MicroRNA-181a promotes angiogenesis in colorectal cancer by targeting SRCIN1 to promote the SRC/VEGF signaling pathway. Cell Death Dis 2018; 9:438. [PMID: 29739921 PMCID: PMC5941226 DOI: 10.1038/s41419-018-0490-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 03/03/2018] [Accepted: 03/16/2018] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is a very common metastatic tumor with active angiogenesis that requires active angiogenesis. Recently, increased microRNA-181a-5p (miR-181a) expression was found to be significantly associated with liver metastasis and poor outcome in CRC patients. In this study, the role of miR-181a in tumor angiogenesis was further investigated. Capillary tube formation assays were used to demonstrate the ability of miR-181a to promote tumor angiogenesis. Bioinformatics analyses identified SRC kinase signaling inhibitor 1 (SRCIN1) as a potential target of miR-181a. Next, two CRC cell lines (HT29 and SW480) were used to clarify the function of miR-181a through SRCIN1 targeting. In addition, the biological effects of SRCIN1 inhibition by miR-181a were examined in vitro by quantitative RT-PCR, western blotting and enzyme-linked immunosorbent assay and in vivo by Matrigel plug angiogenesis assays and immunohistochemical staining. In clinical samples, Fluorescence in situ hybridization and immunofluorescence were performed to detect the relation between miR-181a and SRCIN1. In addition, SRCIN1 protein and miR-181a expression levels in CRC tissues were also measured by western blot and quantitative real-time polymerase chain reaction. MiR-181a markedly augmented the capability of CRC cells to advance tube formation in endothelial cells in vitro. The Matrigel plug assay showed that miR-181a promoted angiogenesis in vivo. In conclusion, miR-181a inhibited SRCIN1, which caused SRC to transform from an inactive status to an active conformation and to trigger vascular endothelial growth factor secretion, leading to increased angiogenesis. MiR-181a dysregulation contributes to angiogenesis in CRC, and downregulation of miR-181a represents a promising, novel strategy to achieve an efficient antiangiogenic response in anti-CRC therapy.
Collapse
Affiliation(s)
- Wu Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, 210009, China
| | - Jialu Li
- Department of Gastroenterology, Tianjin First Center Hospital, 24 Fukang Road, Tianjin, 300192, China
| | - Chaoying You
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Pan Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046, China
| | - Huijin Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046, China
| | - Yan Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046, China
| | - Haiyang Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanqing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046, China
| | - Ruihua Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046, China.
| | - Xi Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS), School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046, China.
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
23
|
Zhang L, Lin J, Ye Y, Oba T, Gentile E, Lian J, Wang J, Zhao Y, Gu J, Wistuba II, Roth JA, Ji L, Wu X. Serum MicroRNA-150 Predicts Prognosis for Early-Stage Non-Small Cell Lung Cancer and Promotes Tumor Cell Proliferation by Targeting Tumor Suppressor Gene SRCIN1. Clin Pharmacol Ther 2017; 103:1061-1073. [PMID: 28891208 DOI: 10.1002/cpt.870] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/23/2017] [Accepted: 08/30/2017] [Indexed: 12/14/2022]
Abstract
This integrative multistage study was aimed to identify circulating microRNAs (miRNAs) as prognostic biomarkers and investigate the treatment target for early-stage non-small cell lung cancer (NSCLC) patients. In stage I-II NSCLC patients, we screened and validated the miRNA ratio signatures predictive of prognosis in serum. In tumor, we found that the expression of miR-150 in identified miRNA signatures was also associated with survival. Increased miR-150 expression promoted NSCLC cell proliferation and migration and vice versa. Specific mRNA cleavage sites targeted by endogenous miR-150 in 3' untranslated region (UTR) of SRCIN1 was identified by utilizing our recently developed novel Stem-Loop-Array reverse-transcription polymerase chain reaction (SLA-RT-PCR) assay. The blocking action of miR-150 resulted in repressed NSCLC cell growth in vitro and knockdown of miR-150 caused substantial tumor volume reduction in vivo. Our findings suggest that miR-150 binding on specific recognition sites in 3' UTR of tumor suppressor gene SRCIN1 present a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Liren Zhang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Lin
- Department of Thoracic & Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuanqing Ye
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Taro Oba
- Department of Thoracic & Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emanuela Gentile
- Department of Thoracic & Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jie Lian
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Wang
- Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yang Zhao
- Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jian Gu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jack A Roth
- Department of Thoracic & Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lin Ji
- Department of Thoracic & Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xifeng Wu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
24
|
Zhang M, Ma F, Xie R, Wu Y, Wu M, Zhang P, Peng Y, Zhao J, Xiong J, Li A, Kequan C, Zhang Y, Liu S, Wang J, Chen X. Overexpression of Srcin1 contributes to the growth and metastasis of colorectal cancer. Int J Oncol 2017; 50:1555-1566. [PMID: 28393242 PMCID: PMC5403293 DOI: 10.3892/ijo.2017.3952] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 03/13/2017] [Indexed: 11/10/2022] Open
Abstract
The adaptor protein Srcin1 is a novel Src-binding protein that regulates Src activation through C-terminal Src kinase (Csk). Srcin1 behaves as a tumour suppressor in breast cancer, but the role of Srcin1 in the development of colorectal cancer (CRC) remains unknown. In the present study, Srcin1 expression in normal tissue was examined by tissue microarray and assessed by immunohistochemistry in 10 patients. In addition, the biological impact of Srcin1 knockdown on CRC cells was investigated in vitro and in vivo. The results showed that Srcin1 was expressed in different types of normal human tissues, whereas its expression was increased in human CRC tissues. Srcin1 expression also correlated with tumour progression. The suppression of Srcin1 induced cell differentiation and G0/G1 cell cycle arrest. Furthermore, Srcin1 increased cell growth as well as the capacity of migration and invasion in CRC cells. Srcin1 induced the activation of the Wnt/β-catenin signalling pathway. Moreover, Srcin1 suppression sensitized cancer cells to 5-fluorouracil (5-FU)-induced apoptosis in vitro and in vivo. Together, these results demonstrate that Srcin1 contributes to CRC carcinogenesis, invasion and metastasis. These findings provide a rationale for a mechanistic approach to CRC treatment based on the development of Srcin1-targeted therapies.
