101
|
Cell Cytoskeleton and Stiffness Are Mechanical Indicators of Organotropism in Breast Cancer. BIOLOGY 2021; 10:biology10040259. [PMID: 33805866 PMCID: PMC8064360 DOI: 10.3390/biology10040259] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary Cancer cell dissemination exhibits organ preference or organotropism. Although the influence of intrinsic biochemical factors on organotropism has been intensely studied, little is known about the roles of mechanical properties of metastatic cancer cells. Our study suggests that there may be a correlation between cell cytoskeleton/stiffness and organotropism. We find that the cytoskeleton and stiffness of breast cancer cell subpopulations with different metastatic preference match the mechanics of the metastasized organs. The modification of cell cytoskeleton significantly influences the organotropism-related gene expression pattern and mechanoresponses on soft substrates which mimic brain tissue stiffness. These findings highlight the key role of cell cytoskeleton in specific organ metastasis, which may not only reflect but also impact the metastatic organ preference. Abstract Tumor metastasis involves the dissemination of tumor cells from the primary lesion to other organs and the subsequent formation of secondary tumors, which leads to the majority of cancer-related deaths. Clinical findings show that cancer cell dissemination is not random but exhibits organ preference or organotropism. While intrinsic biochemical factors of cancer cells have been extensively studied in organotropism, much less is known about the role of cell cytoskeleton and mechanics. Herein, we demonstrate that cell cytoskeleton and mechanics are correlated with organotropism. The result of cell stiffness measurements shows that breast cancer cells with bone tropism are much stiffer with enhanced F-actin, while tumor cells with brain tropism are softer with lower F-actin than their parental cells. The difference in cellular stiffness matches the difference in the rigidity of their metastasized organs. Further, disrupting the cytoskeleton of breast cancer cells with bone tropism not only elevates the expressions of brain metastasis-related genes but also increases cell spreading and proliferation on soft substrates mimicking the stiffness of brain tissue. Stabilizing the cytoskeleton of cancer cells with brain tropism upregulates bone metastasis-related genes while reduces the mechanoadaptation ability on soft substrates. Taken together, these findings demonstrate that cell cytoskeleton and biophysical properties of breast cancer subpopulations correlate with their metastatic preference in terms of gene expression pattern and mechanoadaptation ability, implying the potential role of cell cytoskeleton in organotropism.
Collapse
|
102
|
Link PA, Heise RL, Weinberg SH. Cellular mitosis predicts vessel stability in a mechanochemical model of sprouting angiogenesis. Biomech Model Mechanobiol 2021; 20:1195-1208. [PMID: 33715101 DOI: 10.1007/s10237-021-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 02/22/2021] [Indexed: 11/28/2022]
Abstract
Angiogenesis, the formation of new vessels, occurs in both developmental and pathological contexts. Prior research has investigated vessel formation to identify cellular phenotypes and dynamics associated with angiogenic disease. One major family of proteins involved in angiogenesis are the Rho GTPases, which govern function related to cellular elongation, migration, and proliferation. Using a mechanochemical model coupling Rho GTPase activity and cellular and intercellular mechanics, we investigate the role of cellular mitosis on sprouting angiogenesis. Mitosis-GTPase synchronization was not a strong predictor of GTPase and thus vessel signaling instability, whereas the location of mitotic events was predicted to alter GTPase cycling instabilities. Our model predicts that middle stalk cells undergoing mitosis introduce irregular dynamics in GTPase cycling and may provide a source of aberrant angiogenesis. We also find that cellular and junctional tension exhibit spatial heterogeneity through the vessel, and that tension feedback, specifically in stalk cells, tends to increase the maximum forces generated in the vessel.
Collapse
Affiliation(s)
- Patrick A Link
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Seth H Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
103
|
Differential Splicing of Skipped-exons Predicts Drug Response in Cancer Cell Lines. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:901-912. [PMID: 33662622 PMCID: PMC9402787 DOI: 10.1016/j.gpb.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/07/2019] [Accepted: 08/23/2019] [Indexed: 12/02/2022]
Abstract
Alternative splicing of pre-mRNA transcripts is an important regulatory mechanism that increases the diversity of gene products in eukaryotes. Various studies have linked specific transcript isoforms to altered drug response in cancer; however, few algorithms have incorporated splicing information into drug response prediction. In this study, we evaluated whether basal-level splicing information could be used to predict drug sensitivity by constructing doxorubicin-sensitivity classification models with splicing and expression data. We detailed splicing differences between sensitive and resistant cell lines by implementing quasi-binomial generalized linear modeling (QBGLM) and found altered inclusion of 277 skipped exons. We additionally conducted RNA-binding protein (RBP) binding motif enrichment and differential expression analysis to characterize cis- and trans-acting elements that potentially influence doxorubicin response-mediating splicing alterations. Our results showed that a classification model built with skipped exon data exhibited strong predictive power. We discovered an association between differentially spliced events and epithelial-mesenchymal transition (EMT) and observed motif enrichment, as well as differential expression of RBFOX and ELAVL RBP family members. Our work demonstrates the potential of incorporating splicing data into drug response algorithms and the utility of a QBGLM approach for fast, scalable identification of relevant splicing differences between large groups of samples.
Collapse
|
104
|
Barcelona‐Estaje E, Dalby MJ, Cantini M, Salmeron‐Sanchez M. You Talking to Me? Cadherin and Integrin Crosstalk in Biomaterial Design. Adv Healthc Mater 2021; 10:e2002048. [PMID: 33586353 DOI: 10.1002/adhm.202002048] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/20/2021] [Indexed: 12/21/2022]
Abstract
While much work has been done in the design of biomaterials to control integrin-mediated adhesion, less emphasis has been put on functionalization of materials with cadherin ligands. Yet, cell-cell contacts in combination with cell-matrix interactions are key in driving embryonic development, collective cell migration, epithelial to mesenchymal transition, and cancer metastatic processes, among others. This review focuses on the incorporation of both cadherin and integrin ligands in biomaterial design, to promote what is called the "adhesive crosstalk." First, the structure and function of cadherins and their role in eliciting mechanotransductive processes, by themselves or in combination with integrin mechanosensing, are introduced. Then, biomaterials that mimic cell-cell interactions, and recent applications to get insights in fundamental biology and tissue engineering, are critically discussed.
Collapse
Affiliation(s)
- Eva Barcelona‐Estaje
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | - Matthew J. Dalby
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | - Marco Cantini
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | | |
Collapse
|
105
|
Islam MS, Molley TG, Ireland J, Kruzic JJ, Kilian KA. Magnetic Nanocomposite Hydrogels for Directing Myofibroblast Activity in Adipose‐Derived Stem Cells. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Md Shariful Islam
- School of Materials Science and Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - Thomas G. Molley
- School of Materials Science and Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - Jake Ireland
- School of Chemistry Australian Centre for Nanomedicine The University of New South Wales Sydney NSW 2052 Australia
| | - Jamie J. Kruzic
- School of Mechanical and Manufacturing Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - Kristopher A. Kilian
- School of Materials Science and Engineering The University of New South Wales Sydney NSW 2052 Australia
- School of Chemistry Australian Centre for Nanomedicine The University of New South Wales Sydney NSW 2052 Australia
| |
Collapse
|
106
|
McCreery KP, Xu X, Scott AK, Fajrial AK, Calve S, Ding X, Neu CP. Nuclear Stiffness Decreases with Disruption of the Extracellular Matrix in Living Tissues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006699. [PMID: 33470544 PMCID: PMC7891867 DOI: 10.1002/smll.202006699] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/04/2020] [Indexed: 05/04/2023]
Abstract
Reciprocal interactions between the cell nucleus and the extracellular matrix lead to macroscale tissue phenotype changes. However, little is known about how the extracellular matrix environment affects gene expression and cellular phenotype in the native tissue environment. Here, it is hypothesized that enzymatic disruption of the tissue matrix results in a softer tissue, affecting the stiffness of embedded cell and nuclear structures. The aim is to directly measure nuclear mechanics without perturbing the native tissue structure to better understand nuclear interplay with the cell and tissue microenvironments. To accomplish this, an atomic force microscopy needle-tip probe technique that probes nuclear stiffness in cultured cells to measure the nuclear envelope and cell membrane stiffness within native tissue is expanded. This technique is validated by imaging needle penetration and subsequent repair of the plasma and nuclear membranes of HeLa cells stably expressing the membrane repair protein CHMP4B-GFP. In the native tissue environment ex vivo, it is found that while enzymatic degradation of viable cartilage tissues with collagenase 3 (MMP-13) and aggrecanase-1 (ADAMTS-4) decreased tissue matrix stiffness, cell and nuclear membrane stiffness is also decreased. Finally, the capability for cell and nucleus elastography using the AFM needle-tip technique is demonstrated. These results demonstrate disruption of the native tissue environment that propagates to the plasma membrane and interior nuclear envelope structures of viable cells.
