1
|
Cazares O, Chen M, Menendez J, Molinuevo R, Thomas G, Cervantes J, Yee M, Cadell M, Durham M, Zhu Y, Strietzel C, Bubolz JW, Hinck L. SLIT Loss or Sequestration Increases Mammary Alveologenesis and Lactogenesis. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001264. [PMID: 39381643 PMCID: PMC11461027 DOI: 10.17912/micropub.biology.001264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/17/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
SLITs comprise a family of secreted proteins that function as ligands for Roundabout (ROBO) receptors. Previous research showed that ROBO1 promotes the differentiation of milk-producing alveolar cells by inhibiting Notch signaling in mammary luminal cells. Here, we show enhanced alveolar development and increased milk production in Slit2-/-;Slit3-/- knockout mammary gland epithelia. This result can also be achieved by intraperitoneal delivery of recombinant ROBO1 extracellular domain fragment, ROBO1-5Ig-Fc, which sequesters SLITs. Together, our phenotypic studies suggest that SLITs restrict alveologenesis and lactogenesis by inhibiting ROBO1.
Collapse
Affiliation(s)
| | - Min Chen
- University of California, Santa Cruz, CA, USA
| | | | | | - Gwen Thomas
- University of California, Santa Cruz, CA, USA
| | | | - Michael Yee
- University of California, Santa Cruz, CA, USA
| | | | | | - Yaqi Zhu
- Zoetis (United States), Kalamazoo, MI, United States
| | | | | | | |
Collapse
|
2
|
Ma H, Zhang T. Histone demethylase KDM3B mediates matrix stiffness-induced osteogenic differentiation of adipose-derived stem cells. Arch Biochem Biophys 2024; 757:110028. [PMID: 38768746 DOI: 10.1016/j.abb.2024.110028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Biomechanical signals in the extracellular niche are considered promising for programming the lineage specification of stem cells. Recent studies have reported that biomechanics, such as the microstructure of nanomaterials, can induce adipose-derived stem cells (ASCs) to differentiate into osteoblasts, mediating gene regulation at the epigenetic level. Therefore, in this study, transcriptome expression levels of histone demethylases in ASCs were screened after treatment with different matrix stiffnesses, and histone lysine demethylase 3B (KDM3B) was found to promote osteogenic differentiation of ASCs in response to matrix stiffness, indicating a positive modulatory effect on this biological process. ASCs exhibited widespread and polygonal shapes with a distinct bundle-like expression of vinculin parallel to the axial cytoskeleton along the cell margins on the stiff matrix rather than round shapes with a smeared and shorter expression on the soft matrix. Comparatively rigid polydimethylsiloxane material directed ASCs into an osteogenic phenotype in inductive culture media via the upregulation of osteocalcin, alkaline phosphatase, and runt-related transcription factor 2. Treatment with KDM3B-siRNA decreased the expression of osteogenic differentiation markers and impaired mitochondrial dynamics and mitochondrial membrane potential. These results illustrate the critical role of KDM3B in the biomechanics-induced osteogenic commitment of ASCs and provide new avenues for the further application of stem cells as potential therapeutics for bone regeneration.
Collapse
Affiliation(s)
- Huangshui Ma
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
3
|
Qiu H, Fu Y, Guo Z, Zhang X, Wang X, Wu H. Dysregulated microRNAs and long non-coding RNAs associated with extracellular matrix stiffness. Exp Cell Res 2024; 437:114014. [PMID: 38547959 DOI: 10.1016/j.yexcr.2024.114014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Extracellular matrix (ECM) stiffness regulates development and homeostasis in vivo and affects both physiological and pathological processes. A variety of studies have demonstrated that mRNAs, such as Piezo1, integrin β1, and Yes-associated protein (YAP)/tafazzin (TAZ), can sense the mechanical signals induced by ECM stiffness and transmit them from the extracellular space into the cytoplasm. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), have been reported to play important roles in various cellular processes. Therefore, the interactions between ncRNAs and ECM stiffness, as well as the underlying molecular mechanisms, have become intriguing. In this review, we summarize recent findings on miRNAs and lncRNAs that interact with ECM stiffness. Several miRNAs and lncRNAs are involved in the progression of liver cancer, breast cancer, osteosarcoma, and cardiovascular diseases under the regulation of ECM stiffness. Through these ncRNAs, cellular behaviors including cell differentiation, proliferation, adhesion, migration, invasion, and epithelial-mesenchymal transition (EMT) are affected by ECM stiffness. We also integrate the ncRNA signaling pathways associated with ECM stiffness, in which typical signaling pathways like integrin β1/TGFβ1, phosphatidylinositol-3 kinase (PI3K)/AKT, and EMT are involved. Although our understanding of the relationships between ncRNAs and ECM stiffness is still limited, further investigations may provide new insights for disease treatment. ECM-associated ncRNAs may serve as disease biomarkers or be targeted by drugs.
Collapse
Affiliation(s)
- Huimin Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Yangpu, 200093, Shanghai, China; Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Yi Fu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Zhinan Guo
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China; School of Sports and Health, Shanghai University of Sport, Yangpu, 200438, Shanghai, China.
| | - Xinjia Zhang
- School of Medical Instruments, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Xinyue Wang
- School of Medical Instruments, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Hailong Wu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| |
Collapse
|
4
|
Zhao S, Liu H, Wang H, He X, Tang J, Qi S, Yang R, Xie J. Inhibition of phosphatidylinositol 3-kinase catalytic subunit alpha by miR-203a-3p reduces hypertrophic scar formation via phosphatidylinositol 3-kinase/AKT/mTOR signaling pathway. BURNS & TRAUMA 2024; 12:tkad048. [PMID: 38179473 PMCID: PMC10762504 DOI: 10.1093/burnst/tkad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 01/06/2024]
Abstract
Background Hypertrophic scar (HS) is a common fibroproliferative skin disease that currently has no truly effective therapy. Given the importance of phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) in hypertrophic scar formation, the development of therapeutic strategies for endogenous inhibitors against PIK3CA is of great interest. Here, we explored the molecular mechanisms underlying the protective effects of miR-203a-3p (PIK3CA inhibitor) against excessive scar. Methods Bioinformatic analysis, immunohistochemistry, immunofluorescence, miRNA screening and fluorescence in situ hybridization assays were used to identify the possible pathways and target molecules mediating HS formation. A series of in vitro and in vivo experiments were used to clarify the role of PIK3CA and miR-203a-3p in HS. Mechanistically, transcriptomic sequencing, immunoblotting, dual-luciferase assay and rescue experiments were executed. Results Herein, we found that PIK3CA and the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway were upregulated in scar tissues and positively correlated with fibrosis. We then identified miR-203a-3p as the most suitable endogenous inhibitor of PIK3CA. miR-203a-3p suppressed the proliferation, migration, collagen synthesis and contractility as well as the transdifferentiation of fibroblasts into myofibroblasts in vitro, and improved the morphology and histology of scars in vivo. Mechanistically, miR-203a-3p attenuated fibrosis by inactivating the PI3K/AKT/mTOR pathway by directly targeting PIK3CA. Conclusions PIK3CA and the PI3K/AKT/mTOR pathway are actively involved in scar fibrosis and miR-203a-3p might serve as a potential strategy for hypertrophic scar therapy through targeting PIK3CA and inactivating the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Shixin Zhao
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Hengdeng Liu
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Hanwen Wang
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Xuefeng He
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Jinming Tang
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Shaohai Qi
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| | - Julin Xie
- Department of Burns, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, No. 58 Zhongshan Second Road, Yuexiu District, Guangzhou, Guangdong, 510062, China
| |
Collapse
|
5
|
Kim OH, Jeon TJ, Shin YK, Lee HJ. Role of extrinsic physical cues in cancer progression. BMB Rep 2023; 56:287-295. [PMID: 37037673 PMCID: PMC10230013 DOI: 10.5483/bmbrep.2023-0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 07/22/2023] Open
Abstract
The tumor microenvironment (TME) is a complex system composed of many cell types and an extracellular matrix (ECM). During tumorigenesis, cancer cells constantly interact with cellular components, biochemical cues, and the ECM in the TME, all of which make the environment favorable for cancer growth. Emerging evidence has revealed the importance of substrate elasticity and biomechanical forces in tumor progression and metastasis. However, the mechanisms underlying the cell response to mechanical signals-such as extrinsic mechanical forces and forces generated within the TME-are still relatively unknown. Moreover, having a deeper understanding of the mechanisms by which cancer cells sense mechanical forces and transmit signals to the cytoplasm would substantially help develop effective strategies for cancer treatment. This review provides an overview of biomechanical forces in the TME and the intracellular signaling pathways activated by mechanical cues as well as highlights the role of mechanotransductive pathways through mechanosensors that detect the altering biomechanical forces in the TME. as an adjuvant for cancer immunotherapy.[BMB Reports 2023; 56(5): 287-295].
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Tae Jin Jeon
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, Korea
| | - Yong Kyoo Shin
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
6
|
Kim OH, Jeon TJ, Shin YK, Lee HJ. Role of extrinsic physical cues in cancer progression. BMB Rep 2023; 56:287-295. [PMID: 37037673 PMCID: PMC10230013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/12/2023] Open
Abstract
The tumor microenvironment (TME) is a complex system composed of many cell types and an extracellular matrix (ECM). During tumorigenesis, cancer cells constantly interact with cellular components, biochemical cues, and the ECM in the TME, all of which make the environment favorable for cancer growth. Emerging evidence has revealed the importance of substrate elasticity and biomechanical forces in tumor progression and metastasis. However, the mechanisms underlying the cell response to mechanical signals-such as extrinsic mechanical forces and forces generated within the TME-are still relatively unknown. Moreover, having a deeper understanding of the mechanisms by which cancer cells sense mechanical forces and transmit signals to the cytoplasm would substantially help develop effective strategies for cancer treatment. This review provides an overview of biomechanical forces in the TME and the intracellular signaling pathways activated by mechanical cues as well as highlights the role of mechanotransductive pathways through mechanosensors that detect the altering biomechanical forces in the TME. as an adjuvant for cancer immunotherapy.[BMB Reports 2023; 56(5): 287-295].