Collapse
Affiliation(s)
- Mengnan Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Feng Ma
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ruyi Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Meiyan Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Pei Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ying Peng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jinjun Zhao
- Department of Rheumatism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jing Xiong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Cheng Kequan
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Yali Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xueqing Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
25
|
Grasso S, Chapelle J, Salemme V, Aramu S, Russo I, Vitale N, Verdun di Cantogno L, Dallaglio K, Castellano I, Amici A, Centonze G, Sharma N, Lunardi S, Cabodi S, Cavallo F, Lamolinara A, Stramucci L, Moiso E, Provero P, Albini A, Sapino A, Staaf J, Di Fiore PP, Bertalot G, Pece S, Tosoni D, Confalonieri S, Iezzi M, Di Stefano P, Turco E, Defilippi P. The scaffold protein p140Cap limits ERBB2-mediated breast cancer progression interfering with Rac GTPase-controlled circuitries. Nat Commun 2017; 8:14797. [PMID: 28300085 PMCID: PMC5357316 DOI: 10.1038/ncomms14797] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 01/27/2017] [Indexed: 12/29/2022] Open
Abstract
The docking protein p140Cap negatively regulates tumour cell features. Its relevance on breast cancer patient survival, as well as its ability to counteract relevant cancer signalling pathways, are not fully understood. Here we report that in patients with ERBB2-amplified breast cancer, a p140Cap-positive status associates with a significantly lower probability of developing a distant event, and a clear difference in survival. p140Cap dampens ERBB2-positive tumour cell progression, impairing tumour onset and growth in the NeuT mouse model, and counteracting epithelial mesenchymal transition, resulting in decreased metastasis formation. One major mechanism is the ability of p140Cap to interfere with ERBB2-dependent activation of Rac GTPase-controlled circuitries. Our findings point to a specific role of p140Cap in curbing the aggressiveness of ERBB2-amplified breast cancers and suggest that, due to its ability to impinge on specific molecular pathways, p140Cap may represent a predictive biomarker of response to targeted anti-ERBB2 therapies. p140Cap adaptor proteins interfere with adhesion and growth factor-dependent signalling in cancer cells but the mechanisms are unclear. Here the authors show that p140Cap interferes with ERBB2-dependent activation of Rac GTPase-controlled circuitries reducing metastasis and cancer progression.
Collapse
Affiliation(s)
- Silvia Grasso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Jennifer Chapelle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Simona Aramu
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Isabella Russo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | | | - Katiuscia Dallaglio
- Research Infrastructure, IRCCS Arcispedale Santa Maria Nuova, 42100 Reggio Emilia, Italy
| | | | - Augusto Amici
- Department of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Nanaocha Sharma
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Serena Lunardi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Alessia Lamolinara
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, 66100 Chieti, Italy
| | - Lorenzo Stramucci
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, 66100 Chieti, Italy
| | - Enrico Moiso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Adriana Albini
- Scientific and Technology Pole, IRCCS MultiMedica, 20100 Milan, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy
| | - Johan Staaf
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund 22100, Sweden
| | - Pier Paolo Di Fiore
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, 20100 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20100 Milan, Italy
| | - Giovanni Bertalot
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy
| | - Salvatore Pece
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20100 Milan, Italy
| | - Daniela Tosoni
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy
| | - Stefano Confalonieri
- Molecular Medicine Program, European Institute of Oncology, 20100 Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, 20100 Milan, Italy
| | - Manuela Iezzi
- Department of Medicine and Aging Science, Center of Excellence on Aging and Translational Medicine (CeSi-Met), G. D'Annunzio University, Chieti-Pescara, 66100 Chieti, Italy
| | - Paola Di Stefano
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
26
|
Yang Y, Ding L, Guo ZK, Zheng XL, Wang LS, Sun HY, Jin ZG, Wang HX. The epigenetically-regulated miR-34a targeting c-SRC suppresses RAF/MEK/ERK signaling pathway in K-562 cells. Leuk Res 2017; 55:91-96. [PMID: 28157629 DOI: 10.1016/j.leukres.2017.01.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/05/2017] [Accepted: 01/13/2017] [Indexed: 11/17/2022]
Abstract
Previous reports show that miR-34a suppressed K-562 cell proliferation and contributed to megakaryocytic differentiation of K-562 cells. Here, we reported that miR-34a, a tumor suppressor gene, is down-regulated in the K-562 cells and chronic myeloid leukemia (CML) patients due to aberrant DNA hypermethylation. c-SRC is a target of miR-34a. Restoring miR-34a expression resulted in down-regulation of c-SRC and phosphorylated (Tyr416) c-SRC protein in K-562 cells, which consequently triggered suppression of the RAF/MEK/ERK signaling pathway to decrease cell proliferation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Hematology, Chinese PLA Air Force General Hospital, 30 Fucheng Road, Beijing 100042, China