Collapse
Affiliation(s)
- Kaitlin P. McCreery
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Xin Xu
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Adrienne K. Scott
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Apresio K. Fajrial
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Sarah Calve
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Xiaoyun Ding
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Corey P. Neu
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
- Address correspondence to
| |
Collapse
|
107
|
Mechanical strain induces phenotypic changes in breast cancer cells and promotes immunosuppression in the tumor microenvironment. J Transl Med 2020; 100:1503-1516. [PMID: 32572176 PMCID: PMC7686122 DOI: 10.1038/s41374-020-0452-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BCa) proliferates within a complex, three-dimensional microenvironment amid heterogeneous biochemical and biophysical cues. Understanding how mechanical forces within the tumor microenvironment (TME) regulate BCa phenotype is of great interest. We demonstrate that mechanical strain enhanced the proliferation and migration of both estrogen receptor+ and triple-negative (TNBC) human and mouse BCa cells. Furthermore, a critical role for exosomes derived from cells subjected to mechanical strain in these pro-tumorigenic effects was identified. Exosome production by TNBC cells increased upon exposure to oscillatory strain (OS), which correlated with elevated cell proliferation. Using a syngeneic, orthotopic mouse model of TNBC, we identified that preconditioning BCa cells with OS significantly increased tumor growth and myeloid-derived suppressor cells (MDSCs) and M2 macrophages in the TME. This pro-tumorigenic myeloid cell enrichment also correlated with a decrease in CD8+ T cells. An increase in PD-L1+ exosome release from BCa cells following OS supported additive T cell inhibitory functions in the TME. The role of exosomes in MDSC and M2 macrophage was confirmed in vivo by cytotracking fluorescent exosomes, derived from labeled 4T1.2 cells, preconditioned with OS. In addition, in vivo internalization and intratumoral localization of tumor-cell derived exosomes was observed within MDSCs, M2 macrophages, and CD45-negative cell populations following direct injection of fluorescently-labeled exosomes. Our data demonstrate that exposure to mechanical strain promotes invasive and pro-tumorigenic phenotypes in BCa cells, indicating that mechanical strain can impact the growth and proliferation of cancer cell, alter exosome production by BCa, and induce immunosuppression in the TME by dampening anti-tumor immunity.
Collapse
|
108
|
Meli VS, Atcha H, Veerasubramanian PK, Nagalla RR, Luu TU, Chen EY, Guerrero-Juarez CF, Yamaga K, Pandori W, Hsieh JY, Downing TL, Fruman DA, Lodoen MB, Plikus MV, Wang W, Liu WF. YAP-mediated mechanotransduction tunes the macrophage inflammatory response. SCIENCE ADVANCES 2020; 6:eabb8471. [PMID: 33277245 PMCID: PMC7717914 DOI: 10.1126/sciadv.abb8471] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/20/2020] [Indexed: 05/11/2023]
Abstract
Macrophages are innate immune cells that adhere to the extracellular matrix within tissues. However, how matrix properties regulate their function remains poorly understood. Here, we report that the adhesive microenvironment tunes the macrophage inflammatory response through the transcriptional coactivator YAP. We find that adhesion to soft hydrogels reduces inflammation when compared to adhesion on stiff materials and is associated with reduced YAP expression and nuclear localization. Substrate stiffness and cytoskeletal polymerization, but not adhesive confinement nor contractility, regulate YAP localization. Furthermore, depletion of YAP inhibits macrophage inflammation, whereas overexpression of active YAP increases inflammation. Last, we show in vivo that soft materials reduce expression of inflammatory markers and YAP in surrounding macrophages when compared to stiff materials. Together, our studies identify YAP as a key molecule for controlling inflammation and sensing stiffness in macrophages and may have broad implications in the regulation of macrophages in health and disease.
Collapse
Affiliation(s)
- Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Hamza Atcha
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Praveen Krishna Veerasubramanian
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Raji R Nagalla
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Thuy U Luu
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Esther Y Chen
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Kosuke Yamaga
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - William Pandori
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Jessica Y Hsieh
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Timothy L Downing
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
109
|
Ogle ME, Doron G, Levy MJ, Temenoff JS. Hydrogel Culture Surface Stiffness Modulates Mesenchymal Stromal Cell Secretome and Alters Senescence. Tissue Eng Part A 2020; 26:1259-1271. [DOI: 10.1089/ten.tea.2020.0030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Molly E. Ogle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Gilad Doron
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Matthew J. Levy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Johnna S. Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
110
|
Jang B, Kim A, Hwang J, Song HK, Kim Y, Oh ES. Emerging Role of Syndecans in Extracellular Matrix Remodeling in Cancer. J Histochem Cytochem 2020; 68:863-870. [PMID: 32623937 PMCID: PMC7711240 DOI: 10.1369/0022155420930112] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
The extracellular matrix (ECM) offers a structural basis for regulating cell functions while also acting as a collection point for bioactive molecules and connective tissue cells. To perform pathological functions under a pathological condition, the involved cells need to regulate the ECM to support their altered functions. This is particularly common in the development of cancer. The ECM has been recognized as a key driver of cancer development and progression, and ECM remodeling occurs at all stages of cancer progression. Thus, cancer cells need to change the ECM to support relevant cell surface adhesion receptor-mediated cell functions. In this context, it is interesting to examine how cancer cells regulate ECM remodeling, which is critical to tumor malignancy and metastatic progression. Here, we review how the cell surface adhesion receptor, syndecan, regulates ECM remodeling as cancer progresses, and explore how this can help us better understand ECM remodeling under these pathological conditions.
Collapse
Affiliation(s)
- Bohee Jang
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Ayoung Kim
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jisun Hwang
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Hyun-Kuk Song
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Yunjeon Kim
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Eok-Soo Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
111
|
Song JJ, Go YY, Lee JK, Lee BS, Park SK, Jung H, Lee JH, Chang J. Transcriptomic analysis of tobacco-flavored E-cigarette and menthol-flavored E-cigarette exposure in the human middle ear. Sci Rep 2020; 10:20799. [PMID: 33247188 PMCID: PMC7699635 DOI: 10.1038/s41598-020-77816-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 11/16/2020] [Indexed: 11/09/2022] Open
Abstract
Electronic cigarettes (e-cigarettes) are the most widely used electronic nicotine delivery systems and are designed to imitate smoking and aid in smoking cessation. Although the number of e-cigarette users is increasing rapidly, especially among young adults and adolescents, the potential health impacts and biologic effects of e-cigarettes still need to be elucidated. Our previous study demonstrated the cytotoxic effects of electronic liquids (e-liquids) in a human middle ear epithelial cell (HMEEC-1) line, which were affected by the manufacturer and flavoring agents regardless of the presence of nicotine. In this study, we aimed to evaluate the gene expression profile and identify potential molecular modulator genes and pathways in HMEEC-1 exposed to two different e-liquids (tobacco- and menthol-flavored). HMEEC-1 was exposed to e-liquids, and RNA sequencing, functional analysis, and pathway analysis were conducted to identify the resultant transcriptomic changes. A total of 843 genes were differentially expressed following exposure to the tobacco-flavored e-liquid, among which 262 genes were upregulated and 581 were downregulated. Upon exposure to the menthol-flavored e-liquid, a total of 589 genes were differentially expressed, among which 228 genes were upregulated and 361 were downregulated. Among the signaling pathways associated with the differentially expressed genes mediated by tobacco-flavored e-liquid exposure, several key molecular genes were identified, including IL6 (interleukin 6), PTGS2 (prostaglandin-endoperoxide synthase 2), CXCL8 (C-X-C motif chemokine ligand 8), JUN (Jun proto-oncogene), FOS (Fos proto-oncogene), and TP53 (tumor protein 53). Under menthol-flavored e-liquid treatment, MMP9 (matrix metallopeptidase 9), PTGS2 (prostaglandin-endoperoxide synthase 2), MYC (MYC proto-oncogene, bHLH transcription factor), HMOX1 (heme oxygenase 1), NOS3 (nitric oxide synthase 3), and CAV1 (caveolin 1) were predicted as key genes. In addition, we identified related cellular processes, including inflammatory responses, oxidative stress and carcinogenesis, under exposure to tobacco- and menthol-flavored e-liquids. We identified differentially expressed genes and related cellular processes and gene signaling pathways after e-cigarette exposure in human middle ear cells. These findings may provide useful evidence for understanding the effect of e-cigarette exposure.
Collapse
Affiliation(s)
- Jae-Jun Song
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Korea
| | - Yoon Young Go
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Korea
| | - Jong Kyou Lee
- Department of Otolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 948-1, Daerim 1-dong, Yeongdeunpo-gu, Seoul, 150-950, Korea
| | - Bum Sang Lee
- Department of Otolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 948-1, Daerim 1-dong, Yeongdeunpo-gu, Seoul, 150-950, Korea
| | - Su-Kyoung Park
- Department of Otolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 948-1, Daerim 1-dong, Yeongdeunpo-gu, Seoul, 150-950, Korea
| | - Harry Jung
- Institute of New Frontier Research Team, Hallym Clinical and Translation Science Institute, Hallym University, Chuncheon, Republic of Korea
| | - Jun Ho Lee
- Department of Otolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 948-1, Daerim 1-dong, Yeongdeunpo-gu, Seoul, 150-950, Korea
| | - Jiwon Chang
- Department of Otolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 948-1, Daerim 1-dong, Yeongdeunpo-gu, Seoul, 150-950, Korea.
| |
Collapse
|
112
|
Mechanical suppression of breast cancer cell invasion and paracrine signaling to osteoclasts requires nucleo-cytoskeletal connectivity. Bone Res 2020; 8:40. [PMID: 33298883 PMCID: PMC7673025 DOI: 10.1038/s41413-020-00111-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 01/10/2023] Open
Abstract
Exercise benefits the musculoskeletal system and reduces the effects of cancer. The effects of exercise are multifactorial, where metabolic changes and tissue adaptation influence outcomes. Mechanical signals, a principal component of exercise, are anabolic to the musculoskeletal system and restrict cancer progression. We examined the mechanisms through which cancer cells sense and respond to low-magnitude mechanical signals introduced in the form of vibration. Low-magnitude, high-frequency vibration was applied to human breast cancer cells in the form of low-intensity vibration (LIV). LIV decreased matrix invasion and impaired secretion of osteolytic factors PTHLH, IL-11, and RANKL. Furthermore, paracrine signals from mechanically stimulated cancer cells, reduced osteoclast differentiation and resorptive capacity. Disconnecting the nucleus by knockdown of SUN1 and SUN2 impaired LIV-mediated suppression of invasion and osteolytic factor secretion. LIV increased cell stiffness; an effect dependent on the LINC complex. These data show that mechanical vibration reduces the metastatic potential of human breast cancer cells, where the nucleus serves as a mechanosensory apparatus to alter cell structure and intercellular signaling.
