1
|
Fairfield H, Costa S, Falank C, Farrell M, Murphy CS, D’Amico A, Driscoll H, Reagan MR. Multiple Myeloma Cells Alter Adipogenesis, Increase Senescence-Related and Inflammatory Gene Transcript Expression, and Alter Metabolism in Preadipocytes. Front Oncol 2021; 10:584683. [PMID: 33680918 PMCID: PMC7930573 DOI: 10.3389/fonc.2020.584683] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/23/2020] [Indexed: 12/27/2022] Open
Abstract
Within the bone marrow microenvironment, mesenchymal stromal cells (MSCs) are an essential precursor to bone marrow adipocytes and osteoblasts. The balance between this progenitor pool and mature cells (adipocytes and osteoblasts) is often skewed by disease and aging. In multiple myeloma (MM), a cancer of the plasma cell that predominantly grows within the bone marrow, as well as other cancers, MSCs, preadipocytes, and adipocytes have been shown to directly support tumor cell survival and proliferation. Increasing evidence supports the idea that MM-associated MSCs are distinct from healthy MSCs, and their gene expression profiles may be predictive of myeloma patient outcomes. Here we directly investigate how MM cells affect the differentiation capacity and gene expression profiles of preadipocytes and bone marrow MSCs. Our studies reveal that MM.1S cells cause a marked decrease in lipid accumulation in differentiating 3T3-L1 cells. Also, MM.1S cells or MM.1S-conditioned media altered gene expression profiles of both 3T3-L1 and mouse bone marrow MSCs. 3T3-L1 cells exposed to MM.1S cells before adipogenic differentiation displayed gene expression changes leading to significantly altered pathways involved in steroid biosynthesis, the cell cycle, and metabolism (oxidative phosphorylation and glycolysis) after adipogenesis. MM.1S cells induced a marked increase in 3T3-L1 expression of MM-supportive genes including Il-6 and Cxcl12 (SDF1), which was confirmed in mouse MSCs by qRT-PCR, suggesting a forward-feedback mechanism. In vitro experiments revealed that indirect MM exposure prior to differentiation drives a senescent-like phenotype in differentiating MSCs, and this trend was confirmed in MM-associated MSCs compared to MSCs from normal donors. In direct co-culture, human mesenchymal stem cells (hMSCs) exposed to MM.1S, RPMI-8226, and OPM-2 prior to and during differentiation, exhibited different levels of lipid accumulation as well as secreted cytokines. Combined, our results suggest that MM cells can inhibit adipogenic differentiation while stimulating expression of the senescence associated secretory phenotype (SASP) and other pro-myeloma molecules. This study provides insight into a novel way in which MM cells manipulate their microenvironment by altering the expression of supportive cytokines and skewing the cellular diversity of the marrow.
Collapse
Affiliation(s)
- Heather Fairfield
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Carolyne Falank
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Mariah Farrell
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Biology Department, University of Southern Maine, Portland, ME, United States
| | - Connor S. Murphy
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Anastasia D’Amico
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Biology Department, University of Southern Maine, Portland, ME, United States
| | - Heather Driscoll
- Biology Department, Norwich University, Northfield, VT, United States
| | - Michaela R. Reagan
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Biology Department, University of Southern Maine, Portland, ME, United States,*Correspondence: Michaela R. Reagan,
| |
Collapse
|
2
|
Miao L, Liu HY, Zhou C, He X. LINC00612 enhances the proliferation and invasion ability of bladder cancer cells as ceRNA by sponging miR-590 to elevate expression of PHF14. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:143. [PMID: 30940184 PMCID: PMC6444615 DOI: 10.1186/s13046-019-1149-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