| | - Li Ding
- Department of Hematology, Chinese PLA Air Force General Hospital, 30 Fucheng Road, Beijing 100042, China
| | - Zi-Kuan Guo
- Beijing Institute of Radiation Medicine, Chinese PLA Academy of Military Medical Science, Beijing 100850, China
| | - Xiao-Li Zheng
- Department of Hematology, Chinese PLA Air Force General Hospital, 30 Fucheng Road, Beijing 100042, China
| | - Li-Sheng Wang
- Beijing Institute of Radiation Medicine, Chinese PLA Academy of Military Medical Science, Beijing 100850, China
| | - Hui-Yan Sun
- Beijing Institute of Radiation Medicine, Chinese PLA Academy of Military Medical Science, Beijing 100850, China
| | - Zhan-Guo Jin
- Aerospace Balance Medical Center, Chinese PLA Air Force General Hospital, Beijing 100042, China
| | - Heng-Xiang Wang
- Department of Hematology, Chinese PLA Air Force General Hospital, 30 Fucheng Road, Beijing 100042, China.
| |
Collapse
|
27
|
Yang F, Luo LJ, Zhang L, Wang DD, Yang SJ, Ding L, Li J, Chen D, Ma R, Wu JZ, Tang JH. MiR-346 promotes the biological function of breast cancer cells by targeting SRCIN1 and reduces chemosensitivity to docetaxel. Gene 2016; 600:21-28. [PMID: 27913185 DOI: 10.1016/j.gene.2016.11.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/08/2016] [Accepted: 11/24/2016] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) are a class of highly conserved small noncoding RNAs that play pivotal roles at the post-transcriptional level in the biological function of various cancers, including breast cancer. In our study, miR-346 mimic, inhibitor, negative control or si-SRCIN1 were transfected into MCF-7 and MCF-7/Doc cells, respectively. Quantitative real time PCR (qRT-PCR) was used to measure miR-346 and SRCIN1 mRNA expressions and western blot was used to detect the expression of SRCIN1 in protein level. CCK-8 and colony formation were employed to verify cell viability and proliferation. Flow cytometry showed the apoptosis. Transwell was performed to detect migration and invasion. The luciferase reporter assay data showed the target correlation of miR-346 and SRCIN1. Firstly, we found that the expression of miR-346 was higher in breast cancer tissues than in their paired corresponding non-cancerous tissues and there was significant inversed correlation between miR-346 and SRCIN1. Overexpression of miR-346 promoted cell proliferation, colony formation, migration and invasion, and reduced apoptosis, sensitivity to Docetaxel (Doc). SRCIN1 was identified as a direct target of miR-346, whose silencing promoted cell proliferation and the IC50 of Doc. Moreover, SRCIN1 silencing reduced the effect of miR-346 down-expression. Taken together, miR-346 may function as an oncogenic miRNA and mediate chemosensitivity to docetaxel through targeting SRCIN1 in breast cancer, targeted modulation of miR-346 expression may became a potential strategy for the treatment.
Collapse
Affiliation(s)
- Fan Yang
- Xuzhou Medical University, Xuzhou, Xuzhou 221004, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China.
| | - Long-Ji Luo
- Xuzhou Medical University, Xuzhou, Xuzhou 221004, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Lei Zhang
- Xuzhou Medical University, Xuzhou, Xuzhou 221004, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Dan-Dan Wang
- Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China; The First Clinical School of Nanjing Medical University, Nanjing 210009, China
| | - Su-Jin Yang
- Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China; The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China
| | - Li Ding
- China Pharmaceutical University, Nanjing 21009, China
| | - Jian Li
- Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China; The Forth Clinical School of Nanjing Medical University, Nanjing 210009, China
| | - Dan Chen
- Research Center of Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Rong Ma
- Research Center of Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Jian-Zhong Wu
- Research Center of Clinical Oncology, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China
| | - Jin-Hai Tang
- Xuzhou Medical University, Xuzhou, Xuzhou 221004, China; Department of General Surgery, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Baiziting 42, Nanjing 210009, China; Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
28
|
Wang P, Wang H, Li X, Liu Y, Zhao C, Zhu D. SRCIN1 Suppressed Osteosarcoma Cell Proliferation and Invasion. PLoS One 2016; 11:e0155518. [PMID: 27513473 PMCID: PMC4981393 DOI: 10.1371/journal.pone.0155518] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/30/2016] [Indexed: 02/05/2023] Open
Abstract
SRCIN1 (SRC kinase signalling inhibitor 1) is a new tumor suppressor gene. Previous studies showed that SRCIN1 played a tumor suppressor role in the development of lung cancer and breast cancer. However, the role of SRCIN1 in osteosarcoma is still unknown. In this study, we demonstrated that SRCIN1 was downregulated in osteosarcoma cell lines compared with osteoblastic cell line. Moreover, SRCIN1 was downregulated in osteosarcoma tissues compared with the adjacent tissues. Further investigation revealed that overexpression of SRCIN1 inhibited the osteosarcoma cell line MG-63 proliferation. This effect was confirmed by measuring the ki-67 and PCNA expression. SRCIN1 overexpression promoted E-cadherin expression and suppressed N-cadherin, Vimentin and Snail expression, suggesting that SRCIN1 overexpression inhibited EMT of the osteosarcoma cell. In addition, ectopic expression of SRCIN1 inhibited the MG-63 cell colony formation and invasion. These data suggested that SRCIN1 acted as a tumor suppressor gene in the development of osteosarcoma.