Collapse
|
113
|
Chuang YC, Chang HM, Li CY, Cui Y, Lee CL, Chen CS. Reactive Oxygen Species and Inflammatory Responses of Macrophages to Substrates with Physiological Stiffness. ACS APPLIED MATERIALS & INTERFACES 2020; 12:48432-48441. [PMID: 33064443 DOI: 10.1021/acsami.0c16638] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Macrophages play essential roles in innate immunity and their functions can be activated by different signals at pathological sites. Concerning changes in the rigidity of the microenvironment as a disease progresses, the influence of stiffened substrates on macrophage physiology remains elusive. In this study, to evaluate the effect of stiffened substrates on macrophages, we used J774A.1 cells as the macrophage model to investigate its mechanoinflammation responses using engineered polymeric substrates with various physiological rigidities (approximately 0.6 to 100 kPa). Under lipopolysaccharide (LPS) and adenosine triphosphate (ATP) stress, approximately 4-fold higher cytoplasmic reactive oxygen species (ROS) were triggered in cells on the softer substrate, compared with cells on the stiff substrates. The enhanced ROS response was found to be regulated mainly by NADPH oxidase. Moreover, mitochondrial ROS (mtROS), a crucial intracellular ROS source, are produced in response to substrate rigidity. The results showed higher mtROS production when cells were grown on a soft substrate with LPS/ATP stimuli, and the mechano-mtROS alteration was eliminated by Rho kinase inhibitor Y-27632. We suggest that substrate rigidity can coincide with LPS/ATP in regulating the ROS generation of macrophages. As a result of the pivotal role of ROS in regulating inflammation, increased NLRP-3 inflammasome formation and higher NO secretion (an approximately 300% increase) were observed with macrophages grown on soft substrates. Although no substantial genomic distinction was identified in our experiments, based on the phenotypic and functional results, softer substrates prime macrophages toward the proinflammatory (M1)-like phenotype. In summary, this study demonstrated the mechanosensitive inflammatory response of macrophages and the alteration of ROS, as secondary inflammation signals, may contribute to the functional status of macrophages. These findings not only provide an alternative interpretation of the functional transitions of macrophages influenced by substrate rigidity but may also support the manipulation of the inflammatory responses of macrophages via physical microenvironment modifications.
Collapse
Affiliation(s)
- Yung-Chu Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Hsaio-Ming Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
- School of Engineering, University of California Merced, Merced, California 95343, United States
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yujia Cui
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | | | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
114
|
Buwa N, Mazumdar D, Balasubramanian N. Caveolin1 Tyrosine-14 Phosphorylation: Role in Cellular Responsiveness to Mechanical Cues. J Membr Biol 2020; 253:509-534. [PMID: 33089394 DOI: 10.1007/s00232-020-00143-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The plasma membrane is a dynamic lipid bilayer that engages with the extracellular microenvironment and intracellular cytoskeleton. Caveolae are distinct plasma membrane invaginations lined by integral membrane proteins Caveolin1, 2, and 3. Caveolae formation and stability is further supported by additional proteins including Cavin1, EHD2, Pacsin2 and ROR1. The lipid composition of caveolar membranes, rich in cholesterol and phosphatidylserine, actively contributes to caveolae formation and function. Post-translational modifications of Cav1, including its phosphorylation of the tyrosine-14 residue (pY14Cav1) are vital to its function in and out of caveolae. Cells that experience significant mechanical stress are seen to have abundant caveolae. They play a vital role in regulating cellular signaling and endocytosis, which could further affect the abundance and distribution of caveolae at the PM, contributing to sensing and/or buffering mechanical stress. Changes in membrane tension in cells responding to multiple mechanical stimuli affects the organization and function of caveolae. These mechanical cues regulate pY14Cav1 levels and function in caveolae and focal adhesions. This review, along with looking at the mechanosensitive nature of caveolae, focuses on the role of pY14Cav1 in regulating cellular mechanotransduction.
Collapse
Affiliation(s)
- Natasha Buwa
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Debasmita Mazumdar
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Nagaraj Balasubramanian
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India.
| |
Collapse
|
115
|
Limzerwala JF, Jeganathan KB, Kloeber JA, Davies BA, Zhang C, Sturmlechner I, Zhong J, Fierro Velasco R, Fields AP, Yuan Y, Baker DJ, Zhou D, Li H, Katzmann DJ, van Deursen JM. FoxM1 insufficiency hyperactivates Ect2-RhoA-mDia1 signaling to drive cancer. NATURE CANCER 2020; 1:1010-1024. [PMID: 34841254 PMCID: PMC8623810 DOI: 10.1038/s43018-020-00116-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 08/17/2020] [Indexed: 01/28/2023]
Abstract
FoxM1 activates genes that regulate S-G2-M cell-cycle progression and, when overexpressed, is associated with poor clinical outcome in multiple cancers. Here we identify FoxM1 as a tumor suppressor in mice that, through its N-terminal domain, binds to and inhibits Ect2 to limit the activity of RhoA GTPase and its effector mDia1, a catalyst of cortical actin nucleation. FoxM1 insufficiency impedes centrosome movement through excessive cortical actin polymerization, thereby causing the formation of non-perpendicular mitotic spindles that missegregate chromosomes and drive tumorigenesis in mice. Importantly, low FOXM1 expression correlates with RhoA GTPase hyperactivity in multiple human cancer types, indicating that suppression of the newly discovered Ect2-RhoAmDia1 oncogenic axis by FoxM1 is clinically relevant. Furthermore, by dissecting the domain requirements through which FoxM1 inhibits Ect2 GEF activity, we provide mechanistic insight for the development of pharmacological approaches that target protumorigenic RhoA activity.
Collapse
Affiliation(s)
- Jazeel F Limzerwala
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jake A Kloeber
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, USA
| | - Brian A Davies
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ines Sturmlechner
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jian Zhong
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Raul Fierro Velasco
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alan P Fields
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Yaxia Yuan
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Daohong Zhou
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - David J Katzmann
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
116
|
Park S, Jung WH, Pittman M, Chen J, Chen Y. The Effects of Stiffness, Fluid Viscosity, and Geometry of Microenvironment in Homeostasis, Aging, and Diseases: A Brief Review. J Biomech Eng 2020; 142:100804. [PMID: 32803227 PMCID: PMC7477718 DOI: 10.1115/1.4048110] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/05/2020] [Indexed: 12/12/2022]
Abstract
Cells sense biophysical cues in the micro-environment and respond to the cues biochemically and biophysically. Proper responses from cells are critical to maintain the homeostasis in the body. Abnormal biophysical cues will cause pathological development in the cells; pathological or aging cells, on the other hand, can alter their micro-environment to become abnormal. In this minireview, we discuss four important biophysical cues of the micro-environment-stiffness, curvature, extracellular matrix (ECM) architecture and viscosity-in terms of their roles in health, aging, and diseases.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218; Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218; Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| | - Wei-Hung Jung
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218; Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218; Department of Mechanical Engineering, Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| | - Matthew Pittman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218; Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218; Department of Mechanical Engineering, Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| | - Junjie Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218; Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218; Department of Mechanical Engineering, Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218; Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218; Department of Mechanical Engineering, Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
117
|
Lu T, Lin X, Pan YH, Yang N, Ye S, Zhang Q, Wang C, Zhu R, Zhang T, Wisniewski TM, Cao Z, Ding BS, Dang S, Zhang W. ADAMTS18 Deficiency Leads to Pulmonary Hypoplasia and Bronchial Microfibril Accumulation. iScience 2020; 23:101472. [PMID: 32882513 PMCID: PMC7476315 DOI: 10.1016/j.isci.2020.101472] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 07/02/2020] [Accepted: 08/17/2020] [Indexed: 01/06/2023] Open
Abstract
ADAMTSs (a disintegrin and metalloproteinase with thrombospondin motifs) are secreted metalloproteinases that play a major role in the assembly and degradation of the extracellular matrix (ECM). In this study, we show that ADAMTS18, produced by the epithelial cells of distal airways and mesenchymal cells in lung apex at early embryonic stages, serves as a morphogen in lung development. ADAMTS18 deficiency leads to reduced number and length of bronchi, tipped lung apexes, and dilated alveoli. These developmental defects worsen lipopolysaccharide-induced acute lung injury and bleomycin-induced lung fibrosis in adult Adamts18-deficient mice. ADAMTS18 deficiency also causes increased levels of fibrillin1 and fibrillin2, bronchial microfibril accumulation, decreased focal adhesion kinase signaling, and disruption of F-actin organization. Our findings indicate that ECM homeostasis mediated by ADAMTS18 is pivotal in airway branching morphogenesis.
Collapse
Affiliation(s)
- Tiantian Lu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Xiaotian Lin
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Ning Yang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Shuai Ye
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Qi Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Caiyun Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Rui Zhu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Tianhao Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Thomas M. Wisniewski
- Departments of Neurology, Pathology and Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Zhongwei Cao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Suying Dang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| |
Collapse
|
118
|
Fagotto F. EpCAM as Modulator of Tissue Plasticity. Cells 2020; 9:E2128. [PMID: 32961790 PMCID: PMC7563481 DOI: 10.3390/cells9092128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 01/01/2023] Open
Abstract
The Epithelial Cell Adhesion Molecule or EpCAM is a well-known marker highly expressed in carcinomas and showing a strong correlation with poor cancer prognosis. While its name relates to its proposed function as a cell adhesion molecule, EpCAM has been shown to have various signalling functions. In particular, it has been identified as an important positive regulator of cell adhesion and migration, playing an essential role in embryonic morphogenesis as well as intestinal homeostasis. This activity is not due to its putative adhesive function, but rather to its ability to repress myosin contractility by impinging on a PKC signalling cascade. This mechanism confers EpCAM the unique property of favouring tissue plasticity. I review here the currently available data, comment on possible connections with other properties of EpCAM, and discuss the potential significance in the context of cancer invasion.
