Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Cancer Lett ; 596: 217022, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38849014

ABSTRACT

We previously reported that extracellular matrix protein 1 isoform a (ECM1a) promotes epithelial ovarian cancer (EOC) through autocrine signaling by binding to cell surface receptors αXß2. However, the role of ECM1a as a secretory molecule in the tumor microenvironment is rarely reported. In this study, we constructed murine Ecm1-knockout mice and human ECM1a-knockin mice and further generated orthotopic or peritoneal xenograft tumor models to mimic the different metastatic stages of EOC. We show that ECM1a induces oncogenic metastasis of orthotopic xenograft tumors, but inhibits early-metastasis of peritoneal xenograft tumors. ECM1a remodels extracellular matrices (ECM) and promotes remote metastases by recruiting and transforming bone marrow mesenchymal stem cells (BMSCs) into platelet-derived growth factor receptor beta (PDGFRß+) cancer-associated fibroblasts (CAFs) and facilitating the secretion of angiopoietin-like protein 2 (ANGPTL2). Competing with ECM1a, ANGPTL2 also binds to integrin αX through the P1/P2 peptides, resulting in negative effects on BMSC differentiation. Collectively, this study reveals the dual functions of ECM1a in remodeling of TME during tumor progression, emphasizing the complexity of EOC phenotypic heterogeneity and metastasis.


Subject(s)
Angiopoietin-Like Protein 2 , Cancer-Associated Fibroblasts , Extracellular Matrix Proteins , Mice, Knockout , Ovarian Neoplasms , Tumor Microenvironment , Animals , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Humans , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Mice , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Angiopoietin-like Proteins/metabolism , Angiopoietin-like Proteins/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Neoplasm Metastasis
2.
Acta Histochem ; 126(1): 152133, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38266317

ABSTRACT

Osteoporosis (OP) is a common disease among older adults. The promotion of osteoblast differentiation plays a crucial role in alleviating OP symptoms. Extracellular matrix protein 1 (ECM1) has been reported to be closely associated with osteogenic differentiation. In this study, we constructed U2OS cell lines with ECM1 knockdown and ECM1a overexpression based on knockdown, and identified the target miRNA (miR-1260b) by sequencing. Overexpression of miR-1260b promoted the osteogenic differentiation of U2OS and MG63 cells, as demonstrated by increased alkaline phosphatase (ALP) activity, matrix mineralization, and Runt-Related Transcription Factor 2 (RUNX2), Osteopontin (OPN), Collagen I (COL1A1), and Osteocalcin (OCN) protein expressions, whereas low expression of miR-1260b had the opposite effect. In addition, miR-1260b expression was decreased in OP patients than in non-OP patients. Next, we predicted the target gene of miRNA through TargetScan and miRDB and found that miR-1260b negatively regulated GDP dissociation inhibitor 1 (GDI1) by directly binding to its 3'-untranslated region. Subsequent experiments revealed that GDI1 overexpression decreased ALP activity and calcium deposit, reduced RUNX2, OPN, COL1A1, and OCN expression levels, and reversed the effects of miR-1260b on osteogenic differentiation. In conclusion, ECM1-related miR-1260b promotes osteogenic differentiation by targeting GDI1 in U2OS and MG63 cells. Thus, this study has significant implication for osteoporosis treatment.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors , MicroRNAs , Osteoporosis , Humans , Aged , Osteogenesis/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Cells, Cultured , MicroRNAs/metabolism , Cell Differentiation/genetics , Osteoporosis/metabolism , Extracellular Matrix Proteins
3.
Nutrients ; 14(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36145166

ABSTRACT

Wentilactone A (WA) is a tetranorditerpenoid isolated from marine algae. We previously found that WA inhibited cancer cell proliferation with little toxicity. In this study, we show that high expression of extracellular matrix protein-1 (ECM1) promotes cancer cell cisplatin resistance, and the secreted ECM1 activates normal fibroblasts (NFs) to transform cells with characteristics of cancer-associated fibroblasts (CAFs). Transcription of the ECM1 gene is regulated largely by NF-κB through EP881C/T-EP266C binding sites. WA supresses the phosphorylation of NF-κB through inhibition of the upstream IKK/IκB phoshorylation to block the expression of ECM1, which reverses the cisplatin-induced activation of NF-κB/ECM1. On the contrary, cisplatin facilitates phosphorylation of NF-κB to enhance the expression of ECM1. These results highlight ECM1 as a potential target for treatment of cisplatin-resistant cancers associated with the ECM1 activated signaling. In addition, WA reverses cisplatin resistance by targeting both tumor cells and the tumor microenvironment through IKK/IκB/NF-κB signaling to reduce the expression of the ECM1 protein.