Collapse
Affiliation(s)
- Peng Wang
- Department of Orthopedics, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Hu Wang
- Department of Orthopedics, First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Xiaotao Li
- Department of Orthopedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, 154000, China
| | - Ying Liu
- Clinical medicine Grade Four, Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Chengbin Zhao
- Department of Orthopedics, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
- * E-mail: (CZ); (DZ)
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University Daqing, Daqing, 163319, China
- Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin, 150081, China
- * E-mail: (CZ); (DZ)
| |
Collapse
|
29
|
Ye L, Wang H, Liu B. miR-211 promotes non-small cell lung cancer proliferation by targeting SRCIN1. Tumour Biol 2015; 37:1151-7. [PMID: 26277787 DOI: 10.1007/s13277-015-3835-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that, when dysregulated, are involved in the initiation and progression of various cancers, including lung cancer, in humans. In the current study, qRT-PCR was performed to measure miR-211 expression in human non-small cell lung cancer (NSCLC) cell lines and tissues. Cell proliferation, cell cycle, colony formation, and invasion were performed to detect the functional role of miR-211 in human NSCLC cell line. We used luciferase reporter assay to find the potential target of miR-211. We found that miR-211 expression was upregulated in human non-small cell lung cancer (NSCLC) cell lines and tissues. The overexpression of miR-211 enhanced NSCLC cell proliferation, colony formation, and invasion. SRC kinase signaling inhibitor 1 (SRCIN1) was identified as a direct target of miR-211. SRCIN1 silencing promoted cell proliferation, and SRCIN1 expression was downregulated in human NSCLC cell lines. Thus, miR-211 may function as an oncogenic miRNA in NSCLC, partly by regulating SRCIN1, and the modulation of miR-211 expression represents a potential strategy for the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Leiguang Ye
- Department of Pulmonary Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China, 150040
| | - Hui Wang
- Department of Pulmonary Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China, 150040
| | - Baogang Liu
- Department of Pulmonary Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China, 150040.
| |
Collapse
|
30
|
Xu X, Wang W, Su N, Zhu X, Yao J, Gao W, Hu Z, Sun Y. miR-374a promotes cell proliferation, migration and invasion by targeting SRCIN1 in gastric cancer. FEBS Lett 2014; 589:407-13. [PMID: 25554419 DOI: 10.1016/j.febslet.2014.12.027] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 12/11/2014] [Accepted: 12/18/2014] [Indexed: 12/25/2022]
Abstract
MicroRNAs (miRNAs) play a prominent role in gastric cancer (GC) initiation and progression. In this study, we found that miR-374a expression was up-regulated in human GC cell lines and tissues. Inhibition of miR-374a suppressed GC cell proliferation, migration and invasion in vitro and slowed tumor growth in vivo. SRC kinase signaling inhibitor 1 (SRCIN1) was identified as a direct target of miR-374a. Silencing of SRCIN1 significantly enhanced cell proliferation, migration and invasion, whereas SRCIN1 reintroduction partially abrogated the oncogenic effects of miR-374a. Taken together, these findings suggest that miR-374a functions as a candidate oncogene in GC by directly targeting SRCIN1. miR-374a may therefore be useful as a promising therapeutic target for malignant GC.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Weijun Wang
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Ning Su
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Xujun Zhu
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Jun Yao
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Wenchao Gao
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Zhiqian Hu
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Yanping Sun
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China.
| |
Collapse
|
31
|
Xiao X, Chen B, Liu X, Liu P, Zheng G, Ye F, Tang H, Xie X. Diallyl disulfide suppresses SRC/Ras/ERK signaling-mediated proliferation and metastasis in human breast cancer by up-regulating miR-34a. PLoS One 2014; 9:e112720. [PMID: 25396727 PMCID: PMC4232521 DOI: 10.1371/journal.pone.0112720] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/14/2014] [Indexed: 12/20/2022] Open
Abstract
Diallyl disulfide (DADS) is one of the major volatile components of garlic oil. DADS has various biological properties, including anticancer, antiangiogenic, and antioxidant effects. However, the anticancer mechanisms of DADS in human breast cancer have not been elucidated, particularly in vivo. In this study, we demonstrated that the expression of miR-34a was up-regulated in DADS-treated MDA-MB-231 cells. miR-34a not only inhibited breast cancer growth but also enhanced the antitumor effect of DADS, both in vitro and in vivo. Furthermore, Src was identified as a target of miR-34a, with miR-34a inhibiting SRC expression and consequently triggering the suppression of the SRC/Ras/ERK pathway. These results suggest that DADS could be a promising anticancer agent for breast cancer. miR-34a may also demonstrate a potential gene therapy agent that could enhance the antitumor effects of DADS.