Collapse
Affiliation(s)
- François Fagotto
- CRBM, University of Montpellier and CNRS, 34293 Montpellier, France
| |
Collapse
|
119
|
Balion Z, Sipailaite E, Stasyte G, Vailionyte A, Mazetyte-Godiene A, Seskeviciute I, Bernotiene R, Phopase J, Jekabsone A. Investigation of Cancer Cell Migration and Proliferation on Synthetic Extracellular Matrix Peptide Hydrogels. Front Bioeng Biotechnol 2020; 8:773. [PMID: 33014989 PMCID: PMC7498748 DOI: 10.3389/fbioe.2020.00773] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023] Open
Abstract
Chemical and mechanical properties of a tumor microenvironment are essential players in cancer progression, and it is important to precisely control the extracellular conditions while designing cancer in vitro models. The study investigates synthetic hydrogel matrices from multi-arm polyethylene glycol (PEG) functionalized with collagen-like peptide (CLP) CG(PKG)4(POG)4(DOG)4 alone and conjugated with either cell adhesion peptide RGD (mimicking fibronectin) or IKVAV (mimicking laminin). Human glioblastoma HROG36, rat C6 glioma cells, and A375 human melanoma cells were grown on the hydrogels and monitored for migration, proliferation, projected cell area, cell shape index, size and number, distribution of focal contacts in individual cells, and focal adhesion number. PEG-CLP-RGD induced migration of both glioma cell lines and also stimulated proliferation (assessed as metabolic activity) of HROG36 cells. Migration of C6 cells were also stimulated by PEG-CLP-IKVAV. These responses strongly correlated with the changes in adhesion and morphology parameters of individual cells – projected cell area, cell shape index, and focal contact number. Melanoma A375 cell proliferation was increased by PEG-CLP-RGD, and this was accompanied by a decrease in cell shape index. However, neither RGD nor IKVAV conjugated to PEG-CLP stimulated migratory capacity of A375 cells. Taken together, the study presents synthetic scaffolds with extracellular matrix (ECM)-mimicking peptides that allow for the exploration of the effect of ECM signaling to cancer cells.
Collapse
Affiliation(s)
- Zbigniev Balion
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Emilija Sipailaite
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Gabija Stasyte
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Agne Vailionyte
- Ferentis UAB, Vilnius, Lithuania.,Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania
| | - Airina Mazetyte-Godiene
- Ferentis UAB, Vilnius, Lithuania.,Department of Nanoengineering, Center for Physical Sciences and Technology, Vilnius, Lithuania
| | - Ieva Seskeviciute
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Bernotiene
- Laboratory of Molecular Neurobiology, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jaywant Phopase
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Aiste Jekabsone
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Laboratory of Molecular Neurobiology, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
120
|
Sankaranarayanan S, Kessler SA. Growing straight through walls. eLife 2020; 9:e61647. [PMID: 32867921 PMCID: PMC7462601 DOI: 10.7554/elife.61647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 11/13/2022] Open
Abstract
The pollen tube in a flowering plant grows in a direction that is influenced by the mechanical properties of the stigma papillae and the organization of structures called cortical microtubules inside these cells.
Collapse
Affiliation(s)
- Subramanian Sankaranarayanan
- Department of Botany and Plant Pathology, Purdue UniversityWest LafayetteUnited States
- Purdue Center for Plant Biology, Purdue UniversityWest LafayetteUnited States
| | - Sharon A Kessler
- Department of Botany and Plant Pathology, Purdue UniversityWest LafayetteUnited States
- Purdue Center for Plant Biology, Purdue UniversityWest LafayetteUnited States
| |
Collapse
|
121
|
Stamenović D, Smith ML. Tensional homeostasis at different length scales. SOFT MATTER 2020; 16:6946-6963. [PMID: 32696799 DOI: 10.1039/d0sm00763c] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Tensional homeostasis is a phenomenon of fundamental importance in mechanobiology. It refers to the ability of organs, tissues, and cells to respond to external disturbances by maintaining a homeostatic (set point) level of mechanical stress (tension). It is well documented that breakdown in tensional homeostasis is the hallmark of progression of diseases, including cancer and atherosclerosis. In this review, we surveyed quantitative studies of tensional homeostasis with the goal of providing characterization of this phenomenon across a broad range of length scales, from the organ level to the subcellular level. We considered both static and dynamics approaches that have been used in studies of this phenomenon. Results that we found in the literature and that we obtained from our own investigations suggest that tensional homeostasis is an emergent phenomenon driven by collective rheostatic mechanisms associated with focal adhesions, and by a collective action of cells in multicellular forms, whose impact on tensional homeostasis is cell type-dependent and cell microenvironment-dependent. Additionally, the finding that cadherins, adhesion molecules that are important for formation of cell-cell junctions, promote tensional homeostasis even in single cells, demonstrates their relevance as a signaling moiety.
Collapse
Affiliation(s)
- Dimitrije Stamenović
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA.
| | | |
Collapse
|
122
|
Wei Q, Young J, Holle A, Li J, Bieback K, Inman G, Spatz JP, Cavalcanti-Adam EA. Soft Hydrogels for Balancing Cell Proliferation and Differentiation. ACS Biomater Sci Eng 2020; 6:4687-4701. [PMID: 33455192 DOI: 10.1021/acsbiomaterials.0c00854] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hydrogels have been widely explored for the delivery of cells in a variety of regenerative medicine applications due to their ability to mimic both the biochemical and physical cues of cell microniches. For bone regeneration, in particular, stiff hydrogels mimicking osteoid stiffness have been utilized due to the fact that stiff substrates favor stem cell osteogenic differentiation. Unlike cell adhesion in two dimensions, three-dimensional hydrogels offer mechanical stimulation but limit the cell spreading and growth due to the dense matrix network. Therefore, we designed degradable, soft hydrogels (∼0.5 kPa) mimicking the soft bone marrow stiffness, with incorporated matrix metalloproteinase (MMP)-cleavable sites and RGD-based adhesive sites, to enhance the spreading and proliferation of the encapsulated cells, which are commonly inhibited in nondegradable and/or stiff implants. When the hydrogels were cultured on rigid surfaces to mirror the microenvironment of bone defects in vivo, the cells were shown to migrate toward the interface and differentiate down the osteogenic lineage, enhanced by the codelivery of bone morphogenetic protein-2 (BMP-2). Furthermore, this soft hydrogel might find applications in therapeutic interventions since it is easily injectable and cost-efficient. Taken together, we have designed a new system to balance cell growth and differentiation for improving hydrogel-based bone regenerative medicine strategies.
Collapse
Affiliation(s)
- Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065 Chengdu, China
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120 Heidelberg, Germany
| | - Jennifer Young
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120 Heidelberg, Germany
| | - Andrew Holle
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120 Heidelberg, Germany
| | - Jie Li
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120 Heidelberg, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University and German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany
| | - Gareth Inman
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, U.K
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120 Heidelberg, Germany
| | - Elisabetta A Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120 Heidelberg, Germany
- Central Scientific Facility "Cellular Biotechnology", MPI for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
| |
Collapse
|
123
|
Alpha KM, Xu W, Turner CE. Paxillin family of focal adhesion adaptor proteins and regulation of cancer cell invasion. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:1-52. [PMID: 32859368 PMCID: PMC7737098 DOI: 10.1016/bs.ircmb.2020.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The paxillin family of proteins, including paxillin, Hic-5, and leupaxin, are focal adhesion adaptor/scaffolding proteins which localize to cell-matrix adhesions and are important in cell adhesion and migration of both normal and cancer cells. Historically, the role of these proteins in regulating the actin cytoskeleton through focal adhesion-mediated signaling has been well documented. However, studies in recent years have revealed additional functions in modulating the microtubule and intermediate filament cytoskeletons to affect diverse processes including cell polarization, vesicle trafficking and mechanosignaling. Expression of paxillin family proteins in stromal cells is also important in regulating tumor cell migration and invasion through non-cell autonomous effects on the extracellular matrix. Both paxillin and Hic-5 can also influence gene expression through a variety of mechanisms, while their own expression is frequently dysregulated in various cancers. Accordingly, these proteins may serve as valuable targets for novel diagnostic and treatment approaches in cancer.
Collapse
Affiliation(s)
- Kyle M Alpha
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weiyi Xu
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Christopher E Turner
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States.
| |
Collapse
|
124
|
Abstract
Despite decades of laboratory, epidemiological and clinical research, breast cancer incidence continues to rise. Breast cancer remains the leading cancer-related cause of disease burden for women, affecting one in 20 globally and as many as one in eight in high-income countries. Reducing breast cancer incidence will likely require both a population-based approach of reducing exposure to modifiable risk factors and a precision-prevention approach of identifying women at increased risk and targeting them for specific interventions, such as risk-reducing medication. We already have the capacity to estimate an individual woman's breast cancer risk using validated risk assessment models, and the accuracy of these models is likely to continue to improve over time, particularly with inclusion of newer risk factors, such as polygenic risk and mammographic density. Evidence-based risk-reducing medications are cheap, widely available and recommended by professional health bodies; however, widespread implementation of these has proven challenging. The barriers to uptake of, and adherence to, current medications will need to be considered as we deepen our understanding of breast cancer initiation and begin developing and testing novel preventives.