Background Bladder cancer (BC) is a common type of cancer that involves tumors of the urinary system and poses a serious threat to human health. Long noncoding RNAs (lncRNAs) have emerged as crucial biomarkers and regulators in many cancers. Novel lncRNA biomarkers in BC urgently need to be investigated in regard to its function and regulatory mechanisms. Methods Identification of differentially expressed lncRNAs in BC tissue was performed via microarray analysis. To investigate the biological functions of LINC00612, loss-of-function and gain-of-function experiments were performed in vitro and in vivo. Bioinformatics analysis, dual-luciferase reporter assays, AGO2-RIP assays, RNA pull-down assays, real-time quantitative PCR (RT-qPCR) arrays, fluorescence in situ hybridization assays, and western blot assays were conducted to explore the underlying mechanisms of competitive endogenous RNAs (ceRNAs). Results LINC00612 was upregulated in BC tissues and cell lines. Functionally, downregulation of LINC00612 inhibited cell proliferation and invasion in vitro and in vivo, whereas overexpression of LINC00612 resulted in the opposite effects. Bioinformatics analysis and luciferase assays revealed that miR-590 was a direct target of LINC0061, which was validated by dual-luciferase reporter assays, AGO2-RIP assays, RNA pull-down assays, RT-qPCR arrays, and rescue experiments. Additionally, miR-590 was shown to directly target the PHD finger protein 14 (PHF14) gene. LNIC00612 modulated the expression of E-cadherin and vimentin by competitively sponging miR-590 to elevate the expression of PHF14, thus affecting BC cellular epithelial-mesenchymal transition (EMT). Conclusions Our results indicate that LINC00612 enhances the proliferation and invasion ability of BC cells by sponging miR-590 to upregulate PHF14 expression and promote BC cellular EMT, suggesting that LINC00612 may act as a potential biomarker and therapeutic target for BC.
Collapse
Affiliation(s)
- Liying Miao
- Department of Hemodialysis, The Third Affiliated Hospital of Soochow University, Changzhou Shi, China
| | - Hong Yue Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cuixing Zhou
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Xiaozhou He
- Department of Hemodialysis, The Third Affiliated Hospital of Soochow University, Changzhou Shi, China. .,Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| |
Collapse
|
5
|
Wang S, Fan W, Wan B, Tu M, Jin F, Liu F, Xu H, Han P. Characterization of long noncoding RNA and messenger RNA signatures in melanoma tumorigenesis and metastasis. PLoS One 2017; 12:e0172498. [PMID: 28225791 PMCID: PMC5321451 DOI: 10.1371/journal.pone.0172498] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/06/2017] [Indexed: 01/10/2023] Open
Abstract
The incidence of melanoma, the most aggressive and life-threatening form of skin cancer, has significantly risen over recent decades. Therefore, it is essential to identify the mechanisms that underlie melanoma tumorigenesis and metastasis and to explore novel and effective melanoma treatment strategies. Accumulating evidence s uggests that aberrantly expressed long noncoding RNAs (lncRNAs) have vital functions in multiple cancers. However, lncRNA functions in melanoma tumorigenesis and metastasis remain unclear. In this study, we investigated lncRNA and messenger RNA (mRNA) expression profiles in primary melanomas, metastatic melanomas and normal skin samples from the Gene Expression Omnibus database. We used GSE15605 as the training set (n = 74) and GSE7553 as the validation set (n = 58). In three comparisons (primary melanoma versus normal skin, metastatic melanoma versus normal skin, and metastatic melanoma versus primary melanoma), 178, 295 and 48 lncRNAs and 847, 1758, and 295 mRNAs were aberrantly expressed, respectively. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses to examine the differentially expressed mRNAs, and potential core lncRNAs were predicted by lncRNA-mRNA co-expression networks. Based on our results, 15 lncRNAs and 144 mRNAs were significantly associated with melanoma tumorigenesis and metastasis. A subsequent analysis suggested a critical role for a five-lncRNA signature during melanoma tumorigenesis and metastasis. Low expression of U47924.27 was significantly associated with decreased survival of patients with melanoma. To the best of our knowledge, this study is the first to explore the expression patterns of lncRNAs and mRNAs during melanoma tumorigenesis and metastasis by re-annotating microarray data from the Gene Expression Omnibus (GEO) microarray dataset. These findings reveal potential roles for lncRNAs during melanoma tumorigenesis and metastasis and provide a rich candidate reservoir for future studies.
Collapse
Affiliation(s)
- Siqi Wang
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Wenliang Fan
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bing Wan
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Mengqi Tu
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Feng Jin
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Radiology, The First Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People’s Republic of China
| | - Fang Liu
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- * E-mail: (PH); (HX)
| | - Ping Han
- Department of Radiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- * E-mail: (PH); (HX)
| |
Collapse
|