Collapse
Affiliation(s)
- Xiangsheng Xiao
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Bo Chen
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xiaoping Liu
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Peng Liu
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Guopei Zheng
- Affiliated Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Feng Ye
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Hailin Tang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
32
|
Wang N, Liang H, Zhou Y, Wang C, Zhang S, Pan Y, Wang Y, Yan X, Zhang J, Zhang CY, Zen K, Li D, Chen X. miR-203 suppresses the proliferation and migration and promotes the apoptosis of lung cancer cells by targeting SRC. PLoS One 2014; 9:e105570. [PMID: 25140799 PMCID: PMC4139332 DOI: 10.1371/journal.pone.0105570] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/21/2014] [Indexed: 12/19/2022] Open
Abstract
SRC, also known as proto-oncogene c-Src, is a non-receptor tyrosine kinase that plays an important role in cancer progression by promoting survival, angiogenesis, proliferation, and invasion pathways. In this study, we found that SRC protein levels were consistently upregulated in lung cancer tissues, but that SRC mRNA levels varied randomly, suggesting that a post-transcriptional mechanism was involved in SRC regulation. Because microRNAs (miRNAs) are powerful post-transcriptional regulators of gene expression, we used bioinformatic analyses to search for miRNAs that potentially target SRC. We identified specific targeting sites for miR-203 in the 3'-untranslated region (3'-UTR) of SRC. We then experimentally validated miR-203 as a direct regulator of SRC using cell transfection and luciferase assays and showed that miR-203 inhibited SRC expression and consequently triggered suppression of the SRC/Ras/ERK pathway. Finally, we demonstrated that the repression of SRC by miR-203 suppressed the proliferation and migration and promoted the apoptosis of lung cancer cells. In summary, this study provides the first clues regarding the role of miR-203 as a tumor suppressor in lung cancer cells through the inhibition of SRC translation.
Collapse
Affiliation(s)
- Nan Wang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Hongwei Liang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Yong Zhou
- Department of Thoracic and Cardiovascular surgery, Affiliated Gulou Hospital, Medical college of Nanjing University, Nanjing, Jiangsu, China
| | - Chen Wang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Suyang Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Yi Pan
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Yanbo Wang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Xin Yan
- The Comprehensive Cancer Center of Drum Tower Hospital affiliated to Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, China
| | - Junfeng Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Chen-Yu Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Ke Zen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Donghai Li
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Xi Chen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
p140Cap regulates memory and synaptic plasticity through Src-mediated and citron-N-mediated actin reorganization. J Neurosci 2014; 34:1542-53. [PMID: 24453341 DOI: 10.1523/jneurosci.2341-13.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A major challenge in the neuroscience field is the identification of molecules and pathways that control synaptic plasticity and memory. Dendritic spines play a pivotal role in these processes, as the major sites of excitatory synapses in neuronal communication. Previous studies have shown that the scaffold protein p140Cap localizes into dendritic spines and that its knockdown negatively modulates spine shape in culture. However, so far, there is no information on its in vivo relevance. By using a knock-out mouse model, we here demonstrate that p140Cap is a key element for both learning and synaptic plasticity. Indeed, p140Cap(-/-) mice are impaired in object recognition test, as well as in LTP and in LTD measurements. The in vivo effects of p140Cap loss are presumably attenuated by noncell-autonomous events, since primary neurons obtained from p140Cap(-/-) mice show a strong reduction in number of mushroom spines and abnormal organization of synapse-associated F-actin. These phenotypes are most likely caused by a local reduction of the inhibitory control of RhoA and of cortactin toward the actin-depolymerizing factor cofilin. These events can be controlled by p140Cap through its capability to directly inhibit the activation of Src kinase and by its binding to the scaffold protein Citron-N. Altogether, our results provide new insight into how protein associated with dynamic microtubules may regulate spine actin organization through interaction with postsynaptic density components.
Collapse
|
34
|
Cao M, Hou D, Liang H, Gong F, Wang Y, Yan X, Jiang X, Wang C, Zhang J, Zen K, Zhang CY, Chen X. miR-150 promotes the proliferation and migration of lung cancer cells by targeting SRC kinase signalling inhibitor 1. Eur J Cancer 2014; 50:1013-24. [PMID: 24456795 DOI: 10.1016/j.ejca.2013.12.024] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 10/30/2013] [Accepted: 12/27/2013] [Indexed: 11/15/2022]
Abstract
microRNAs (miRNAs) are a class of endogenously expressed, small non-coding RNAs that play an important role in the regulation of gene expression at the post-transcriptional level. Dysregulation of miRNAs is associated with a variety of diseases, including lung cancer. In the present study, miR-150 was found to be significantly upregulated in lung cancer clinical specimens by quantitative real-time polymerase chain reaction (RT-PCR). Using bioinformatics analysis, v-src avian sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (SRC) kinase signalling inhibitor 1 (SRCIN1), an important regulator of SRC activity, was predicted to be a potential target of miR-150. Furthermore, an inverse correlation between miR-150 and SRCIN1 protein levels, but not mRNA levels, was identified in human lung cancer tissue samples. By overexpressing or knocking down miR-150 in lung adenocarcinoma A549 cells and H1975 cells, it was experimentally validated that miR-150 is a direct regulator of SRCIN1. It was further confirmed that miR-150 directly recognises the 3'-untranslated region (3'-UTR) of SRCIN1 transcript with a luciferase reporter assay. Finally, it was demonstrated that the repression of SRCIN1 by miR-150 consequently triggered the activation of the Src/focal adhesion kinase (FAK) and Src/Ras/extracellular signal-regulated kinase (ERK) pathway, which eventually promoted the proliferation and migration of A549 cells, and this promotion by miR-150 could be reversed by overexpressing SRCIN1. Taken together, our findings provide the first clues regarding the role of miR-150 as an oncogene in lung cancer through the inhibition of SRCIN1 translation.