Collapse
Affiliation(s)
- Kara L Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.
| | - Jack Cuzick
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Kelly-Anne Phillips
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
125
|
Gheorghe RO, Deftu A, Filippi A, Grosu A, Bica-Popi M, Chiritoiu M, Chiritoiu G, Munteanu C, Silvestro L, Ristoiu V. Silencing the Cytoskeleton Protein Iba1 (Ionized Calcium Binding Adapter Protein 1) Interferes with BV2 Microglia Functioning. Cell Mol Neurobiol 2020; 40:1011-1027. [PMID: 31950314 PMCID: PMC11448859 DOI: 10.1007/s10571-020-00790-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/07/2020] [Indexed: 01/31/2023]
Abstract
Iba1 (ionized calcium binding adapter protein 1) is a cytoskeleton protein specific only for microglia and macrophages, where it acts as an actin-cross linking protein. Although frequently regarded as a marker of activation, its involvement in cell migration, membrane ruffling, phagocytosis or in microglia remodeling during immunological surveillance of the brain suggest that Iba1 is not a simple cytoskeleton protein, but a signaling molecule involved in specific signaling pathways. In this study we investigated if Iba1 could also represent a drug target, and tested the hypothesis that its specific silencing with customized Iba1-siRNA can modulate microglia functioning. The results showed that Iba1-silenced BV2 microglia migrate less due to reduced proliferation and cell adhesion, while their phagocytic activity and P2x7 functioning was significantly increased. Our data are the proof of concept that Iba1 protein is a new microglia target, which opens a new therapeutic avenue for modulating microglia behavior.
Collapse
Affiliation(s)
- Roxana-Olimpia Gheorghe
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Alexandru Deftu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Alexandru Filippi
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of the Romanian Academy, Bucharest, Romania
| | - Andreea Grosu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Melania Bica-Popi
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania
| | - Marioara Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Gabriela Chiritoiu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Cristian Munteanu
- Department of Molecular Cell Biology, Institute of Biochemistry, Romanian Academy, Splaiul Independentei 296, 06003, Bucharest, Romania
| | - Luigi Silvestro
- Pharma Serv International, Sabinelor 52, 050853, Bucharest, Romania
| | - Violeta Ristoiu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, Sector 5, 050095, Bucharest, Romania.
| |
Collapse
|
126
|
Domingues C, Geraldo AM, Anjo SI, Matos A, Almeida C, Caramelo I, Lopes-da-Silva JA, Paiva A, Carvalho J, Pires das Neves R, Manadas B, Grãos M. Cofilin-1 Is a Mechanosensitive Regulator of Transcription. Front Cell Dev Biol 2020; 8:678. [PMID: 32903827 PMCID: PMC7438942 DOI: 10.3389/fcell.2020.00678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical properties of the extracellular environment are interrogated by cells and integrated through mechanotransduction. Many cellular processes depend on actomyosin-dependent contractility, which is influenced by the microenvironment’s stiffness. Here, we explored the influence of substrate stiffness on the proteome of proliferating undifferentiated human umbilical cord-matrix mesenchymal stem/stromal cells. The relative abundance of several proteins changed significantly by expanding cells on soft (∼3 kPa) or stiff substrates (GPa). Many such proteins are associated with the regulation of the actin cytoskeleton, a major player of mechanotransduction and cell physiology in response to mechanical cues. Specifically, Cofilin-1 levels were elevated in cells cultured on soft comparing with stiff substrates. Furthermore, Cofilin-1 was de-phosphorylated (active) and present in the nuclei of cells kept on soft substrates, in contrast with phosphorylated (inactive) and widespread distribution in cells on stiff. Soft substrates promoted Cofilin-1-dependent increased RNA transcription and faster RNA polymerase II-mediated transcription elongation. Cofilin-1 is part of a novel mechanism linking mechanotransduction and transcription.
Collapse
Affiliation(s)
- Catarina Domingues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - A Margarida Geraldo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Sandra Isabel Anjo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - André Matos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Cláudio Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Inês Caramelo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | | | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - João Carvalho
- Centro de Física da Universidade de Coimbra (CFisUC), Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal.,Biocant, Technology Transfer Association, Cantanhede, Portugal
| |
Collapse
|
127
|
Stromal CCL2 Signaling Promotes Mammary Tumor Fibrosis through Recruitment of Myeloid-Lineage Cells. Cancers (Basel) 2020; 12:cancers12082083. [PMID: 32731354 PMCID: PMC7465971 DOI: 10.3390/cancers12082083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is correlated with breast tumor desmoplasia, leading to diminished chemotherapy response and disease-free survival. Obesity causes chronic, macrophage-driven inflammation within breast tissue, initiated by chemokine ligand 2 (CCL2) signaling from adipose stromal cells. To understand how CCL2-induced inflammation alters breast tumor pathology, we transplanted oncogenically transformed human breast epithelial cells with breast stromal cells expressing CCL2 or empty vector into murine mammary glands and examined tumor formation and progression with time. As tumors developed, macrophages were rapidly recruited, followed by the emergence of cancer-associated fibroblasts (CAFs) and collagen deposition. Depletion of CD11b + myeloid lineage cells early in tumor formation reduced tumor growth, CAF numbers, and collagen deposition. CCL2 expression within developing tumors also enhanced recruitment of myeloid progenitor cells from the bone marrow into the tumor site. The myeloid progenitor cell population contained elevated numbers of fibrocytes, which exhibited platelet-derived growth factor receptor-alpha (PDGFRα)-dependent colony formation and growth in vitro. Together, these results suggest that chronic inflammation induced by CCL2 significantly enhances tumor growth and promotes the formation of a desmoplastic stroma through early recruitment of macrophages and fibrocytes into the tumor microenvironment. Fibrocytes may be a novel target in the tumor microenvironment to reduce tumor fibrosis and enhance treatment responses for obese breast cancer patients.
Collapse
|
128
|
Wu Y, Tang H, Liu L, He Q, Zhao L, Huang Z, Yang J, Cao C, Chen J, Wang A. Biomimetic titanium implant coated with extracellular matrix enhances and accelerates osteogenesis. Nanomedicine (Lond) 2020; 15:1779-1793. [PMID: 32705940 DOI: 10.2217/nnm-2020-0047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To evaluate the biological function of titanium implants coated with cell-derived mineralized extracellular matrix, which mimics a bony microenvironment. Materials & methods: A biomimetic titanium implant was fabricated primarily by modifying the titanium surface with TiO2 nanotubes or sand-blasted, acid-etched topography, then was coated with mineralized extracellular matrix constructed by culturing bone marrow mesenchymal stromal cells. The osteogenic ability of biomimetic titanium surface in vitro and in vivo were evaluated. Results: In vitro and in vivo studies revealed that the biomimetic titanium implant enhanced and accelerated osteogenesis of bone marrow stromal cells by increasing cell proliferation and calcium deposition. Conclusion: By combining surface topography modification with biological coating, the results provided a valuable method to produce biomimetic titanium implants with excellent osteogenic ability.
Collapse
Affiliation(s)
- Yu Wu
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Haikuo Tang
- Department of Oral & Maxillofacial Surgery, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Lin Liu
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Qianting He
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Luodan Zhao
- Department of Stomatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China
| | - Zhexun Huang
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Jinghong Yang
- Department of Prosthodontics, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Congyuan Cao
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Jie Chen
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Anxun Wang
- Department of Oral & Maxillofacial Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| |
Collapse
|
129
|
Nguyen AK, Kilian KA. Physicochemical Tools for Visualizing and Quantifying Cell-Generated Forces. ACS Chem Biol 2020; 15:1731-1746. [PMID: 32530602 DOI: 10.1021/acschembio.0c00304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To discern how mechanical forces coordinate biological outcomes, methods that map cell-generated forces in a spatiotemporal manner, and at cellular length scales, are critical. In their native environment, whether it be within compact multicellular three-dimensional structures or sparsely populated fibrillar networks of the extracellular matrix, cells are constantly exposed to a slew of physical forces acting on them from all directions. At the same time, cells exert highly localized forces of their own on their surroundings and on neighboring cells. Together, the generation and transmission of these forces can control diverse cellular activities and behavior as well as influence cell fate decisions. To thoroughly understand these processes, we must first be able to characterize and measure such forces. However, our experimental needs and technical capabilities are in discord-while it is apparent that we should study cell-generated forces within more biologically relevant 3D environments, this goal remains challenging because of caveats associated with complex "sensing-transduction-readout" modalities. In this Review, we will discuss the latest techniques for measuring cell-generated forces. We will highlight recent advances in traction force microscopy and examine new alternative approaches for quantifying cell-generated forces, both of individual cells and within 3D tissues. Finally, we will explore the future direction of novel cellular force-sensing tools in the context of mechanobiology and next-generation biomaterials design.
Collapse
Affiliation(s)
- Ashley K. Nguyen
- School of Chemistry, School of Materials Science and Engineering, Australian Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Kristopher A. Kilian
- School of Chemistry, School of Materials Science and Engineering, Australian Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
130
|
Hopkins AA, Murphy R, Irnaten M, Wallace DM, Quill B, O'Brien C. The role of lamina cribrosa tissue stiffness and fibrosis as fundamental biomechanical drivers of pathological glaucoma cupping. Am J Physiol Cell Physiol 2020; 319:C611-C623. [PMID: 32667866 DOI: 10.1152/ajpcell.00054.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The primary biomechanical driver of pathological glaucomatous cupping remains unknown. Finite element modeling indicates that stress and strain play key roles. In this article, primarily a review, we utilize known biomechanical data and currently unpublished results from our lab to propose a three-stage, tissue stiffness-based model to explain glaucomatous cupping occurring at variable levels of translaminar pressure (TLP). In stage 1, a short-term increase in TLP gradient induces a transient increase in lamina cribrosa (LC) strain. Beyond a critical level of strain, the tissue stiffness rises steeply provoking cellular responses via integrin-mediated mechanotransduction. This early mechanoprotective cellular contraction reduces strain, which reduces tissue stiffness by return of the posteriorly deflected LC to baseline. In stage 2 a prolonged period of TLP increase elicits extracellular matrix (ECM) production leading to fibrosis, increasing baseline tissue stiffness and strain and diminishing the contractile ability/ability to return to the baseline LC position. This is supported by our three-dimensional collagen contraction assays, which show significantly reduced capacity to contract in glaucoma compared with normal LC cells. Second, 15% cyclic strain in LC cells over 24 h elicits a typical increase in ECM profibrotic genes in normal LC cells but a highly blunted response in glaucoma LC cells. Stage 3 is characterized by persistent fibrosis causing further stiffening and inducing a feed-forward ECM production cycle. Repeated cycles of increased strain and stiffness with profibrotic ECM deposition prevent optic nerve head (ONH) recoil from the new deflected position. This incremental maladaptive modeling leads to pathological ONH cupping.