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Tae Jin Jeon
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, Korea
| | - Yong Kyoo Shin
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
7
|
Xie N, Xiao C, Shu Q, Cheng B, Wang Z, Xue R, Wen Z, Wang J, Shi H, Fan D, Liu N, Xu F. Cell response to mechanical microenvironment cues via Rho signaling: From mechanobiology to mechanomedicine. Acta Biomater 2023; 159:1-20. [PMID: 36717048 DOI: 10.1016/j.actbio.2023.01.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/30/2023]
Abstract
Mechanical cues in the cell microenvironment such as those from extracellular matrix properties, stretching, compression and shear stress, play a critical role in maintaining homeostasis. Upon sensing mechanical stimuli, cells can translate these external forces into intracellular biochemical signals to regulate their cellular behaviors, but the specific mechanisms of mechanotransduction at the molecular level remain elusive. As a subfamily of the Ras superfamily, Rho GTPases have been recognized as key intracellular mechanotransduction mediators that can regulate multiple cell activities such as proliferation, migration and differentiation as well as biological processes such as cytoskeletal dynamics, metabolism, and organ development. However, the upstream mechanosensors for Rho proteins and downstream effectors that respond to Rho signal activation have not been well illustrated. Moreover, Rho-mediated mechanical signals in previous studies are highly context-dependent. In this review, we systematically summarize the types of mechanical cues in the cell microenvironment and provide recent advances on the roles of the Rho-based mechanotransduction in various cell activities, physiological processes and diseases. Comprehensive insights into the mechanical roles of Rho GTPase partners would open a new paradigm of mechanomedicine for a variety of diseases. STATEMENT OF SIGNIFICANCE: In this review, we highlight the critical role of Rho GTPases as signal mediators to respond to physical cues in microenvironment. This article will add a distinct contribution to this set of knowledge by intensively addressing the relationship between Rho signaling and mechanobiology/mechanotransduction/mechanomedcine. This topic has not been discussed by the journal, nor has it yet been developed by the field. The comprehensive picture that will develop, from molecular mechanisms and engineering methods to disease treatment strategies, represents an important and distinct contribution to the field. We hope that this review would help researchers in various fields, especially clinicians, oncologists and bioengineers, who study Rho signal pathway and mechanobiology/mechanotransduction, understand the critical role of Rho GTPase in mechanotransduction.
Collapse
Affiliation(s)
- Ning Xie
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Cailan Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Qiuai Shu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ziwei Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Runxin Xue
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Zhang Wen
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Jinhai Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Haitao Shi
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an Shaanxi 710049, China.
| | - Na Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
8
|
Zhang Q, Wang P, Fang X, Lin F, Fang J, Xiong C. Collagen gel contraction assays: From modelling wound healing to quantifying cellular interactions with three-dimensional extracellular matrices. Eur J Cell Biol 2022; 101:151253. [PMID: 35785635 DOI: 10.1016/j.ejcb.2022.151253] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 06/06/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cells respond to and actively remodel the extracellular matrix (ECM). The dynamic and bidirectional interaction between cells and ECM, especially their mechanical interactions, has been found to play an essential role in triggering a series of complex biochemical and biomechanical signal pathways and in regulating cellular functions and behaviours. The collagen gel contraction assay (CGCA) is a widely used method to investigate cell-ECM interactions in 3D environments and provides a mechanically associated readout reflecting 3D cellular contractility. In this review, we summarize various versions of CGCA, with an emphasis on recent high-throughput and low-consumption CGCA techniques. More importantly, we focus on the technique of force monitoring during the contraction of collagen gel, which provides a quantitative characterization of the overall forces generated by all the resident cells in the collagen hydrogel. Accordingly, we present recent biological applications of the CGCA, which have expanded from the initial wound healing model to other studies concerning cell-ECM interactions, including fibrosis, cancer, tissue repair and the preparation of biomimetic microtissues.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Pudi Wang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Xu Fang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Feng Lin
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Jing Fang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
9
|
Yin T, Zhao H. miR-152-3p impedes the malignant phenotypes of hepatocellular carcinoma by repressing roundabout guidance receptor 1. Cell Mol Biol Lett 2022; 27:22. [PMID: 35236289 PMCID: PMC8903719 DOI: 10.1186/s11658-022-00322-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND miR-152-3p functions as a tumour suppressor in the progression of hepatic tumorigenesis. Herein, we further discussed the prognostic significance and immune infiltration of miR-152-3p and its potential gene target in hepatocellular carcinoma (HCC). METHODS The Cancer Genome Atlas (TCGA), Integrative Molecular Database of Hepatocellular Carcinoma (HCCDB), Human Protein Atlas (HPA) and Kaplan-Meier Plotter databases were used to evaluate miR-152-3p and roundabout guidance receptor 1 (ROBO1) expression, prognosis and immune infiltration. In vitro cell experiments, including cell proliferation and apoptosis, were evaluated using Cell Counting Kit 8 (CCK8) and terminal-deoxynucleotidyl transferase-mediated nick end labelling (TUNEL) assays. RESULTS Up-regulation of ROBO1 functioned as an oncogene associated with poor prognosis, immune cell enrichment and cell proliferation in HCC. ROBO1 was significantly positively correlated with the enrichment of multiple immune cells and their biomarkers. Enrichment of type-2 T-helper (Th2) cells is an unfavourable biomarker of HCC prognosis. GSEA revealed that ROBO1 correlated with apoptosis, mitosis and carcinogenic signalling pathways. Suppression of cell proliferation and the enhancement of cell apoptosis by miR-152-3p mimics were counteracted by overexpression of ROBO1 in HCC cells. CONCLUSION ROBO1 expression is positively correlated with multiple immune checkpoint molecules, suggesting that ROBO1 may be a potential drug target to enhance the potency of immunotherapy. The miR-152-3p/ROBO1 signalling axis contributes to malignant progression and provides a prospective immunotherapeutic target for HCC.
Collapse
Affiliation(s)
- Tao Yin
- Department of General Surgery, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan, Chifeng, 024005, China.
| | - Haonan Zhao
- Department of General Surgery, Affiliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan, Chifeng, 024005, China
| |
Collapse
|
10
|
Zheng S, Hu C, Lin H, Li G, Xia R, Zhang X, Su D, Li Z, Zhou Q, Chen R. circCUL2 induces an inflammatory CAF phenotype in pancreatic ductal adenocarcinoma via the activation of the MyD88-dependent NF-κB signaling pathway. J Exp Clin Cancer Res 2022; 41:71. [PMID: 35189958 PMCID: PMC8862589 DOI: 10.1186/s13046-021-02237-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/27/2021] [Indexed: 12/29/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is characterized by clusters of cancer cells surrounded by a dense desmoplastic stroma. However, little is known about stromal cell heterogeneity in the pancreatic tumor microenvironment. Methods We conducted circRNA profiling in primary fibroblasts by high-throughput sequencing and detected circCUL2 levels in PDAC tissues by qRT–PCR. We subsequently investigated the effect of circCUL2 on inflammatory cancer-associated fibroblast (iCAF) activation, heterogeneity and protumor activity by ELISA, flow cytometry, colony formation and transwell assays in vitro and by xenograft models in vivo. The regulatory effect of circCUL2 on miR-203a-3p/MyD88/IL6 was examined by RNA pulldown, FISH, and luciferase reporter assays. Results We identified that circCUL2 was specifically expressed in cancer-associated fibroblasts (CAFs) but not in cancer cells. Moreover, the enrichment of circCUL2 in tumor tissues was significantly correlated with the poor prognosis of PDAC patients. Upregulation of circCUL2 expression in normal fibroblasts (NFs) induced the iCAF phenotype, and then iCAFs promoted PDAC progression through IL6 secretion in vitro. Furthermore, circCUL2-transduced NFs promoted tumorigenesis and metastasis of PDAC cells in vivo, which was blocked by an anti-IL6 antibody. Mechanistically, circCUL2 functioned as a ceRNA and modulated the miR-203a-3p/MyD88/NF-κB/IL6 axis, thereby further activating the STAT3 signaling pathway in pancreatic cancer cells to induce PDAC progression. Conclusions We showed that the circCUL2/miR-203a-5p/MyD88/NF-κB/IL6 axis contributes to the induction of iCAFs and established a distinct fibroblast niche for PDAC progression, which could help the development of strategies that selectively target tumor-promoting CAFs in PDAC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02237-6.
Collapse
|
11
|
Nguyen Ho-Bouldoires TH, Sollier K, Zamfirov L, Broders-Bondon F, Mitrossilis D, Bermeo S, Guerin CL, Chipont A, Champenois G, Leclère R, André N, Ranno L, Michel A, Ménager C, Meseure D, Demené C, Tanter M, Fernández-Sánchez ME, Farge E. Ret kinase-mediated mechanical induction of colon stem cells by tumor growth pressure stimulates cancer progression in vivo. Commun Biol 2022; 5:137. [PMID: 35177769 PMCID: PMC8854631 DOI: 10.1038/s42003-022-03079-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 01/26/2022] [Indexed: 12/20/2022] Open
Abstract
How mechanical stress actively impacts the physiology and pathophysiology of cells and tissues is little investigated in vivo. The colon is constantly submitted to multi-frequency spontaneous pulsatile mechanical waves, which highest frequency functions, of 2 s period, remain poorly understood. Here we find in vivo that high frequency pulsatile mechanical stresses maintain the physiological level of mice colon stem cells (SC) through the mechanosensitive Ret kinase. When permanently stimulated by a magnetic mimicking-tumor growth analogue pressure, we find that SC levels pathologically increase and undergo mechanically induced hyperproliferation and tumorigenic transformation. To mimic the high frequency pulsatile mechanical waves, we used a generator of pulsed magnetic force stimulation in colonic tissues pre-magnetized with ultra-magnetic liposomes. We observed the pulsatile stresses using last generation ultra-wave dynamical high-resolution imaging. Finally, we find that the specific pharmacological inhibition of Ret mechanical activation induces the regression of spontaneous formation of SC, of CSC markers, and of spontaneous sporadic tumorigenesis in Apc mutated mice colons. Consistently, in human colon cancer tissues, Ret activation in epithelial cells increases with tumor grade, and partially decreases in leaking invasive carcinoma. High frequency pulsatile physiological mechanical stresses thus constitute a new niche that Ret-dependently fuels mice colon physiological SC level. This process is pathologically over-activated in the presence of permanent pressure due to the growth of tumors initiated by pre-existing genetic alteration, leading to mechanotransductive self-enhanced tumor progression in vivo, and repressed by pharmacological inhibition of Ret. Ho-Bouldoires, Sollier, Zamfirov and Broders-Bondon et al. show that high frequency pulsatile mechanical stresses maintain the physiological level of mice colon stem cells through the mechanosensitive Ret kinase and that Ret activation is elevated in human colon cancer tissue. They go on to show that the maintenance of such stimulation in the form of tumour growth pressure results in mechanically-induced hyperproliferation and tumorigenic transformation of stem cells, which can be prevented by Ret kinase inhibition.