Collapse
Affiliation(s)
- Minghui Cao
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Dongxia Hou
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Hongwei Liang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Fei Gong
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Yilei Wang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Xin Yan
- Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Tianjin 300060, China
| | - Xiaohong Jiang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Chen Wang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Junfeng Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Ke Zen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China
| | - Chen-Yu Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China.
| | - Xi Chen
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
35
|
Carraro DM, Koike Folgueira MAA, Garcia Lisboa BC, Ribeiro Olivieri EH, Vitorino Krepischi AC, de Carvalho AF, de Carvalho Mota LD, Puga RD, do Socorro Maciel M, Michelli RAD, de Lyra EC, Grosso SHG, Soares FA, Achatz MIADSW, Brentani H, Moreira-Filho CA, Brentani MM. Comprehensive analysis of BRCA1, BRCA2 and TP53 germline mutation and tumor characterization: a portrait of early-onset breast cancer in Brazil. PLoS One 2013; 8:e57581. [PMID: 23469205 PMCID: PMC3586086 DOI: 10.1371/journal.pone.0057581] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/22/2013] [Indexed: 11/25/2022] Open
Abstract
Germline mutations in BRCA1, BRCA2 and TP53 genes have been identified as one of the most important disease-causing issues in young breast cancer patients worldwide. The specific defective biological processes that trigger germline mutation-associated and -negative tumors remain unclear. To delineate an initial portrait of Brazilian early-onset breast cancer, we performed an investigation combining both germline and tumor analysis. Germline screening of the BRCA1, BRCA2, CHEK2 (c.1100delC) and TP53 genes was performed in 54 unrelated patients <35 y; their tumors were investigated with respect to transcriptional and genomic profiles as well as hormonal receptors and HER2 expression/amplification. Germline mutations were detected in 12 out of 54 patients (22%) [7 in BRCA1 (13%), 4 in BRCA2 (7%) and one in TP53 (2%) gene]. A cancer familial history was present in 31.4% of the unrelated patients, from them 43.7% were carriers for germline mutation (37.5% in BRCA1 and in 6.2% in the BRCA2 genes). Fifty percent of the unrelated patients with hormone receptor-negative tumors carried BRCA1 mutations, percentage increasing to 83% in cases with familial history of cancer. Over-representation of DNA damage-, cellular and cell cycle-related processes was detected in the up-regulated genes of BRCA1/2-associated tumors, whereas cell and embryo development-related processes were over-represented in the up-regulated genes of BRCA1/2-negative tumors, suggesting distinct mechanisms driving the tumorigenesis. An initial portrait of the early-onset breast cancer patients in Brazil was generated pointing out that hormone receptor-negative tumors and positive familial history are two major risk factors for detection of a BRCA1 germline mutation. Additionally, the data revealed molecular factors that potentially trigger the tumor development in young patients.
Collapse
Affiliation(s)
- Dirce Maria Carraro
- Laboratory of Genomics and Molecular Biology, A.C. Camargo Hospital, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Repetto D, Aramu S, Boeri Erba E, Sharma N, Grasso S, Russo I, Jensen ON, Cabodi S, Turco E, Di Stefano P, Defilippi P. Mapping of p140Cap phosphorylation sites: the EPLYA and EGLYA motifs have a key role in tyrosine phosphorylation and Csk binding, and are substrates of the Abl kinase. PLoS One 2013; 8:e54931. [PMID: 23383002 PMCID: PMC3561454 DOI: 10.1371/journal.pone.0054931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 12/17/2012] [Indexed: 12/11/2022] Open
Abstract
Protein phosphorylation tightly regulates specific binding of effector proteins that control many diverse biological functions of cells (e. g. signaling, migration and proliferation). p140Cap is an adaptor protein, specifically expressed in brain, testis and epithelial cells, that undergoes phosphorylation and tunes its interactions with other regulatory molecules via post-translation modification. In this work, using mass spectrometry, we found that p140Cap is in vivo phosphorylated on tyrosine (Y) within the peptide GEGLpYADPYGLLHEGR (from now on referred to as EGLYA) as well as on three serine residues. Consistently, EGLYA has the highest score of in silico prediction of p140Cap phosphorylation. To further investigate the p140Cap function, we performed site specific mutagenesis on tyrosines inserted in EGLYA and EPLYA, a second sequence with the same highest score of phosphorylation. The mutant protein, in which both EPLYA/EGLYA tyrosines were converted to phenylalanine, was no longer tyrosine phosphorylated, despite the presence of other tyrosine residues in p140Cap sequence. Moreover, this mutant lost its ability to bind the C-terminal Src kinase (Csk), previously shown to interact with p140Cap by Far Western analysis. In addition, we found that in vitro and in HEK-293 cells, the Abelson kinase is the major kinase involved in p140Cap tyrosine phosphorylation on the EPLYA and EGLYA sequences. Overall, these data represent an original attempt to in vivo characterise phosphorylated residues of p140Cap. Elucidating the function of p140Cap will provide novel insights into its biological activity not only in normal cells, but also in tumors.