Collapse
Affiliation(s)
- Alan A Hopkins
- Clinical Research Centre, Catherine McAuley Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Rory Murphy
- Clinical Research Centre, Catherine McAuley Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Mustapha Irnaten
- Clinical Research Centre, Catherine McAuley Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Deborah M Wallace
- Clinical Research Centre, Catherine McAuley Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Barry Quill
- Clinical Research Centre, Catherine McAuley Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Colm O'Brien
- Clinical Research Centre, Catherine McAuley Centre, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
131
|
McNeill MC, Wray J, Sala-Newby GB, Hindmarch CCT, Smith SA, Ebrahimighaei R, Newby AC, Bond M. Nuclear actin regulates cell proliferation and migration via inhibition of SRF and TEAD. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118691. [PMID: 32119877 PMCID: PMC7262588 DOI: 10.1016/j.bbamcr.2020.118691] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/20/2022]
Abstract
Actin dynamics regulate cell behaviour in response to physiological signals. Here we demonstrate a novel role for nuclear actin in inhibiting cell proliferation and migration. We demonstrate that physiological signals that elevate cAMP, which is anti-mitogenic in vascular smooth muscle cells, increases nuclear actin monomer levels. Expression of a nuclear-targeted polymerisation-defective actin mutant (NLS-ActinR62D) inhibited proliferation and migration. Preventing nuclear actin monomer accumulation by enhancing its nuclear export or polymerisation reversed the anti-mitogenic and anti-migratory effects of cAMP. Transcriptomic analysis identified repression of proliferation and migration associated genes regulated by serum response factor (SRF) and TEA Domain (TEAD) transcription factors. Accordingly, NLS-ActinR62D inhibited SRF and TEAD activity and target gene expression, and these effects were reversed by constitutively-active mutants of the TEAD and SRF co-factors YAP, TAZ and MKL1. In summary, intranuclear actin inhibits proliferation and migration by inhibiting YAP-TEAD and MKL-SRF activity. This mechanism explains the anti-mitogenic and anti-migratory properties of physiological signals that elevate cAMP. SUMMARY: McNeill et al show that increased levels of intranuclear actin monomer inhibit cell proliferation and migration by inhibiting MKL1-SRF and YAP/TAZ-TEAD-dependent gene expression. This mechanism mediates the anti-mitogenic and anti-migratory effects of physiological signals that elevate cyclic-AMP.
Collapse
Affiliation(s)
- Madeleine C McNeill
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Jason Wray
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Graciela B Sala-Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Charles C T Hindmarch
- Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Kingston, ON K7L3N6, Canada
| | - Sarah A Smith
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Reza Ebrahimighaei
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Andrew C Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Mark Bond
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| |
Collapse
|
132
|
Vaeyens MM, Jorge-Peñas A, Barrasa-Fano J, Shapeti A, Roeffaers M, Van Oosterwyck H. Actomyosin-dependent invasion of endothelial sprouts in collagen. Cytoskeleton (Hoboken) 2020; 77:261-276. [PMID: 32588525 DOI: 10.1002/cm.21624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/11/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
During sprouting angiogenesis-the growth of blood vessels from the existing vasculature-endothelial cells (ECs) adopt an elongated invasive form and exert forces at cell-cell and cell-matrix interaction sites. These cell shape changes and cellular tractions require extensive reorganizations of the actomyosin network. However, the respective roles of actin and myosin for endothelial sprouting are not fully elucidated. In this study, we further investigate these roles by treating 2D-migrating and 3D-sprouting ECs with chemical compounds targeting either myosin or actin. These treatments affected the endothelial cytoskeleton drastically and reduced the invasive response in a compound-specific manner; pointing toward a tight control of the actin and myosin activity during sprouting. Clusters in the data further illustrate that endothelial sprout morphology is sensitive to the in vitro model mechanical microenvironment and directs future research toward mechanical substrate guidance as a strategy for promoting engineered tissue vascularization. In summary, our results add to a growing corpus of research highlighting a key role of the cytoskeleton for sprouting angiogenesis.
Collapse
Affiliation(s)
- Marie-Mo Vaeyens
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alvaro Jorge-Peñas
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jorge Barrasa-Fano
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Apeksha Shapeti
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Roeffaers
- Department of Microbial and Molecular Systems (M2S), Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
133
|
SB772077B (SB77) Alleviated the Aqueous Outflow Resistance Mediated by Cyclic Mechanical Stress in Perfused Human Cadaveric Eyes. Sci Rep 2020; 10:10202. [PMID: 32576873 PMCID: PMC7311383 DOI: 10.1038/s41598-020-67087-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/03/2020] [Indexed: 01/08/2023] Open
Abstract
The intraocular pressure lowering property of a new rho kinase inhibitor, SB772077B (SB77) has been previously demonstrated in perfused human cadaveric eyes. In this study, the efficacy of SB77 in alleviating the aqueous outflow resistance mediated by cyclic mechanical stress in perfused human cadaveric eyes was investigated. A human anterior segment perfusion culture model was used to investigate the effect of cyclic intraocular pressure (IOP) on aqueous outflow facility in presence or absence of SB77. The status of RhoA activation and the downstream effector molecule myosin-light chain phosphorylation (p-MLC) was investigated by Western blot. Cyclic mechanical stress resulted in decrease in aqueous outflow facility (-19.79 ± 4.93%; p = 0.019) in perfused human eyes and treatment with SB77 (50 µM) significantly enhanced outflow facility by 15% (p = 0.05). The increase in outflow facility by SB77 was confirmed with the inactivation of RhoA/ROCK signaling and decreased expression of extracellular matrix markers. SB77 effectively reduced the outflow resistance mediated by cyclic IOP and thus may be a potential clinical candidate for the management of glaucoma.
Collapse
|
134
|
Wu S, Zhang D, Bai J, Zheng H, Deng J, Gou Z, Gao C. Adsorption of serum proteins on titania nanotubes and its role on regulating adhesion and migration of mesenchymal stem cells. J Biomed Mater Res A 2020; 108:2305-2318. [DOI: 10.1002/jbm.a.36987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/28/2020] [Accepted: 04/04/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Sai Wu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and EngineeringZhejiang University Hangzhou China
| | - Deteng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and EngineeringZhejiang University Hangzhou China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University Hangzhou China
| | - Jun Bai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and EngineeringZhejiang University Hangzhou China
| | - Honghao Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and EngineeringZhejiang University Hangzhou China
| | - Jun Deng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and EngineeringZhejiang University Hangzhou China
| | - Zhongru Gou
- Bio‐nanomaterials and Regenerative Medicine Research Division, Zhejiang‐California International Nanosystem InstituteZhejiang University Hangzhou China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and EngineeringZhejiang University Hangzhou China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University Hangzhou China
| |
Collapse
|
135
|
Bin J, Nie S, Tang Z, Kang A, Fu Z, Hu Y, Liao Q, Xiong W, Zhou Y, Tang Y, Jiang J. Long noncoding RNA EPB41L4A-AS1 functions as an oncogene by regulating the Rho/ROCK pathway in colorectal cancer. J Cell Physiol 2020; 236:523-535. [PMID: 32557646 DOI: 10.1002/jcp.29880] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/20/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors worldwide. In terms of cancer-related death, colon cancer ranks second and third among men and women, respectively, and the incidence is increasing annually. Accumulating evidence have indicated that long noncoding RNA (lncRNA) plays an important role in tumorigenesis. In this study, we found that lncRNA EPB41L4A-AS1 was highly expressed in CRC tissues and was associated with poor prognosis and tumor metastasis in patients with CRC. In vitro studies showed that the knockdown of EPB41L4A-AS1 inhibited the proliferation, migration, invasion, and epithelial-mesenchymal transition of CRC cells. Mechanically, we found that EPB41L4A-AS1 may participate in the development of CRC by activating the Rho/Rho-associated protein kinase signaling pathway. Collectively, these results demonstrated that EPB41L4A-AS1 can promote the proliferation, invasion, and migration of CRC, and it may be a novel biomarker for the diagnosis and targeted treatment of CRC.