Collapse
Affiliation(s)
- Thanh Huong Nguyen Ho-Bouldoires
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France
| | - Kévin Sollier
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France
| | - Laura Zamfirov
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France.,Physics for Medicine Paris, ESPCI ParisTech, PSL Research University, Inserm U1273, F-75005, Paris, France
| | - Florence Broders-Bondon
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France
| | - Démosthène Mitrossilis
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France.,Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St., 115 27, Athens, Greece
| | - Sebastian Bermeo
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France
| | | | - Anna Chipont
- Cytometry Platform, Institut Curie, Paris, France
| | - Gabriel Champenois
- Platform of Investigative Pathology, Institut Curie, 75248, Paris, France
| | - Renaud Leclère
- Platform of Investigative Pathology, Institut Curie, 75248, Paris, France
| | - Nicolas André
- Platform of Investigative Pathology, Institut Curie, 75248, Paris, France
| | - Laurent Ranno
- NEEL Institut, CNRS, Grenoble Alpes University, F-38042, Grenoble, France
| | - Aude Michel
- Sorbonne Université, Laboratoire PHENIX Physico-chimie des Electrolytes et Nanosystèmes Interfaciaux, CNRS UMR 8234, F-75005, Paris, France
| | - Christine Ménager
- Sorbonne Université, Laboratoire PHENIX Physico-chimie des Electrolytes et Nanosystèmes Interfaciaux, CNRS UMR 8234, F-75005, Paris, France
| | - Didier Meseure
- Platform of Investigative Pathology, Institut Curie, 75248, Paris, France
| | - Charlie Demené
- Physics for Medicine Paris, ESPCI ParisTech, PSL Research University, Inserm U1273, F-75005, Paris, France
| | - Mickael Tanter
- Physics for Medicine Paris, ESPCI ParisTech, PSL Research University, Inserm U1273, F-75005, Paris, France
| | - Maria Elena Fernández-Sánchez
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France.
| | - Emmanuel Farge
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire de Physico-Chimie Curie, Mechanics and Genetics of Embryonic and Tumoral Development team, INSERM, F-75005, Paris, France.
| |
Collapse
|
12
|
Yang H, Zhou S, Lan D, Bin Y, Bao W, Wang M, Huang F, Peng Z. The expression of Slit2 and Robo1 increased during retinoic acid syndrome in acute promyelocytic leukemia and impacted differentiated cell migration. Transl Oncol 2022; 18:101370. [PMID: 35182953 PMCID: PMC8857660 DOI: 10.1016/j.tranon.2022.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/25/2022] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
The upregulation of Robo1 and Slit2 was first found in APL patients. The positive correlation between Robo1/Slit2 and retinoic acid syndrome was first demonstrated. It was demonstrated for the first time that Slit2 induces the migration of differentiated cells. Slit2 did not inhibit il8-induced differentiated cell migration.
Retinoic acid syndrome (RAS) is a serious complication developed during the induction therapy of acute promyelocytic leukemia (APL). Cytokines and differentiated cells migration play important roles in the development of RAS. Slit guidance ligand 2 (Slit2) and roundabout 1 (Robo1) involve in cell migration. Our study aimed to investigate the expression of Slit2 and Robo1 in APL and check whether they affected promyelocytes migration. 62 cases of newly diagnosed APL patients were involved and received all-trans retinoic acid (ATRA) and arsenic trioxide as induction therapy. Bone marrow cells (BMCs) were obtained on days 0 and 28, and promyelocytes and plasma were collected from day 1 to day 21. The expression of Robo1 in promyelocytes, and that of Slit2 and cytokines, including IL-8,IL-1β and others, in serum were monitored. 20 healthy individuals donated their cells as control. Of the 62 APL patients, 16 (25.81%) patients developed RAS. The expression of Robo1, Slit2 and IL-8 increased significantly with the development of RAS. In the 16 patients with RAS, levels of Slit2, Robo1 and IL-8 were higher during the development of RAS than before or after the RAS (P < 0.05). RhSlit2-N and rhIL-8 induced cells migration, and the migration induced by IL-8 was not inhibited by rhSlit2-N. Elevated Slit2 and Robo1 levels might be useful markers for the diagnosis and treatment of RAS. The levels of Slit2, Robo1 and IL-8 showed a positive correlation with the severity of RAS. Slit2 and IL-8 promoted the migration of differentiated cells.
Collapse
Affiliation(s)
- Haiyan Yang
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shengsheng Zhou
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dong Lan
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yehong Bin
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Wenguang Bao
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Man Wang
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Fengxiang Huang
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhigang Peng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
13
|
Cazares O, Chatterjee S, Lee P, Strietzel C, Bubolz JW, Harburg G, Howard J, Katzman S, Sanford J, Hinck L. Alveolar progenitor differentiation and lactation depends on paracrine inhibition of notch via ROBO1/CTNNB1/JAG1. Development 2021; 148:dev199940. [PMID: 34758082 PMCID: PMC8627605 DOI: 10.1242/dev.199940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/23/2021] [Indexed: 11/09/2022]
Abstract
In the mammary gland, how alveolar progenitor cells are recruited to fuel tissue growth with each estrus cycle and pregnancy remains poorly understood. Here, we identify a regulatory pathway that controls alveolar progenitor differentiation and lactation by governing Notch activation in mouse. Loss of Robo1 in the mammary gland epithelium activates Notch signaling, which expands the alveolar progenitor cell population at the expense of alveolar differentiation, resulting in compromised lactation. ROBO1 is expressed in both luminal and basal cells, but loss of Robo1 in basal cells results in the luminal differentiation defect. In the basal compartment, ROBO1 inhibits the expression of Notch ligand Jag1 by regulating β-catenin (CTNNB1), which binds the Jag1 promoter. Together, our studies reveal how ROBO1/CTTNB1/JAG1 signaling in the basal compartment exerts paracrine control of Notch signaling in the luminal compartment to regulate alveolar differentiation during pregnancy.
Collapse
Affiliation(s)
- Oscar Cazares
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Sharmila Chatterjee
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Pinky Lee
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA 95064, USA
| | | | - J. W. Bubolz
- Zoetis Inc. 333 Portage Street, Building 300, Kalamazoo, MI 49007, USA
| | - Gwyndolen Harburg
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Jon Howard
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
| | - Sol Katzman
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
| | - Jeremy Sanford
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Lindsay Hinck
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| |
Collapse
|
14
|
Li S, Qiao S, Li N, Zhu X. MiR-744 Functions as an Oncogene Through Direct Binding to c-Fos Promoter and Facilitates Non-small Cell Lung Cancer Progression. Ann Surg Oncol 2021; 29:1465-1475. [PMID: 34599436 DOI: 10.1245/s10434-021-10688-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/04/2021] [Indexed: 11/18/2022]
Abstract
Metastasis is the leading cause of death in non-small cell lung cancer (NSCLC) patients. Previously, we reported that miR-744 exerted proto-oncogenic function in nasopharyngeal carcinoma, but the role of miR-744 during NSCLC development has not been established. We focused on the function and molecular mechanism of miR-744 in NSCLC. The clinical cohort data from TCGA were analyzed for the correlation of miR-744 and outcomes in NSCLC patients. Gain- and loss-of-function experiment was performed by transfection with miR-744 agomir or antagomir in NSCLC cell lines. The expression of mRNA and protein were analyzed by qPCR assays and Western blotting respectively. Cellular proliferation, migration, and invasion were analyzed by CCK8 assays, wound healing, and transwell assays, respectively. Promoter activities and gene transcription were analyzed by luciferase reporter assays. Xenograft model was applied for in vivo study. High miR-744 expression correlated with lymph node metastasis and poor prognosis in NSCLC patient. MiR-744 aggravated the growth, invasion, and metastasis of NSCLC cells eventually induced the malignant phenotype and promotes radio/chemoresistance in vitro. The -1195 to -1227 and -298 to -323 bp upstream of c-FOS gene was observed to bind with miR-744. Lastly, miR-744 acted as a tumor promoter in lung cancer growth and metastasis in vivo. Taken together, our results indicated that miR-744 up-regulated c-Fos by binding with its promoter contributed to development of NSCLC cells malignant phenotype. Our findings highlight the potential value of miR-744, which may serve as a possible therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Shangbiao Li
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Simiao Qiao
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Na Li
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Zhu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
The Role of miRNAs in Extracellular Matrix Repair and Chronic Fibrotic Lung Diseases. Cells 2021; 10:cells10071706. [PMID: 34359876 PMCID: PMC8304879 DOI: 10.3390/cells10071706] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
The lung extracellular matrix (ECM) plays a key role in the normal architecture of the lung, from embryonic lung development to mechanical stability and elastic recoil of the breathing adult lung. The lung ECM can modulate the biophysical environment of cells through ECM stiffness, porosity, topography and insolubility. In a reciprocal interaction, lung ECM dynamics result from the synthesis, degradation and organization of ECM components by the surrounding structural and immune cells. Repeated lung injury and repair can trigger a vicious cycle of aberrant ECM protein deposition, accompanied by elevated ECM stiffness, which has a lasting effect on cell and tissue function. The processes governing the resolution of injury repair are regulated by several pathways; however, in chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary disease (IPF) these processes are compromised, resulting in impaired cell function and ECM remodeling. Current estimates show that more than 60% of the human coding transcripts are regulated by miRNAs. miRNAs are small non-coding RNAs that regulate gene expressions and modulate cellular functions. This review is focused on the current knowledge of miRNAs in regulating ECM synthesis, degradation and topography by cells and their dysregulation in asthma, COPD and IPF.