Collapse
Affiliation(s)
- Daniele Repetto
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | - Simona Aramu
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | | | - Nanaocha Sharma
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | - Silvia Grasso
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | - Isabella Russo
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | - Ole N. Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Sara Cabodi
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
| | - Paola Di Stefano
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
- * E-mail: (PD); (PDS)
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, Università degli Studi di Torino, Torino, Italy
- * E-mail: (PD); (PDS)
| |
Collapse
|
37
|
Prostaglandins in cancer cell adhesion, migration, and invasion. Int J Cell Biol 2012; 2012:723419. [PMID: 22505934 PMCID: PMC3299390 DOI: 10.1155/2012/723419] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/08/2011] [Indexed: 12/21/2022] Open
Abstract
Prostaglandins exert a profound influence over the adhesive, migratory, and invasive behavior of cells during the development and progression of cancer. Cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) are upregulated in inflammation and cancer. This results in the production of prostaglandin E2 (PGE2), which binds to and activates G-protein-coupled prostaglandin E1–4 receptors (EP1–4). Selectively targeting the COX-2/mPGES-1/PGE2/EP1–4 axis of the prostaglandin pathway can reduce the adhesion, migration, invasion, and angiogenesis. Once stimulated by prostaglandins, cadherin adhesive connections between epithelial or endothelial cells are lost. This enables cells to invade through the underlying basement membrane and extracellular matrix (ECM). Interactions with the ECM are mediated by cell surface integrins by “outside-in signaling” through Src and focal adhesion kinase (FAK) and/or “inside-out signaling” through talins and kindlins. Combining the use of COX-2/mPGES-1/PGE2/EP1–4 axis-targeted molecules with those targeting cell surface adhesion receptors or their downstream signaling molecules may enhance cancer therapy.
Collapse
|
38
|
Giron-Michel J, Azzi S, Khawam K, Mortier E, Caignard A, Devocelle A, Ferrini S, Croce M, François H, Lecru L, Charpentier B, Chouaib S, Azzarone B, Eid P. Interleukin-15 plays a central role in human kidney physiology and cancer through the γc signaling pathway. PLoS One 2012; 7:e31624. [PMID: 22363690 PMCID: PMC3283658 DOI: 10.1371/journal.pone.0031624] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 01/16/2012] [Indexed: 12/16/2022] Open
Abstract
The ability of Interleukin-15 (IL-15) to activate many immune antitumor mechanisms renders the cytokine a good candidate for the therapy of solid tumors, particularly renal cell carcinoma. Although IL-15 is being currently used in clinical trials, the function of the cytokine on kidney's components has not been extensively studied; we thus investigated the role of IL-15 on normal and tumor renal epithelial cells. Herein, we analyzed the expression and the biological functions of IL-15 in normal renal proximal tubuli (RPTEC) and in their neoplastic counterparts, the renal clear cell carcinomas (RCC). This study shows that RPTEC express a functional heterotrimeric IL-15Rαβγc complex whose stimulation with physiologic concentrations of rhIL-15 is sufficient to inhibit epithelial mesenchymal transition (EMT) commitment preserving E-cadherin expression. Indeed, IL-15 is not only a survival factor for epithelial cells, but it can also preserve the renal epithelial phenotype through the γc-signaling pathway, demonstrating that the cytokine possess a wide range of action in epithelial homeostasis. In contrast, in RCC in vitro and in vivo studies reveal a defect in the expression of γc-receptor and JAK3 associated kinase, which strongly impacts IL-15 signaling. Indeed, in the absence of the γc/JAK3 couple we demonstrate the assembly of an unprecedented functional high affinity IL-15Rαβ heterodimer, that in response to physiologic concentrations of IL-15, triggers an unbalanced signal causing the down-regulation of the tumor suppressor gene E-cadherin, favoring RCC EMT process. Remarkably, the rescue of IL-15/γc-dependent signaling (STAT5), by co-transfecting γc and JAK3 in RCC, inhibits EMT reversion. In conclusion, these data highlight the central role of IL-15 and γc-receptor signaling in renal homeostasis through the control of E-cadherin expression and preservation of epithelial phenotype both in RPTEC (up-regulation) and RCC (down-regulation).