Collapse
Affiliation(s)
- Jie Bin
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shaolin Nie
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ziyuan Tang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Anding Kang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhongping Fu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yingbin Hu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Central Laboratory, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Xiong
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yujuan Zhou
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yanyan Tang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Central Laboratory, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiarui Jiang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
136
|
From 2646 to 15: differentially regulated microRNAs between progenitors from normal myometrium and leiomyoma. Am J Obstet Gynecol 2020; 222:596.e1-596.e9. [PMID: 31874141 DOI: 10.1016/j.ajog.2019.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 11/15/2019] [Accepted: 12/16/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Uterine leiomyomas (fibroids) are smooth muscle neoplasms of the myometrial layer of the uterus and are the most common benign tumors in women. Although their etiology is still unclear, progenitor cells seem to be implicated. OBJECTIVE To identify the dysregulated pathways involved in leiomyoma onset by microRNA profiling of progenitor cells isolated from normal myometrium and leiomyoma tissue. MATERIALS AND METHODS Pairs of normal myometrium and uterine fibroid specimens were collected from 12 myomectomy patients. Myometrial progenitor cells and leiomyoma progenitor cells were isolated and characterized for stemness. After total RNA extraction and profiling of their 2646 microRNAs, DIANA-miRPath analysis was applied to find any dysregulated pathways. RESULTS Only 30 microRNAs showed a significant differential regulation between myometrial progenitor cells and leiomyoma progenitor cells. Removal of those that had values close to the cut-off or that were not consistent among triplicates left 15 microRNAs, of which 7 were downregulated and 8 were upregulated in leiomyoma progenitor cells compared to myometrial progenitor cells. According to DIANA-miRPath analysis, the 7 downregulated microRNAs (hsa-miR-146b-5p; hsa-miR-335-3p; hsa-miR-335-5p; hsa-miR-135b-5p; hsa-miR-10a-3p; hsa-miR-10a-5p; hsa-miR-200a-3p) are all related to 3 pathways, "ECM-receptor interaction" (33 targeted genes), "Adherens junction" (33 targeted genes), and "Hippo signaling" (69 targeted genes), whereas the 8 upregulated miRNAs (hsa-miR-146a-5p; hsa-miR-576-3p; hsa-miR-122-5p; hsa-miR-1246; hsa-miR-595; hsa-miR-658; hsa-miR-4284; hsa-miR-924) are related to 4 pathways, "PI3K-Akt signaling pathway" (71 targeted genes), "Pathways in Cancer" (80 targeted genes), "Cell Cycle" (37 targeted genes), and "Regulation of actin cytoskeleton" (41 targeted genes). CONCLUSION The findings that only 15 of 2646 microRNAs are differentially regulated in normal myometrium and leiomyoma and that they are involved in 7 dysregulated pathways provides interesting insights into the development of uterine fibroids, and lends support to the hypothesis that leiomyoma onset is the result of alterations affecting progenitor cells.
Collapse
|
137
|
van Dijk CG, Louzao-Martinez L, van Mulligen E, Boermans B, Demmers JA, van den Bosch TP, Goumans MJ, Duncker DJ, Verhaar MC, Cheng C. Extracellular Matrix Analysis of Human Renal Arteries in Both Quiescent and Active Vascular State. Int J Mol Sci 2020; 21:E3905. [PMID: 32486169 PMCID: PMC7313045 DOI: 10.3390/ijms21113905] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/30/2023] Open
Abstract
In vascular tissue engineering strategies, the addition of vascular-specific extracellular matrix (ECM) components may better mimic the in vivo microenvironment and potentially enhance cell-matrix interactions and subsequent tissue growth. For this purpose, the exact composition of the human vascular ECM first needs to be fully characterized. Most research has focused on characterizing ECM components in mature vascular tissue; however, the developing fetal ECM matches the active environment required in vascular tissue engineering more closely. Consequently, we characterized the ECM protein composition of active (fetal) and quiescent (mature) renal arteries using a proteome analysis of decellularized tissue. The obtained human fetal renal artery ECM proteome dataset contains higher levels of 15 ECM proteins versus the mature renal artery ECM proteome, whereas 16 ECM proteins showed higher levels in the mature tissue compared to fetal. Elastic ECM proteins EMILIN1 and FBN1 are significantly enriched in fetal renal arteries and are mainly produced by cells of mesenchymal origin. We functionally tested the role of EMILIN1 and FBN1 by anchoring the ECM secreted by vascular smooth muscle cells (SMCs) to glass coverslips. This ECM layer was depleted from either EMILIN1 or FBN1 by using siRNA targeting of the SMCs. Cultured endothelial cells (ECs) on this modified ECM layer showed alterations on the transcriptome level of multiple pathways, especially the Rho GTPase controlled pathways. However, no significant alterations in adhesion, migration or proliferation were observed when ECs were cultured on EMILIN1- or FNB1-deficient ECM. To conclude, the proteome analysis identified unique ECM proteins involved in the embryonic development of renal arteries. Alterations in transcriptome levels of ECs cultured on EMILIN1- or FBN1-deficient ECM showed that these candidate proteins could affect the endothelial (regenerative) response.
Collapse
Affiliation(s)
- Christian G.M. van Dijk
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Laura Louzao-Martinez
- Center for Proteomics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (L.L.-M.); (J.A.A.D.)
| | - Elise van Mulligen
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Bart Boermans
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Jeroen A.A. Demmers
- Center for Proteomics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (L.L.-M.); (J.A.A.D.)
| | | | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands;
| | - Dirk J. Duncker
- Experimental Cardiology, Department of Cardiology, Thorax center, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands;
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Caroline Cheng
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
- Experimental Cardiology, Department of Cardiology, Thorax center, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands;
| |
Collapse
|
138
|
Role of Collagen Fiber Morphology on Ovarian Cancer Cell Migration Using Image-Based Models of the Extracellular Matrix. Cancers (Basel) 2020; 12:cancers12061390. [PMID: 32481580 PMCID: PMC7352517 DOI: 10.3390/cancers12061390] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Remodeling of the extracellular matrix (ECM) is an important part in the development and progression of many epithelial cancers. However, the biological significance of collagen alterations in ovarian cancer has not been well established. Here we investigated the role of collagen fiber morphology on cancer cell migration using tissue engineered scaffolds based on high-resolution Second-Harmonic Generation (SHG) images of ovarian tumors. The collagen-based scaffolds are fabricated by multiphoton excited (MPE) polymerization, which is a freeform 3D method affording submicron resolution feature sizes (~0.5 µm). This capability allows the replication of the collagen fiber architecture, where we constructed models representing normal stroma, high-risk tissue, benign tumors, and high-grade tumors. These were seeded with normal and ovarian cancer cell lines to investigate the separate roles of the cell type and matrix morphology on migration dynamics. The primary finding is that key cell–matrix interactions such as motility, cell spreading, f-actin alignment, focal adhesion, and cadherin expression are mainly determined by the collagen fiber morphology to a larger extent than the initial cell type. Moreover, we found these aspects were all enhanced for cells on the highly aligned, high-grade tumor model. Conversely, the weakest corresponding responses were observed on the more random mesh-like normal stromal matrix, with the partially aligned benign tumor and high-risk models demonstrating intermediate behavior. These results are all consistent with a contact guidance mechanism. These models cannot be synthesized by other conventional fabrication methods, and we suggest this approach will enable a variety of studies in cancer biology.
Collapse
|
139
|
Xu JL, Lesniak A, Gowen AA. Predictive Modeling of the In Vitro Responses of Preosteoblastic MC3T3-E1 Cells on Polymeric Surfaces Using Fourier Transform Infrared Spectroscopy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:24466-24478. [PMID: 32374584 DOI: 10.1021/acsami.0c04261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Biomaterials' surface properties elicit diverse cellular responses in biomedical and biotechnological applications. Predicting the cell behavior on a polymeric surface is an ongoing challenge due to its complexity. This work proposes a novel modeling methodology based on attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy. Spectra were collected on wetted polymeric surfaces to incorporate both surface chemistry and information on water-polymer interactions. Results showed that predictive models built with spectra from wetted surfaces ("wet spectra") performed much better than models built using spectra acquired from dry surfaces ("dry spectra"), suggesting that the water-polymer interaction is critically important to the prediction of subsequent cell behavior. The best model was seen to predict total area of focal adhesions with coefficient of determination for prediction (R2P) of 0.94 and root-mean-square errors of prediction (RMSEP) of 4.03 μm2 when tested on an independent experimental set. This work offers new insights into our understanding of cell-biomaterial interactions. The presence of carboxyl groups in polymers promoted larger adhesion areas, yet the formation of carbonyl-to-water interaction decreased adhesion areas. Surface wettability, which was related to the water-polymer interaction, was proven to highly influence cell adhesion. The good predictive ability opens new possibilities for high throughput monitoring of cell attachment on polymeric substrates.
Collapse
Affiliation(s)
- Jun-Li Xu
- School of Biosystems and Food Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - Anna Lesniak
- School of Biosystems and Food Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aoife A Gowen
- School of Biosystems and Food Engineering, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
140
|
Kim SK, Jang SD, Kim H, Chung S, Park JK, Kuh HJ. Phenotypic Heterogeneity and Plasticity of Cancer Cell Migration in a Pancreatic Tumor Three-Dimensional Culture Model. Cancers (Basel) 2020; 12:cancers12051305. [PMID: 32455681 PMCID: PMC7281339 DOI: 10.3390/cancers12051305] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 01/01/2023] Open
Abstract
Invasive cancer cell migration is a key feature of metastatic human pancreatic ductal adenocarcinoma (PDAC), yet the underlying mechanisms remain poorly understood. Here, we investigated modes of cancer cell invasion using two pancreatic cancer cell lines with differential epithelial–mesenchymal status, PANC-1 and BxPC-3, under 3D culture conditions. Multicellular tumor spheroids (TSs) were grown in a collagen matrix co-cultured with pancreatic stellate cells (PSCs) using microchannel chips. PANC-1 cells showed individual migration from TSs via invadopodium formation. BxPC-3 cells showed plasticity between collective and individual migration in either mesenchymal mode, with filopodium-like protrusions, or blebby amoeboid mode. These two cell lines showed significantly different patterns of extracellular matrix (ECM) remodeling, with MMP-dependent degradation in a limited area of ECM around invadopodia for PANC-1 cells, or MMP-independent extensive deformation of ECM for BxPC-3 cells. Cancer cell migration out of the collagen channel significantly increased by PSCs and directional cancer cell migration was mediated by fibronectin deposited by PSCs. Our results highlight the phenotypic heterogeneity and plasticity of PDAC cell migration and ECM remodeling under 3D culture conditions. This 3D co-culture model of pancreatic cancer cells and PSCs offers a useful tool for studying cancer cell migration and ECM remodeling to identify and develop potential molecular targets and anti-cancer agents against human PDAC.