Collapse
|
16
|
Northey JJ, Barrett AS, Acerbi I, Hayward MK, Talamantes S, Dean IS, Mouw JK, Ponik SM, Lakins JN, Huang PJ, Wu J, Shi Q, Samson S, Keely PJ, Mukhtar RA, Liphardt JT, Shepherd JA, Hwang ES, Chen YY, Hansen KC, Littlepage LE, Weaver VM. Stiff stroma increases breast cancer risk by inducing the oncogene ZNF217. J Clin Invest 2021; 130:5721-5737. [PMID: 32721948 DOI: 10.1172/jci129249] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Women with dense breasts have an increased lifetime risk of malignancy that has been attributed to a higher epithelial density. Quantitative proteomics, collagen analysis, and mechanical measurements in normal tissue revealed that stroma in the high-density breast contains more oriented, fibrillar collagen that is stiffer and correlates with higher epithelial cell density. microRNA (miR) profiling of breast tissue identified miR-203 as a matrix stiffness-repressed transcript that is downregulated by collagen density and reduced in the breast epithelium of women with high mammographic density. Culture studies demonstrated that ZNF217 mediates a matrix stiffness- and collagen density-induced increase in Akt activity and mammary epithelial cell proliferation. Manipulation of the epithelium in a mouse model of mammographic density supported a causal relationship between stromal stiffness, reduced miR-203, higher levels of the murine homolog Zfp217, and increased Akt activity and mammary epithelial proliferation. ZNF217 was also increased in the normal breast epithelium of women with high mammographic density, correlated positively with epithelial proliferation and density, and inversely with miR-203. The findings identify ZNF217 as a potential target toward which preexisting therapies, such as the Akt inhibitor triciribine, could be used as a chemopreventive agent to reduce cancer risk in women with high mammographic density.
Collapse
Affiliation(s)
- Jason J Northey
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Alexander S Barrett
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Irene Acerbi
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Mary-Kate Hayward
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Stephanie Talamantes
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Ivory S Dean
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Janna K Mouw
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jonathon N Lakins
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Po-Jui Huang
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA
| | - Junmin Wu
- Harper Cancer Research Institute, Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana, USA
| | - Quanming Shi
- Department of Bioengineering, Stanford University, Palo Alto, California, USA
| | - Susan Samson
- Helen Diller Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Jan T Liphardt
- Department of Bioengineering, Stanford University, Palo Alto, California, USA
| | - John A Shepherd
- Population Sciences in the Pacific Program (Cancer Epidemiology), University of Hawaii Cancer Center, University of Hawaii at Manoa, Manoa, Hawaii, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Yunn-Yi Chen
- Department of Pathology, UCSF, San Francisco, California, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA.,Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laurie E Littlepage
- Harper Cancer Research Institute, Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana, USA
| | - Valerie M Weaver
- Department of Surgery.,Center for Bioengineering and Tissue Regeneration, UCSF, San Francisco, California, USA.,Helen Diller Comprehensive Cancer Center, UCSF, San Francisco, California, USA.,Population Sciences in the Pacific Program (Cancer Epidemiology), University of Hawaii Cancer Center, University of Hawaii at Manoa, Manoa, Hawaii, USA.,Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| |
Collapse
|
17
|
Non-coding RNAs modulate function of extracellular matrix proteins. Biomed Pharmacother 2021; 136:111240. [PMID: 33454598 DOI: 10.1016/j.biopha.2021.111240] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/19/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
The extracellular matrix (ECM) creates a multifaceted system for the interaction of diverse structural proteins, matricellular molecules, proteoglycans, hyaluronan, and various glycoproteins that collaborate and bind with each other to produce a bioactive polymer. Alterations in the composition and configuration of ECM elements influence the cellular phenotype, thus participating in the pathogenesis of several human disorders. Recent studies indicate the crucial roles of non-coding RNAs in the modulation of ECM. Several miRNAs such as miR-21, miR-26, miR-19, miR-140, miR-29, miR-30, miR-133 have been dysregulated in disorders that are associated with disruption or breakdown of the ECM. Moreover, expression of MALAT1, PVT1, SRA1, n379519, RMRP, PFL, TUG1, TM1P3, FAS-AS1, PART1, XIST, and expression of other lncRNAs is altered in disorders associated with the modification of ECM components. In the current review, we discuss the role of lncRNAs and miRNAs in the modification of ECM and their relevance with the pathophysiology of human disorders such as cardiac/ lung fibrosis, cardiomyopathy, heart failure, asthma, osteoarthritis, and cancers.
Collapse
|
18
|
The Physiological MicroRNA Landscape in Nipple Aspirate Fluid: Differences and Similarities with Breast Tissue, Breast Milk, Plasma and Serum. Int J Mol Sci 2020; 21:ijms21228466. [PMID: 33187146 PMCID: PMC7696615 DOI: 10.3390/ijms21228466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background: MicroRNAs (miRNAs) target 60% of human messenger RNAs and can be detected in tissues and biofluids without loss of stability during sample processing, making them highly appraised upcoming biomarkers for evaluation of disease. However, reporting of the abundantly expressed miRNAs in healthy samples is often surpassed. Here, we characterized for the first time the physiological miRNA landscape in a biofluid of the healthy breast: nipple aspirate fluid (NAF), and compared NAF miRNA expression patterns with publically available miRNA expression profiles of healthy breast tissue, breast milk, plasma and serum. Methods: MiRNA RT-qPCR profiling of NAF (n = 41) and serum (n = 23) samples from two healthy female cohorts was performed using the TaqMan OpenArray Human Advanced MicroRNA 754-Panel. MiRNA quantification data based on non-targeted or multi-targeted profiling techniques for breast tissue, breast milk, plasma and serum were retrieved from the literature by means of a systematic search. MiRNAs from each individual study were orderly ranked between 1 and 50, combined into an overall ranking per sample type and compared. Results: NAF expressed 11 unique miRNAs and shared 21/50 miRNAs with breast tissue. Seven miRNAs were shared between the five sample types. Overlap between sample types varied between 42% and 62%. Highly ranked NAF miRNAs have established roles in breast carcinogenesis. Conclusion: This is the first study to characterize and compare the unique physiological NAF-derived miRNA landscape with the physiological expression pattern in breast tissue, breast milk, plasma and serum. Breast-specific sources did not mutually overlap more than with systemic sources. Given their established role in carcinogenesis, NAF miRNA assessment could be a valuable tool in breast tumor diagnostics.
Collapse
|
19
|
Role of Slit2 upregulation in recurrent miscarriage through regulation of stromal decidualization. Placenta 2020; 103:1-9. [PMID: 33068960 DOI: 10.1016/j.placenta.2020.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/15/2020] [Accepted: 10/09/2020] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Knockout mouse model has shown a relationship between Slit2/Robo1 signalling and altered fertility. Altered expression by endometrial epithelium and trophoblast and is associated with the pathogenesis of pregnancy complications but few studies have investigated the expression of decidual Slit2 in miscarriage. METHODS Expression profiles of Slit2 and Robo1 were measured in human endometrial tissues during the menstrual cycle phases (n = 30), in decidua tissues from recurrent miscarriage (n = 20) and healthy control (n = 20) at 6-8 weeks of gestation. The hormonal regulation of Slit2/Robo1 expression and the role of Slit2/Robo1 signalling in decidualization was investigated in vitro, along with its effects on β-catenin and MET expression. RESULTS In human endometrium, Slit2 and Robo1 protein expression in stromal cells were decreased between the late-proliferative and early-secretory phase. In recurrent miscarriage patients, decidual expression Slit2 was increased and associated with lower expression of E-cadherin and higher level vimentin compared to controls. In vitro, the expression of Slit2 was downregulated by cAMP and progesterone in hESCs. Upregulation of Slit2 resulted in inhibition of cell decidualization and β-catenin translocation to nucleus. DISCUSSION This study indicates a functional role for Slit2 in endometrial stromal cell decidualization and the pathogenesis of recurrent miscarriage. Aberrant Increase in Slit2 expression may impairs decidualization of endometrial stromal cells leading to recurrent in recurrent miscarriage.