Subject(s)
NF-kappa B , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Cisplatin/pharmacology , Extracellular Matrix Proteins/metabolism , Female , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Ovarian Neoplasms/drug therapy , Phosphorylation , Tumor Microenvironment
4.
Cancer Biol Med ; 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34591414

ABSTRACT

OBJECTIVE: The aims of this study were to examine the prognostic value of SHP-1 in breast cancer, its roles in the regulation of breast cancer cell growth and metastasis, and the underlying mechanisms. METHODS: Tumor specimens from 160 patients with breast cancer and 160 noncancerous tissues were used to examine the expression of SHP-1 and to analyze its association with overall survival through Kaplan-Meier and multivariate Cox regression analyses. RNA sequencing data and the expression and clinical importance of SHP-1 in breast cancer were evaluated with data from The Cancer Genome Atlas. In vitro and in vivo assays were performed to elucidate the effects of SHP-1 on breast cancer cell proliferation and invasion. Confocal immunofluorescence and GST pulldown assays were used to demonstrate the interaction between SHP-1 and epidermal growth factor receptor, as well as its downstream pathways. Immunohistochemistry and The Cancer Genome Atlas database were used to investigate the clinical association between SHP-1 and EGFR in human breast cancer. RESULTS: SHP-1 expression was associated with better survival in patients with breast cancer, whereas SHP-1 expression was negatively correlated with EGFR in human breast cancer. Ectopic SHP-1 expression significantly suppressed breast cancer cell proliferation, migration, and invasion. SHP-1 knockdown induced a more invasive phenotype and accelerated cell growth. Mechanistically, EGFR, a protein directly interacting with SHP-1, mediates the SHP-1-induced inactivation of Ras/Erk/GSK3ß signaling and its downstream effectors. CONCLUSIONS: SHP-1 is an important prognostic biomarker in patients with breast cancer, and the SHP-1-EGFR axis is a promising target for treatment.

5.
Nat Commun ; 12(1): 4230, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244494

ABSTRACT

Extracellular matrix protein-1 (ECM1) promotes tumorigenesis in multiple organs but the mechanisms associated to ECM1 isoform subtypes have yet to be clarified. We report in this study that the secretory ECM1a isoform induces tumorigenesis through the GPR motif binding to integrin αXß2 and the activation of AKT/FAK/Rho/cytoskeleton signaling. The ATP binding cassette subfamily G member 1 (ABCG1) transduces the ECM1a-integrin αXß2 interactive signaling to facilitate the phosphorylation of AKT/FAK/Rho/cytoskeletal molecules and to confer cancer cell cisplatin resistance through up-regulation of the CD326-mediated cell stemness. On the contrary, the non-secretory ECM1b isoform binds myosin and blocks its phosphorylation, impairing cytoskeleton-mediated signaling and tumorigenesis. Moreover, ECM1a induces the expression of the heterogeneous nuclear ribonucleoprotein L like (hnRNPLL) protein to favor the alternative mRNA splicing generating ECM1a. ECM1a, αXß2, ABCG1 and hnRNPLL higher expression associates with poor survival, while ECM1b higher expression associates with good survival. These results highlight ECM1a, integrin αXß2, hnRNPLL and ABCG1 as potential targets for treating cancers associated with ECM1-activated signaling.


Subject(s)
Alternative Splicing , Carcinoma, Ovarian Epithelial/genetics , Extracellular Matrix Proteins/metabolism , Neoplasm Recurrence, Local/epidemiology , Ovarian Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Animals , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/therapy , Cell Line, Tumor , Chemotherapy, Adjuvant , Cisplatin/pharmacology , Cisplatin/therapeutic use , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Extracellular Matrix Proteins/genetics , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Integrin alphaXbeta2/genetics , Integrin alphaXbeta2/metabolism , Kaplan-Meier Estimate , Mice , Middle Aged , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Ovary/pathology , Ovary/surgery , Phosphorylation/genetics , Prognosis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Seq , Signal Transduction/genetics , Xenograft Model Antitumor Assays
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(6): 650-656, 2019 Jun 30.
Article in Chinese | MEDLINE | ID: mdl-31270042

ABSTRACT

OBJECTIVE: To investigate the effect of medium-chain acyl-CoA dehydrogenase (ACADM) on invasion and metastasis of breast cancer cells and explore the underlying mechanism. METHODS: A large cancer genome database was used to analyze the expression of ACADM in breast cancer tissues and normal tissues. The proliferation, migration and invasion of cultured breast cancer MCF-7 and T47D cells with ACADM overexpression or ACADM silencing were evaluated using MTT proliferation assay, EdU assay, Transwell chamber assay, and Boyden invasion assay; Western blotting was used to detect the protein expressions of the related pathway in the cells. In nude mouse models of tail vein metastasis of MCF-7 cells with or without ACADM overexpression, the tumor growth and tumor histopathology were observed using HE staining. RESULTS: Analysis of the Oncomine sample set showed a significantly higher expression level of ACADM in breast cancer tissues than in normal breast tissues (P < 0.05). Overexpression of ACADM obviously enhanced the migration and invasion abilities and promoted the epithelial-mesenchymal transition (EMT) of cultured MCF-7 and T47D cells; conversely, silencing of ACADM significantly suppressed the migration and invasion of the breast cancer cells. In the nude mouse models, ACADM overexpression in MCF-7 cells significantly enhanced their in vivo migration and invasion abilities. CONCLUSIONS: ACADM can promote the EMT process of breast cancer cells and improve the migration and invasion ability. ACADM is an oncogene in breast cancer.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Acyl-CoA Dehydrogenase , Animals , Cell Movement , Cell Proliferation , Humans , MCF-7 Cells , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...