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Sandy Azzi
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Krystel Khawam
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Erwan Mortier
- INSERM UMRS 892, Institut de Recherche Thérapeutique de l'Université de Nantes (IRT UN), Nantes, France
| | - Anne Caignard
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris, France
| | - Aurore Devocelle
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Silvano Ferrini
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Michela Croce
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Hélène François
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Lola Lecru
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Bernard Charpentier
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Salem Chouaib
- INSERM UMR 753, Université de Paris-Sud, Institut Gustave Roussy (IGR), Villejuif, France
| | - Bruno Azzarone
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| | - Pierre Eid
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| |
Collapse
|
39
|
Suppression of breast tumor growth and metastasis by an engineered transcription factor. PLoS One 2011; 6:e24595. [PMID: 21931769 PMCID: PMC3172243 DOI: 10.1371/journal.pone.0024595] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 08/15/2011] [Indexed: 11/19/2022] Open
Abstract
Maspin is a tumor and metastasis suppressor playing an essential role as gatekeeper of tumor progression. It is highly expressed in epithelial cells but is silenced in the onset of metastatic disease by epigenetic mechanisms. Reprogramming of Maspin epigenetic silencing offers a therapeutic potential to lock metastatic progression. Herein we have investigated the ability of the Artificial Transcription Factor 126 (ATF-126) designed to upregulate the Maspin promoter to inhibit tumor progression in pre-established breast tumors in immunodeficient mice. ATF-126 was transduced in the aggressive, mesenchymal-like and triple negative breast cancer line, MDA-MB-231. Induction of ATF expression in vivo by Doxycycline resulted in 50% reduction in tumor growth and totally abolished tumor cell colonization. Genome-wide transcriptional profiles of ATF-induced cells revealed a gene signature that was found over-represented in estrogen receptor positive (ER+) "Normal-like" intrinsic subtype of breast cancer and in poorly aggressive, ER+ luminal A breast cancer cell lines. The comparison transcriptional profiles of ATF-126 and Maspin cDNA defined an overlapping 19-gene signature, comprising novel targets downstream the Maspin signaling cascade. Our data suggest that Maspin up-regulates downstream tumor and metastasis suppressor genes that are silenced in breast cancers, and are normally expressed in the neural system, including CARNS1, SLC8A2 and DACT3. In addition, ATF-126 and Maspin cDNA induction led to the re-activation of tumor suppressive miRNAs also expressed in neural cells, such as miR-1 and miR-34, and to the down-regulation of potential oncogenic miRNAs, such as miR-10b, miR-124, and miR-363. As expected from its over-representation in ER+ tumors, the ATF-126-gene signature predicted favorable prognosis for breast cancer patients. Our results describe for the first time an ATF able to reduce tumor growth and metastatic colonization by epigenetic reactivation of a dormant, normal-like, and more differentiated gene program.
Collapse
|
40
|
p140Cap suppresses the invasive properties of highly metastatic MTLn3-EGFR cells via impaired cortactin phosphorylation. Oncogene 2011; 31:624-33. [DOI: 10.1038/onc.2011.257] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Bray I, Tivnan A, Bryan K, Foley NH, Watters KM, Tracey L, Davidoff AM, Stallings RL. MicroRNA-542-5p as a novel tumor suppressor in neuroblastoma. Cancer Lett 2011; 303:56-64. [PMID: 21310526 DOI: 10.1016/j.canlet.2011.01.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 01/05/2011] [Accepted: 01/19/2011] [Indexed: 01/31/2023]
Abstract
Several studies have implicated the dysregulation of microRNAs in neuroblastoma pathogenesis, an often fatal paediatric cancer arising from precursor cells of the sympathetic nervous system. Our group and others have demonstrated that lower expression of miR-542-5p is highly associated with poor patient survival, indicating a potential tumor suppressive function. Here, we demonstrate that ectopic over-expression of this miRNA decreases the invasive potential of neuroblastoma cell lines in vitro, along with primary tumor growth and metastases in an orthotopic mouse xenograft model, providing the first functional evidence for the involvement of miR-542-5p as a tumor suppressor in any type of cancer.
Collapse
Affiliation(s)
- Isabella Bray
- Department of Cancer Genetics, Royal College of Surgeons in Ireland, York House, York Street, Dublin 2, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
42
|
β1 integrin controls EGFR signaling and tumorigenic properties of lung cancer cells. Oncogene 2011; 30:4087-96. [DOI: 10.1038/onc.2011.107] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
43
|
Cabodi S, del Pilar Camacho-Leal M, Di Stefano P, Defilippi P. Integrin signalling adaptors: not only figurants in the cancer story. Nat Rev Cancer 2010; 10:858-70. [PMID: 21102636 DOI: 10.1038/nrc2967] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Current evidence highlights the ability of adaptor (or scaffold) proteins to create signalling platforms that drive cellular transformation upon integrin-dependent adhesion and growth factor receptor activation. The understanding of the biological effects that are regulated by these adaptors in tumours might be crucial for the identification of new targets and the development of innovative therapeutic strategies for human cancer. In this Review we discuss the relevance of adaptor proteins in signalling that originates from integrin-mediated cell-extracellular matrix (ECM) adhesion and growth factor stimulation in the context of cell transformation and tumour progression. We specifically underline the contribution of p130 Crk-associated substrate (p130CAS; also known as BCAR1), neural precursor cell expressed, developmentally down-regulated 9 (NEDD9; also known as HEF1), CRK and the integrin-linked kinase (ILK)-pinch-parvin (IPP) complex to cancer, along with the more recently identified p140 Cas-associated protein (p140CAP; also known as SRCIN1).
Collapse
Affiliation(s)
- Sara Cabodi
- Molecular Biotechnology Centre and Department of Genetics, Biology and Biochemistry, University of Torino, Via Nizza 52, Torino 10126, Italy
| | | | | | | |
Collapse
|