Collapse
|
20
|
Bhosle VK, Mukherjee T, Huang YW, Patel S, Pang BWF, Liu GY, Glogauer M, Wu JY, Philpott DJ, Grinstein S, Robinson LA. SLIT2/ROBO1-signaling inhibits macropinocytosis by opposing cortical cytoskeletal remodeling. Nat Commun 2020; 11:4112. [PMID: 32807784 PMCID: PMC7431850 DOI: 10.1038/s41467-020-17651-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/08/2020] [Indexed: 01/06/2023] Open
Abstract
Macropinocytosis is essential for myeloid cells to survey their environment and for growth of RAS-transformed cancer cells. Several growth factors and inflammatory stimuli are known to induce macropinocytosis, but its endogenous inhibitors have remained elusive. Stimulation of Roundabout receptors by Slit ligands inhibits directional migration of many cell types, including immune cells and cancer cells. We report that SLIT2 inhibits macropinocytosis in vitro and in vivo by inducing cytoskeletal changes in macrophages. In mice, SLIT2 attenuates the uptake of muramyl dipeptide, thereby preventing NOD2-dependent activation of NF-κB and consequent secretion of pro-inflammatory chemokine, CXCL1. Conversely, blocking the action of endogenous SLIT2 enhances CXCL1 secretion. SLIT2 also inhibits macropinocytosis in RAS-transformed cancer cells, thereby decreasing their survival in nutrient-deficient conditions which resemble tumor microenvironment. Our results identify SLIT2 as a physiological inhibitor of macropinocytosis and challenge the conventional notion that signals that enhance macropinocytosis negatively regulate cell migration, and vice versa.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Tapas Mukherjee
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Yi-Wei Huang
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Bo Wen Frank Pang
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- BenchSci, Suite 201, 559 College Street, Toronto, ON, M6G 1A9, Canada
| | - Guang-Ying Liu
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, 101 Elm Street, Toronto, ON, M5G 2L3, Canada
- Department of Dental Oncology and Maxillofacial Prosthetics, University Health Network, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
- Centre for Advanced Dental Research and Care, Mount Sinai Hospital, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Jane Y Wu
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, 290 Victoria Street, Toronto, ON, M5C 1N8, Canada
| | - Lisa A Robinson
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Division of Nephrology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Paediatrics, Faculty of Medicine, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
21
|
Najminejad H, Farhadihosseinabadi B, Dabaghian M, Dezhkam A, Rigi Yousofabadi E, Najminejad R, Abdollahpour-Alitappeh M, Karimi MH, Bagheri N, Mahi-Birjand M, Ghasemi N, Mazaheri M, Kalantar SM, Seifalian A, Sheikhha MH. Key Regulatory miRNAs and their Interplay with Mechanosensing and Mechanotransduction Signaling Pathways in Breast Cancer Progression. Mol Cancer Res 2020; 18:1113-1128. [PMID: 32430354 DOI: 10.1158/1541-7786.mcr-19-1229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/14/2020] [Accepted: 05/15/2020] [Indexed: 11/16/2022]
Abstract
According to the WHO, breast cancer is the most common cancer in women worldwide. Identification of underlying mechanisms in breast cancer progression is the main concerns of researches. The mechanical forces within the tumor microenvironment, in addition to biochemical stimuli such as different growth factors and cytokines, activate signaling cascades, resulting in various changes in cancer cell physiology. Cancer cell proliferation, invasiveness, migration, and, even, resistance to cancer therapeutic agents are changed due to activation of mechanotransduction signaling. The mechanotransduction signaling is frequently dysregulated in breast cancer, indicating its important role in cancer cell features. So far, a variety of experimental investigations have been conducted to determine the main regulators of the mechanotransduction signaling. Currently, the role of miRNAs has been well-defined in the cancer process through advances in molecular-based approaches. miRNAs are small groups of RNAs (∼22 nucleotides) that contribute to various biological events in cells. The central role of miRNAs in the regulation of various mediators involved in the mechanotransduction signaling has been well clarified over the last decade. Unbalanced expression of miRNAs is associated with different pathologic conditions. Overexpression and downregulation of certain miRNAs were found to be along with dysregulation of mechanotransduction signaling effectors. This study aimed to critically review the role of miRNAs in the regulation of mediators involved in the mechanosensing pathways and clarify how the cross-talk between miRNAs and their targets affect the cell behavior and physiology of breast cancer cells.
Collapse
Affiliation(s)
- Hamid Najminejad
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Behrouz Farhadihosseinabadi
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center (NRC), Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Dabaghian
- Research and Development Department, Razi Vaccine and serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Asiyeh Dezhkam
- Department of Midwifery, School of Nursing and Midwifery, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Reza Najminejad
- Department of Internal Medicine, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | | | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Motahareh Mahi-Birjand
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nasrin Ghasemi
- Abortion Research Centre, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahta Mazaheri
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mehdi Kalantar
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (Ltd), The London BioScience Innovation Centre, London, United Kingdom.
| | - Mohammad Hasan Sheikhha
- Genetics and Biotechnology Lab, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
22
|
Dong Y, Zheng Q, Wang Z, Lin X, You Y, Wu S, Wang Y, Hu C, Xie X, Chen J, Gao D, Zhao Y, Wu W, Liu Y, Ren Z, Chen R, Cui J. Higher matrix stiffness as an independent initiator triggers epithelial-mesenchymal transition and facilitates HCC metastasis. J Hematol Oncol 2019; 12:112. [PMID: 31703598 PMCID: PMC6839087 DOI: 10.1186/s13045-019-0795-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
Background Increased liver stiffness exerts a detrimental role in driving hepatocellular carcinoma (HCC) malignancy and progression, and indicates a high risk of unfavorable outcomes. However, it remains largely unknown how liver matrix stiffness as an independent cue triggers epithelial-mesenchymal transition (EMT) and facilitates HCC metastasis. Methods Buffalo rat HCC models with different liver stiffness backgrounds and an in vitro Col I-coated cell culture system with tunable stiffness were used in the study to explore the effects of matrix stiffness on EMT occurrence and its underlying molecular mechanism. Clinical significance of liver stiffness and key molecules required for stiffness-induced EMT were validated in HCC cohorts with different liver stiffness. Results HCC xenografts grown in higher stiffness liver exhibited worse malignant phenotypes and higher lung metastasis rate, suggesting that higher liver stiffness promotes HCC invasion and metastasis. Cell tests in vitro showed that higher matrix stiffness was able to strikingly strengthen malignant phenotypes and independently induce EMT occurrence in HCC cells, and three signaling pathways converging on Snail expression participated in stiffness-mediated effect on EMT including integrin-mediated S100A11 membrane translocation, eIF4E phosphorylation, and TGF β1 autocrine. Additionally, the key molecules required for stiffness-induced EMT were highly expressed in tumor tissues of HCC patients with higher liver stiffness and correlated with poor tumor differentiation and higher recurrence. Conclusions Higher matrix stiffness as an initiator triggers epithelial-mesenchymal transition (EMT) in HCC cells independently, and three signaling pathways converging on Snail expression contribute to this pathological process. This work highlights a significant role of biomechanical signal in triggering EMT and facilitating HCC invasion and metastasis.
Collapse
Affiliation(s)
- Yinying Dong
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Qiongdan Zheng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiahui Lin
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yang You
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Sifan Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yaohui Wang
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chao Hu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaoying Xie
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Jie Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Weizhong Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yinkun Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China.
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
23
|
Jiang S, Du Y, Liu D, He J, Huang Y, Qin K, Zhou X. Inhibitory Effect of Slit2-N on VEGF165-induced proliferation of vascular endothelia via Slit2-N-Robo4-Akt pathway in choroidal neovascularization. Cell Cycle 2019; 18:1241-1253. [PMID: 31081721 DOI: 10.1080/15384101.2019.1617005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Researches have been focusing on the role of Slit2 in angiogenesis, specifically in cell migration and vessel permeability. Nevertheless, the role of Slit2-N, the bioactive fragment of Slit2, in the proliferation of vascular endothelia in choroidal neovascularization and some related mechanisms have not been studied yet. Thus, our study aimed to explore the role of Slit2-N in proliferation of vascular endothelia and the related mechanisms in choroidal neovascularization. Fluorescein isothiocyanate perfusion and HE staining were performed to evaluate volumes of choroidal neovascularization lesions. The effect of Slit2-N on VEGF165-induced cell proliferation and some related mechanisms were detected by CCK8 assay, flow cytometry, siRNA transfection, and western blotting. We found that Slit2-N reduced volumes of laser-induced choroidal neovascularization networks in vivo. Results of the in vitro study showed Slit2-N reduced VEGF165-induced cell proliferation of both human umbilical vascular endothelial cells and human microvascular endothelial cells possibly via activation of AKT rather than that of ERK1/2. Additionally, Robo4, one of the receptors binding to Slit2-N, was involved in the inhibitory effect of Slit2-N. Generally, our findings revealed the inhibitory role of Slit2-N in proliferation of vascular endothelia and some related mechanisms, and presented some potential targets, molecules along Slit2-N-Robo4-AKT axis, to choroidal neovascularization therapy.
Collapse
Affiliation(s)
- Shaoqiu Jiang
- a Department of Ophthalmology , the Second Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b Chongqing Key Laboratory of Ophthalmology , Chongqing Eye Institute , Chongqing , China
| | - Yong Du
- a Department of Ophthalmology , the Second Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b Chongqing Key Laboratory of Ophthalmology , Chongqing Eye Institute , Chongqing , China
| | - Danning Liu
- a Department of Ophthalmology , the Second Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Junchi He
- c Department of Neurosurgery , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Yike Huang
- b Chongqing Key Laboratory of Ophthalmology , Chongqing Eye Institute , Chongqing , China.,d Department of Ophthalmology , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Ke Qin
- a Department of Ophthalmology , the Second Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b Chongqing Key Laboratory of Ophthalmology , Chongqing Eye Institute , Chongqing , China
| | - Xiyuan Zhou
- a Department of Ophthalmology , the Second Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b Chongqing Key Laboratory of Ophthalmology , Chongqing Eye Institute , Chongqing , China
| |
Collapse
|
24
|
Fan L, Wu Y, Wang J, He J, Han X. Sevoflurane inhibits the migration and invasion of colorectal cancer cells through regulating ERK/MMP-9 pathway by up-regulating miR-203. Eur J Pharmacol 2019; 850:43-52. [PMID: 30685432 DOI: 10.1016/j.ejphar.2019.01.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 02/08/2023]
Abstract
Surgery resection is the primary treatment for colorectal cancer (CRC) patients with the risk of cancer dissemination and metastasis. Sevoflurane is one inhalational anesthesia which regulates migration and invasion in varying cancers. However, the effect of sevoflurane on CRC cells and its mechanism remain poorly understood. In this study, SW620 and HCT116 cells were treated with different concentrations of sevoflurane for 6 h in vitro. We measured the effect of sevoflurane on cell survival, migration and invasion by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide or trans-well assays. Moreover, we explored the interaction between sevoflurane and miR-203 and Roundabout1 (Robo1) as well as the extracellular signal-regulated kinase (ERK) and matrix metalloproteinase-9 (MMP-9) pathway. Results showed that sevoflurane inhibited cell migration and invasion in SW620 and HCT116 cells in a concentration dependent manner. Moreover, different concentrations of sevoflurane suppressed the phosphorylation of ERK. miR-203 expression was impaired while sevoflurane reversed the expression of miR-203 in CRC cells. In addition, inhibition of miR-203 attenuated the inhibitory effect of sevoflurane on cell migration, invasion and phosphorylated ERK level. Notably, MMP-9, as a downstream of ERK, was involved in sevoflurane-mediated processes in CRC cells. Besides, Robo1 was indicated as a target of miR-203 and inhibited by sevoflurane treatment. These results indicated that sevoflurane suppressed cell migration and invasion through regulating ERK/MMP-9 pathway via miR-203/Robo1 in CRC cells, indicating important clinical implications for anesthetic agents to prevent metastasis in CRC.
Collapse
Affiliation(s)
- Lihua Fan
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China.
| | - Yini Wu
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China
| | - Jianping Wang
- Department of Anorectal Surgery, Wenzhou Medical University, The Fifth Affiliated Hospital, Lishui, Zhejiang 323000, China.
| | - Jiaqun He
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China
| | - Xin Han
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China
| |
Collapse
|
25
|
Guo Z, Cao Y. An lncRNA‑miRNA‑mRNA ceRNA network for adipocyte differentiation from human adipose‑derived stem cells. Mol Med Rep 2019; 19:4271-4287. [PMID: 30896814 PMCID: PMC6471198 DOI: 10.3892/mmr.2019.10067] [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] [Received: 06/12/2018] [Accepted: 01/02/2019] [Indexed: 01/08/2023] Open
Abstract
Human adipose tissue‑derived stromal stem cells (HASCs) represent a promising regenerative resource for breast reconstruction and augmentation. However, the mechanisms involved in inducing its adipogenic differentiation remain to be fully elucidated. The present study aimed to comprehensively investigate the expression changes in mRNAs, microRNAs (miRNAs) and long non‑coding (lnc)RNAs during the adipogenic differentiation of HASCs, and screen crucial lncRNA‑miRNA‑mRNA interaction axes using microarray datasets GSE57593, GSE25715 and GSE61302 collected from the Gene Expression Omnibus database. Following pretreatment, differentially expressed genes (DEGs), miRNAs (DE‑miRNAs) or lncRNAs (DE‑lncRNAs) between undifferentiated and differentiated HASCs were identified using the Linear Models for Microarray data method. A protein‑protein interaction (PPI) network was constructed for the DEGs based on protein databases, followed by module analysis. The 'lncRNA‑miRNA‑mRNA' competing endogenous RNA (ceRNA) network was constructed based on the interactions between miRNAs and mRNAs, lncRNAs and miRNAs predicted by the miRWalk and lnCeDB databases. The underlying functions of mRNAs were predicted using the clusterProfiler package. In the present study, 905 DEGs, 36 DE‑miRNAs and 577 DE‑lncRNAs were screened between undifferentiated HASCs and differentiated adipocyte cells. PPI network analysis demonstrated that LEP may be a hub gene, which was also enriched in significant module 5. LEP was predicted to be involved in the Janus kinase‑signal transducer and activator of transcription signaling pathway, and the regulation of inflammatory response. The upregulation of LEP was regulated by downregulated hsa‑miRNA (miR)‑130b‑5p and hsa‑miR‑23a‑5p (or hsa‑miR‑302d‑3p). These miRNAs also respectively interacted with RP11‑552F3.9 (or RP11‑15A1.7), ultimately forming the ceRNA axes. In conclusion, the present study revealed that the RP11‑552F3.9 (RP11‑15A1.7)‑hsa‑miR‑130b‑5p/hsa‑miR‑23a‑5p (hsa‑miR‑302d‑3p)‑LEP interaction axes may be crucial for inducing the adipogenic differentiation of HASCs via involvement in inflammation.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Breast Surgery, The No. 3 Hospital of Nanchang, Nanchang, Jiangxi 330000, P.R. China
| | - Yali Cao
- Department of Breast Surgery, The No. 3 Hospital of Nanchang, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
26
|
Beamish IV, Hinck L, Kennedy TE. Making Connections: Guidance Cues and Receptors at Nonneural Cell-Cell Junctions. Cold Spring Harb Perspect Biol 2018; 10:a029165. [PMID: 28847900 PMCID: PMC6211390 DOI: 10.1101/cshperspect.a029165] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The field of axon guidance was revolutionized over the past three decades by the identification of highly conserved families of guidance cues and receptors. These proteins are essential for normal neural development and function, directing cell and axon migration, neuron-glial interactions, and synapse formation and plasticity. Many of these genes are also expressed outside the nervous system in which they influence cell migration, adhesion and proliferation. Because the nervous system develops from neural epithelium, it is perhaps not surprising that these guidance cues have significant nonneural roles in governing the specialized junctional connections between cells in polarized epithelia. The following review addresses roles for ephrins, semaphorins, netrins, slits and their receptors in regulating adherens, tight, and gap junctions in nonneural epithelia and endothelia.
Collapse
Affiliation(s)
- Ian V Beamish
- Department of Neurology & Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Lindsay Hinck
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California 95064
| | - Timothy E Kennedy
- Department of Neurology & Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| |
Collapse
|
27
|
Abstract
miR-203 is an epigenetically silenced tumor-suppressive microRNA in tumors. This study was designed to investigate the effects of miR-203 on the proliferation, migration, invasion, and apoptosis of bladder cancer (BCa) cells. The expression levels of miR-203 in BCa tissues, normal adjacent tissues, and BCa cell lines were detected. BCa cells were transfected with miR-203 mimic and inhibitor to investigate the effect of miR-203 on cell functions and the epithelial-mesenchymal transition (EMT). Cotransfection with miR-203 inhibitor and shRNA of the predicted target gene Twist1 (si-Twist1) was performed to investigate the target relationship of miR-203 and Twist1. The effects of knockdown of Twist1 on cell functions were also investigated. The expression of miR-203 was significantly reduced in BCa tissues and cells, in comparison with the control. miR-203 mimic significantly reduced cell viability, invasion, migration, and EMT, and enhanced cell apoptosis. On the contrary, miR-203 inhibitor showed the opposite results. However, the administration of si-Twist1 cancelled the effect of miR-203 inhibitor on cell proliferation, apoptosis, invasion, and migration. These demonstrated that miR-203 may function as a tumor-suppressive microRNA in BCa by negatively targeting Twist1. Both Twist1 and miR-203 might be explored as potential targets for studying the mechanism related to BCa pathogenesis and therapy.
Collapse
Affiliation(s)
- Jie Shen
- Department of Urology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Jianhua Zhang
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Minhui Xiao
- Department of Urology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Junfeng Yang
- Department of Urology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| | - Ningnan Zhang
- Department of Urology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, P.R. China
| |
Collapse
|
28
|
Wu S, Zheng Q, Xing X, Dong Y, Wang Y, You Y, Chen R, Hu C, Chen J, Gao D, Zhao Y, Wang Z, Xue T, Ren Z, Cui J. Matrix stiffness-upregulated LOXL2 promotes fibronectin production, MMP9 and CXCL12 expression and BMDCs recruitment to assist pre-metastatic niche formation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:99. [PMID: 29728125 PMCID: PMC5935912 DOI: 10.1186/s13046-018-0761-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/13/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Higher matrix stiffness affects biological behavior of tumor cells, regulates tumor-associated gene/miRNA expression and stemness characteristic, and contributes to tumor invasion and metastasis. However, the linkage between higher matrix stiffness and pre-metastatic niche in hepatocellular carcinoma (HCC) is still largely unknown. METHODS We comparatively analyzed the expressions of LOX family members in HCC cells grown on different stiffness substrates, and speculated that the secreted LOXL2 may mediate the linkage between higher matrix stiffness and pre-metastatic niche. Subsequently, we investigated the underlying molecular mechanism by which matrix stiffness induced LOXL2 expression in HCC cells, and explored the effects of LOXL2 on pre-metastatic niche formation, such as BMCs recruitment, fibronectin production, MMPs and CXCL12 expression, cell adhesion, etc. RESULTS: Higher matrix stiffness significantly upregulated LOXL2 expression in HCC cells, and activated JNK/c-JUN signaling pathway. Knockdown of integrin β1 and α5 suppressed LOXL2 expression and reversed the activation of above signaling pathway. Additionally, JNK inhibitor attenuated the expressions of p-JNK, p-c-JUN, c-JUN and LOXL2, and shRNA-c-JUN also decreased LOXL2 expression. CM-LV-LOXL2-OE and rhLOXL2 upregulated MMP9 expression and fibronectin production obviously in lung fibroblasts. Moreover, activation of Akt pathway contributed to LOXL2-induced fibronectin upregulation. LOXL2 in CM as chemoattractant increased motility and invasion of BMCs, implicating a significant role of LOXL2 in BMCs recruitment. Except that, CM-LV-LOXL2-OE as chemoattractant also increased the number of migrated HCC cells, and improved chemokine CXCL12 expression in lung fibroblasts. The number of HCC cells adhered to surface of lung fibroblasts treated with CM-LV-LOXL2-OE was remarkably higher than that of the control cells. These results indicated that the secreted LOXL2 facilitated the motility of HCC cells and strengthened CTCs settlement on the remodeled matrix "soil". CONCLUSION Integrin β1/α5/JNK/c-JUN signaling pathway participates in higher matrix stiffness-induced LOXL2 upregulation in HCC cells. The secreted LOXL2 promotes fibronectin production, MMP9 and CXCL12 expression and BMDCs recruitment to assist pre-metastatic niche formation.
Collapse
Affiliation(s)
- Sifan Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Qiongdan Zheng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Xiaoxia Xing
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yinying Dong
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yaohui Wang
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yang You
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Chao Hu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jie Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Tongchun Xue
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
29
|
Muncie JM, Weaver VM. The Physical and Biochemical Properties of the Extracellular Matrix Regulate Cell Fate. Curr Top Dev Biol 2018; 130:1-37. [PMID: 29853174 DOI: 10.1016/bs.ctdb.2018.02.002] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The extracellular matrix is a complex network of hydrated macromolecular proteins and sugars that, in concert with bound soluble factors, comprise the acellular stromal microenvironment of tissues. Rather than merely providing structural information to cells, the extracellular matrix plays an instructive role in development and is critical for the maintenance of tissue homeostasis. In this chapter, we review the composition of the extracellular matrix and summarize data illustrating its importance in embryogenesis, tissue-specific development, and stem cell differentiation. We discuss how the biophysical and biochemical properties of the extracellular matrix ligate specific transmembrane receptors to activate intracellular signaling that alter cell shape and cytoskeletal dynamics to modulate cell growth and viability, and direct cell migration and cell fate. We present examples describing how the extracellular matrix functions as a highly complex physical and chemical entity that regulates tissue organization and cell behavior through a dynamic and reciprocal dialogue with the cellular constituents of the tissue. We suggest that the extracellular matrix not only transmits cellular and tissue-level force to shape development and tune cellular activities that are key for coordinated tissue behavior, but that it is itself remodeled such that it temporally evolves to maintain the integrated function of the tissue. Accordingly, we argue that perturbations in extracellular matrix composition and structure compromise key developmental events and tissue homeostasis, and promote disease.
Collapse
Affiliation(s)
- Jonathon M Muncie
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA, United States; Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, United States
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
30
|
Northcott JM, Dean IS, Mouw JK, Weaver VM. Feeling Stress: The Mechanics of Cancer Progression and Aggression. Front Cell Dev Biol 2018. [PMID: 29541636 PMCID: PMC5835517 DOI: 10.3389/fcell.2018.00017] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The tumor microenvironment is a dynamic landscape in which the physical and mechanical properties evolve dramatically throughout cancer progression. These changes are driven by enhanced tumor cell contractility and expansion of the growing tumor mass, as well as through alterations to the material properties of the surrounding extracellular matrix (ECM). Consequently, tumor cells are exposed to a number of different mechanical inputs including cell–cell and cell-ECM tension, compression stress, interstitial fluid pressure and shear stress. Oncogenes engage signaling pathways that are activated in response to mechanical stress, thereby reworking the cell's intrinsic response to exogenous mechanical stimuli, enhancing intracellular tension via elevated actomyosin contraction, and influencing ECM stiffness and tissue morphology. In addition to altering their intracellular tension and remodeling the microenvironment, cells actively respond to these mechanical perturbations phenotypically through modification of gene expression. Herein, we present a description of the physical changes that promote tumor progression and aggression, discuss their interrelationship and highlight emerging therapeutic strategies to alleviate the mechanical stresses driving cancer to malignancy.
Collapse
Affiliation(s)
- Josette M Northcott
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Ivory S Dean
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Janna K Mouw
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, United States.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
31
|
Broders-Bondon F, Nguyen Ho-Bouldoires TH, Fernandez-Sanchez ME, Farge E. Mechanotransduction in tumor progression: The dark side of the force. J Cell Biol 2018; 217:1571-1587. [PMID: 29467174 PMCID: PMC5940296 DOI: 10.1083/jcb.201701039] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Broders-Bondon et al. review the pathological mechanical properties of tumor tissues and how abnormal mechanical signals result in oncogenic biochemical signals during tumor progression. Cancer has been characterized as a genetic disease, associated with mutations that cause pathological alterations of the cell cycle, adhesion, or invasive motility. Recently, the importance of the anomalous mechanical properties of tumor tissues, which activate tumorigenic biochemical pathways, has become apparent. This mechanical induction in tumors appears to consist of the destabilization of adult tissue homeostasis as a result of the reactivation of embryonic developmental mechanosensitive pathways in response to pathological mechanical strains. These strains occur in many forms, for example, hypervascularization in late tumors leads to high static hydrodynamic pressure that can promote malignant progression through hypoxia or anomalous interstitial liquid and blood flow. The high stiffness of tumors directly induces the mechanical activation of biochemical pathways enhancing the cell cycle, epithelial–mesenchymal transition, and cell motility. Furthermore, increases in solid-stress pressure associated with cell hyperproliferation activate tumorigenic pathways in the healthy epithelial cells compressed by the neighboring tumor. The underlying molecular mechanisms of the translation of a mechanical signal into a tumor inducing biochemical signal are based on mechanically induced protein conformational changes that activate classical tumorigenic signaling pathways. Understanding these mechanisms will be important for the development of innovative treatments to target such mechanical anomalies in cancer.
Collapse
Affiliation(s)
- Florence Broders-Bondon
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Thanh Huong Nguyen Ho-Bouldoires
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Maria-Elena Fernandez-Sanchez
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Emmanuel Farge
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| |
Collapse
|
32
|
Mechanosensitive adhesion complexes in epithelial architecture and cancer onset. Curr Opin Cell Biol 2018; 50:42-49. [PMID: 29454273 DOI: 10.1016/j.ceb.2018.01.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/10/2018] [Accepted: 01/30/2018] [Indexed: 01/09/2023]
Abstract
Mechanical signals from the extracellular space are paramount to coordinate tissue morphogenesis and homeostasis. Although there is a wide variety of cellular mechanisms involved in transducing extracellular forces, recent literature emphasizes the central role of two main adhesion complexes in epithelial mechanosensitive processes: focal adhesions and adherens junctions. These biomechanical sensors can decode physical signals such as matrix stiffness or intercellular tension into a wide range of coordinated cellular responses, which can impact cell differentiation, migration, and proliferation. Communication between cells and their microenvironment plays a pivotal role both in physiological and pathological conditions. Here we summarize the most recent findings on the biology of these mechanotransduction pathways in epithelial cells, highlighting the extensive amount of biological processes coordinated by cell-matrix and cell-cell adhesion complexes.
Collapse
|
33
|
Mao X, Fan C, Yu X, Chen B, Jin F. DDEFL1 correlated with Rho GTPases activity in breast cancer. Oncotarget 2017; 8:112487-112497. [PMID: 29348842 PMCID: PMC5762527 DOI: 10.18632/oncotarget.22095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/22/2017] [Indexed: 01/13/2023] Open
Abstract
DDEFL1 is related to maintaining a limiting amount of ARF6 in GTP-loaded form by accelerating its GTP hydrolysis activity, which has been implicated in hepatocellular cancer pathogenesis and lung cancer development. We investigated DDEFL1 expression in breast cancer and paired normal breast tissues by immunohistochemistry and found that DDEFL1 expression was significantly associated with tumor size, lymph node metastasis, high content of elastosis and TNM stage but not with menopausal status or age. We detected the mRNA and protein expression of DDEFL1 in breast cancer cell lines by Western blotting and quantitative real-time PCR (qRT-PCR). DDEFL1 was obvious in MDA-MB-435s and MDA-MB-231 but very weak in ZR-75-1. Further experiments were conducted to evaluate the effect of DDEFL1 small interfering RNA (siRNA) transfection on the biological behavior of MDA-MB-231. After transfection, the effects of DDEFL1 inhibition on expression of mRNA and protein were also analyzed by Western blotting and qRT-PCR. Increased apoptosis and invasive ability, decreased cellular proliferation was found in MDA-MB-231 with successful DDEFL1 siRNA transient transfection (p < 0.05). Western blotting and qRT-PCR results showed that the DDEFL1 inhibition up-regulated Caspase-3, Apaf-1, cytochrome c, and Bax expression and down-regulated Bcl-2 expression. The DDEFL1 inhibition also down-regulated the mRNA and protein expression of Rho, CDC42 and Rac1. Our study provided a functional linkage through DDEFL1 with breast cancer biological behaviours by Rho GTPases. Possible implication of our main finding for the DDEFL1 role in breast cancer and the downstream signaling pathways for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xiaoyun Mao
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, China
| | - Xinmiao Yu
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Bo Chen
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Feng Jin
- Department of Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| |
Collapse
|
34
|
Abstract
Biomechanical and biochemical cues within a tissue collaborate across length scales to direct cell fate during development and are critical for the maintenance of tissue homeostasis. Loss of tensional homeostasis in a tissue not only accompanies malignancy but may also contribute to oncogenic transformation. High mechanical stress in solid tumors can impede drug delivery and may additionally drive tumor progression and promote metastasis. Mechanistically, biomechanical forces can drive tumor aggression by inducing a mesenchymal-like switch in transformed cells so that they attain tumor-initiating or stem-like cell properties. Given that cancer stem cells have been linked to metastasis and treatment resistance, this raises the intriguing possibility that the elevated tissue mechanics in tumors could promote their aggression by programming their phenotype toward that exhibited by a stem-like cell.Significance: Recent findings argue that mechanical stress and elevated mechanosignaling foster malignant transformation and metastasis. Prolonged corruption of tissue tension may drive tumor aggression by altering cell fate specification. Thus, strategies that could reduce tumor mechanics might comprise effective approaches to prevent the emergence of treatment-resilient metastatic cancers. Cancer Discov; 7(11); 1224-37. ©2017 AACR.
Collapse
Affiliation(s)
- Jason J Northey
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, California
| | - Laralynne Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, California
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, California.
- Departments of Anatomy, Bioengineering and Therapeutic Sciences, and Radiation Oncology, and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and The Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California
| |
Collapse
|
35
|
Yu CK, Xu T, Assoian RK, Rader DJ. Mining the Stiffness-Sensitive Transcriptome in Human Vascular Smooth Muscle Cells Identifies Long Noncoding RNA Stiffness Regulators. Arterioscler Thromb Vasc Biol 2017; 38:164-173. [PMID: 29051139 DOI: 10.1161/atvbaha.117.310237] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Vascular extracellular matrix stiffening is a risk factor for aortic and coronary artery disease. How matrix stiffening regulates the transcriptome profile of human aortic and coronary vascular smooth muscle cells (VSMCs) is not well understood. Furthermore, the role of long noncoding RNAs (lncRNAs) in the cellular response to stiffening has never been explored. This study characterizes the stiffness-sensitive (SS) transcriptome of human aortic and coronary VSMCs and identifies potential key lncRNA regulators of stiffness-dependent VSMC functions. APPROACH AND RESULTS Aortic and coronary VSMCs were cultured on hydrogel substrates mimicking physiological and pathological extracellular matrix stiffness. Total RNAseq was performed to compare the SS transcriptome profiles of aortic and coronary VSMCs. We identified 3098 genes (2842 protein coding and 157 lncRNA) that were stiffness sensitive in both aortic and coronary VSMCs (false discovery rate <1%). Hierarchical clustering revealed that aortic and coronary VSMCs grouped by stiffness rather than cell origin. Conservation analyses also revealed that SS genes were more conserved than stiffness-insensitive genes. These VSMC SS genes were less tissue-type specific and expressed in more tissues than stiffness-insensitive genes. Using unbiased systems analyses, we identified MALAT1 as an SS lncRNA that regulates stiffness-dependent VSMC proliferation and migration in vitro and in vivo. CONCLUSIONS This study provides the transcriptomic landscape of human aortic and coronary VSMCs in response to extracellular matrix stiffness and identifies novel SS human lncRNAs. Our data suggest that the SS transcriptome is evolutionarily important to VSMCs function and that SS lncRNAs can act as regulators of stiffness-dependent phenotypes.
Collapse
MESH Headings
- Aorta/metabolism
- Aorta/pathology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cluster Analysis
- Computational Biology/methods
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Data Mining/methods
- Extracellular Matrix/genetics
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Humans
- Hydrogels
- Mechanotransduction, Cellular
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Transcriptome
- Vascular Stiffness
Collapse
Affiliation(s)
- Christopher K Yu
- From the Perelman School of Medicine (C.K.Y.), Department of Systems Pharmacology and Translational Therapeutics (T.X., R.K.A.), Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics (T.X., R.K.A.), and Departments of Genetics, Medicine, and Pediatrics, Perelman School of Medicine (D.J.R.), University of Pennsylvania, Philadelphia
- This manuscript was sent to Zahi Fayad, Consulting Editor, for review by expert referees, editorial decision, and final disposition
| | - Tina Xu
- From the Perelman School of Medicine (C.K.Y.), Department of Systems Pharmacology and Translational Therapeutics (T.X., R.K.A.), Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics (T.X., R.K.A.), and Departments of Genetics, Medicine, and Pediatrics, Perelman School of Medicine (D.J.R.), University of Pennsylvania, Philadelphia
- This manuscript was sent to Zahi Fayad, Consulting Editor, for review by expert referees, editorial decision, and final disposition
| | - Richard K Assoian
- From the Perelman School of Medicine (C.K.Y.), Department of Systems Pharmacology and Translational Therapeutics (T.X., R.K.A.), Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics (T.X., R.K.A.), and Departments of Genetics, Medicine, and Pediatrics, Perelman School of Medicine (D.J.R.), University of Pennsylvania, Philadelphia
- This manuscript was sent to Zahi Fayad, Consulting Editor, for review by expert referees, editorial decision, and final disposition
| | - Daniel J Rader
- From the Perelman School of Medicine (C.K.Y.), Department of Systems Pharmacology and Translational Therapeutics (T.X., R.K.A.), Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics (T.X., R.K.A.), and Departments of Genetics, Medicine, and Pediatrics, Perelman School of Medicine (D.J.R.), University of Pennsylvania, Philadelphia.
- This manuscript was sent to Zahi Fayad, Consulting Editor, for review by expert referees, editorial decision, and final disposition.
| |
Collapse
|
36
|
Northey JJ, Przybyla L, Weaver VM. Tissue Force Programs Cell Fate and Tumor Aggression. Cancer Discov 2017; 7:1224-1237. [PMID: 29038232 DOI: 10.1158/2159-8290.cd-16-0733] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 06/07/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
Biomechanical and biochemical cues within a tissue collaborate across length scales to direct cell fate during development and are critical for the maintenance of tissue homeostasis. Loss of tensional homeostasis in a tissue not only accompanies malignancy but may also contribute to oncogenic transformation. High mechanical stress in solid tumors can impede drug delivery and may additionally drive tumor progression and promote metastasis. Mechanistically, biomechanical forces can drive tumor aggression by inducing a mesenchymal-like switch in transformed cells so that they attain tumor-initiating or stem-like cell properties. Given that cancer stem cells have been linked to metastasis and treatment resistance, this raises the intriguing possibility that the elevated tissue mechanics in tumors could promote their aggression by programming their phenotype toward that exhibited by a stem-like cell.Significance: Recent findings argue that mechanical stress and elevated mechanosignaling foster malignant transformation and metastasis. Prolonged corruption of tissue tension may drive tumor aggression by altering cell fate specification. Thus, strategies that could reduce tumor mechanics might comprise effective approaches to prevent the emergence of treatment-resilient metastatic cancers. Cancer Discov; 7(11); 1224-37. ©2017 AACR.
Collapse
Affiliation(s)
- Jason J Northey
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, California
| | - Laralynne Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, California
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco (UCSF), San Francisco, California. .,Departments of Anatomy, Bioengineering and Therapeutic Sciences, and Radiation Oncology, and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and The Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California
| |
Collapse
|
37
|
Zanotelli MR, Bordeleau F, Reinhart-King CA. Subcellular regulation of cancer cell mechanics. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017. [DOI: 10.1016/j.cobme.2017.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
38
|
Miroshnikova YA, Mouw JK, Barnes JM, Pickup MW, Lakins JN, Kim Y, Lobo K, Persson AI, Reis GF, McKnight TR, Holland EC, Phillips JJ, Weaver VM. Tissue mechanics promote IDH1-dependent HIF1α-tenascin C feedback to regulate glioblastoma aggression. Nat Cell Biol 2016; 18:1336-1345. [PMID: 27820599 PMCID: PMC5361403 DOI: 10.1038/ncb3429] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
Increased overall survival for patients with glioma brain tumours is associated with mutations in the metabolic regulator isocitrate dehydrogenase 1 (IDH1). Gliomas develop within a mechanically challenged microenvironment that is characterized by a dense extracellular matrix (ECM) that compromises vascular integrity to induce hypoxia and activate HIF1α. We found that glioma aggression and patient prognosis correlate with HIF1α levels and the stiffness of a tenascin C (TNC)-enriched ECM. Gain- and loss-of-function xenograft manipulations demonstrated that a mutant IDH1 restricts glioma aggression by reducing HIF1α-dependent TNC expression to decrease ECM stiffness and mechanosignalling. Recurrent IDH1-mutant patient gliomas had a stiffer TNC-enriched ECM that our studies attributed to reduced miR-203 suppression of HIF1α and TNC mediated via a tension-dependent positive feedback loop. Thus, our work suggests that elevated ECM stiffness can independently foster glioblastoma aggression and contribute to glioblastoma recurrence via bypassing the protective activity of IDH1 mutational status.
Collapse
Affiliation(s)
- Yekaterina A. Miroshnikova
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | - Janna K. Mouw
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | - J. Matthew Barnes
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | - Michael W. Pickup
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | - Johnathan N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | - Youngmi Kim
- Division of Human Biology and Solid Tumor Translational Research, Fred Hutchinson Cancer Research Center, Department of Neurosurgery and Alvord Brain Tumor Center, University of Washington, Seattle, Washington 98109, USA
| | - Khadjia Lobo
- Magnetic Resonance Science Center, Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94143, USA
| | - Anders I. Persson
- Department of Neurology, University of California, San Francisco, California 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California 94158, USA
- Brain Tumor Research Center, Helen Diller Family Cancer Research Center, University of California San Francisco, San Francisco, California 94143, USA
- UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California San Francisco, San Francisco, California 94143, USA
| | - Gerald F. Reis
- Department of Pathology, University of California, San Francisco, California 94143, USA
| | - Tracy R. McKnight
- Magnetic Resonance Science Center, Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California 94143, USA
| | - Eric C. Holland
- Division of Human Biology and Solid Tumor Translational Research, Fred Hutchinson Cancer Research Center, Department of Neurosurgery and Alvord Brain Tumor Center, University of Washington, Seattle, Washington 98109, USA
| | - Joanna J. Phillips
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California 94158, USA
- Brain Tumor Research Center, Helen Diller Family Cancer Research Center, University of California San Francisco, San Francisco, California 94143, USA
- UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California San Francisco, San Francisco, California 94143, USA
- Department of Pathology, University of California, San Francisco, California 94143, USA
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
- Department of Anatomy and Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California 94143, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
39
|
Xiaoxianggou attenuates atherosclerotic plaque formation in endogenous high Ang II ApoE(-/-) mice via the inhibition of miR-203 on the expression of Ets-2 in endothelial cells. Biomed Pharmacother 2016; 82:173-9. [PMID: 27470353 DOI: 10.1016/j.biopha.2016.04.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 04/29/2016] [Accepted: 04/29/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Atherosclerosis is a chronic immune-inflammatory disorder and one of the leading causes responsible for cardiovascular morbidity and mortality. Traditional Chinese medicine treatment with multi-targets has shown prospects for the therapeutic effect on atherosclerosis. Thus, this study aims to investigate whether xiaoxianggou has benefit for reducing the atherosclerotic plaque area in endogenous high Ang II ApoE(-/-) mice and investigated the underlying mechanisms. METHODS Endogenous high Ang II ApoE(-/-) mice model was generated by using two kidney one clip (2K1C). All mice were treated by intragastric administration with xiaoxianggou two times a week for 16 weeks. En face plaque area was analyzed by oil-red O staining. Serum anti-OxLDL antibodies were measured by ELISA assay. Expression of miR-203 and Ets-2 were evaluated using qRT-RCR and western blotting analysis, respectively. RESULTS This study revealed that xiaoxianggou treatment dose-dependently reduced the atherosclerotic plaque area and serum autoantibodies against oxLDL, elevated miR-203 expression and reduced Ets-2 expression in endogenous high Ang II ApoE(-/-) mice. In primary arterial ECs, Xiaoxianggou reverses the reduced miR-203 expression and the elevated Ets-2 expression induced by AngII, which was further recovered by miR-203 inhibitor. Additionally, miR-203 regulated the expression of Ets-2 by targeting Ets-2-3' UTR. Moreover, miR-203 inhibitor reversed the reduction of atherosclerotic lesion area induced by Xiaoxianggou. CONCLUSIONS These findings present that xiaoxianggou plays an anti-atherosclerotic role in endogenous high Ang II ApoE(-/-) mice model, which is partly due to its antioxidant actions against atherosclerosis and the inhibition of miR-203 on the expression of Ets-2 in endothelial cells.
Collapse
|
40
|
Short B. ROBO-cop protects cells from stiff environment. J Biophys Biochem Cytol 2016. [DOI: 10.1083/jcb.2126iti2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|