Collapse
Affiliation(s)
- Seul-Ki Kim
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea; (S.-K.K.); (S.D.J.)
| | - So Dam Jang
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea; (S.-K.K.); (S.D.J.)
| | - Hyunho Kim
- School of Mechanical Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (H.K.); (S.C.)
| | - Seok Chung
- School of Mechanical Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (H.K.); (S.C.)
| | - Jong Kook Park
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Korea;
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea; (S.-K.K.); (S.D.J.)
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
141
|
Angely C, Ladant D, Planus E, Louis B, Filoche M, Chenal A, Isabey D. Functional and structural consequences of epithelial cell invasion by Bordetella pertussis adenylate cyclase toxin. PLoS One 2020; 15:e0228606. [PMID: 32392246 PMCID: PMC7213728 DOI: 10.1371/journal.pone.0228606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/18/2020] [Indexed: 01/13/2023] Open
Abstract
Bordetella pertussis, the causative agent of whopping cough, produces an adenylate cyclase toxin (CyaA) that plays a key role in the host colonization by targeting innate immune cells which express CD11b/CD18, the cellular receptor of CyaA. CyaA is also able to invade non-phagocytic cells, via a unique entry pathway consisting in a direct translocation of its catalytic domain across the cytoplasmic membrane of the cells. Within the cells, CyaA is activated by calmodulin to produce high levels of cyclic adenosine monophosphate (cAMP) and alter cellular physiology. In this study, we explored the effects of CyaA toxin on the cellular and molecular structure remodeling of A549 alveolar epithelial cells. Using classical imaging techniques, biochemical and functional tests, as well as advanced cell mechanics method, we quantify the structural and functional consequences of the massive increase of intracellular cyclic AMP induced by the toxin: cell shape rounding associated to adhesion weakening process, actin structure remodeling for the cortical and dense components, increase in cytoskeleton stiffness, and inhibition of migration and repair. We also show that, at low concentrations (0.5 nM), CyaA could significantly impair the migration and wound healing capacities of the intoxicated alveolar epithelial cells. As such concentrations might be reached locally during B. pertussis infection, our results suggest that the CyaA, beyond its major role in disabling innate immune cells, might also contribute to the local alteration of the epithelial barrier of the respiratory tract, a hallmark of pertussis.
Collapse
Affiliation(s)
- Christelle Angely
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires (CNRS UMR 3528), Département de Biologie Structurale et Chimie, Institut Pasteur, Paris, France
| | - Emmanuelle Planus
- Institut pour l’Avancée des Biosciences (IAB), Centre de Recherche UGA/ Inserm U1209 / CNRS UMR 5309, La Tronche, France
| | - Bruno Louis
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
| | - Marcel Filoche
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
- Laboratoire de Physique de la Matière Condensée, Ecole Polytechnique, CNRS, IP Paris, Palaiseau, France
| | - Alexandre Chenal
- Unité de Biochimie des Interactions Macromoléculaires (CNRS UMR 3528), Département de Biologie Structurale et Chimie, Institut Pasteur, Paris, France
| | - Daniel Isabey
- Equipe 13, Biomécanique & Appareil Respiratoire, Inserm U955, Créteil, France
- UMR 955, UPEC, Université Paris-Est, Créteil, France
- ERL 7000, CNRS, Créteil, France
- * E-mail:
| |
Collapse
|
142
|
Mojena-Medina D, Martínez-Hernández M, de la Fuente M, García-Isla G, Posada J, Jorcano JL, Acedo P. Design, Implementation, and Validation of a Piezoelectric Device to Study the Effects of Dynamic Mechanical Stimulation on Cell Proliferation, Migration and Morphology. SENSORS 2020; 20:s20072155. [PMID: 32290334 PMCID: PMC7180771 DOI: 10.3390/s20072155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
Cell functions and behavior are regulated not only by soluble (biochemical) signals but also by biophysical and mechanical cues within the cells' microenvironment. Thanks to the dynamical and complex cell machinery, cells are genuine and effective mechanotransducers translating mechanical stimuli into biochemical signals, which eventually alter multiple aspects of their own homeostasis. Given the dominant and classic biochemical-based views to explain biological processes, it could be challenging to elucidate the key role that mechanical parameters such as vibration, frequency, and force play in biology. Gaining a better understanding of how mechanical stimuli (and their mechanical parameters associated) affect biological outcomes relies partially on the availability of experimental tools that may allow researchers to alter mechanically the cell's microenvironment and observe cell responses. Here, we introduce a new device to study in vitro responses of cells to dynamic mechanical stimulation using a piezoelectric membrane. Using this device, we can flexibly change the parameters of the dynamic mechanical stimulation (frequency, amplitude, and duration of the stimuli), which increases the possibility to study the cell behavior under different mechanical excitations. We report on the design and implementation of such device and the characterization of its dynamic mechanical properties. By using this device, we have performed a preliminary study on the effect of dynamic mechanical stimulation in a cell monolayer of an epidermal cell line (HaCaT) studying the effects of 1 Hz and 80 Hz excitation frequencies (in the dynamic stimuli) on HaCaT cell migration, proliferation, and morphology. Our preliminary results indicate that the response of HaCaT is dependent on the frequency of stimulation. The device is economic, easily replicated in other laboratories and can support research for a better understanding of mechanisms mediating cellular mechanotransduction.
Collapse
Affiliation(s)
- Dahiana Mojena-Medina
- Department of Electronics Technology, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (J.P.); (P.A.)
- Correspondence:
| | - Marina Martínez-Hernández
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (M.M.-H.); (M.d.l.F.); (G.G.-I.); (J.L.J.)
| | - Miguel de la Fuente
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (M.M.-H.); (M.d.l.F.); (G.G.-I.); (J.L.J.)
| | - Guadalupe García-Isla
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (M.M.-H.); (M.d.l.F.); (G.G.-I.); (J.L.J.)
| | - Julio Posada
- Department of Electronics Technology, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (J.P.); (P.A.)
| | - José Luis Jorcano
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (M.M.-H.); (M.d.l.F.); (G.G.-I.); (J.L.J.)
| | - Pablo Acedo
- Department of Electronics Technology, Universidad Carlos III de Madrid, 28911 Madrid, Spain; (J.P.); (P.A.)
| |
Collapse
|
143
|
Miller AE, Hu P, Barker TH. Feeling Things Out: Bidirectional Signaling of the Cell-ECM Interface, Implications in the Mechanobiology of Cell Spreading, Migration, Proliferation, and Differentiation. Adv Healthc Mater 2020; 9:e1901445. [PMID: 32037719 PMCID: PMC7274903 DOI: 10.1002/adhm.201901445] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Biophysical cues stemming from the extracellular environment are rapidly transduced into discernible chemical messages (mechanotransduction) that direct cellular activities-placing the extracellular matrix (ECM) as a potent regulator of cell behavior. Dynamic reciprocity between the cell and its associated matrix is essential to the maintenance of tissue homeostasis and dysregulation of both ECM mechanical signaling, via pathological ECM turnover, and internal mechanotransduction pathways contribute to disease progression. This review covers the current understandings of the key modes of signaling used by both the cell and ECM to coregulate one another. By taking an outside-in approach, the inherent complexities and regulatory processes at each level of signaling (ECM, plasma membrane, focal adhesion, and cytoplasm) are captured to give a comprehensive picture of the internal and external mechanoregulatory environment. Specific emphasis is placed on the focal adhesion complex which acts as a central hub of mechanical signaling, regulating cell spreading, migration, proliferation, and differentiation. In addition, a wealth of available knowledge on mechanotransduction is curated to generate an integrated signaling network encompassing the central components of the focal adhesion, cytoplasm and nucleus that act in concert to promote durotaxis, proliferation, and differentiation in a stiffness-dependent manner.
Collapse
Affiliation(s)
- Andrew E Miller
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Ping Hu
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| |
Collapse
|
144
|
Porazinski S, Parkin A, Pajic M. Rho-ROCK Signaling in Normal Physiology and as a Key Player in Shaping the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:99-127. [PMID: 32030687 DOI: 10.1007/978-3-030-35582-1_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Rho-ROCK signaling network has a range of specialized functions of key biological importance, including control of essential developmental processes such as morphogenesis and physiological processes including homeostasis, immunity, and wound healing. Deregulation of Rho-ROCK signaling actively contributes to multiple pathological conditions, and plays a major role in cancer development and progression. This dynamic network is critical in modulating the intricate communication between tumor cells, surrounding diverse stromal cells and the matrix, shaping the ever-changing microenvironment of aggressive tumors. In this chapter, we overview the complex regulation of the Rho-ROCK signaling axis, its role in health and disease, and analyze progress made with key approaches targeting the Rho-ROCK pathway for therapeutic benefit. Finally, we conclude by outlining likely future trends and key questions in the field of Rho-ROCK research, in particular surrounding Rho-ROCK signaling within the tumor microenvironment.
Collapse
Affiliation(s)
- Sean Porazinski
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Ashleigh Parkin
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Marina Pajic
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia.
| |
Collapse
|
145
|
Li W, Zhao J, Wang J, Sun L, Xu H, Sun W, Pan Y, Wang H, Zhang WB. ROCK-TAZ signaling axis regulates mechanical tension-induced osteogenic differentiation of rat cranial sagittal suture mesenchymal stem cells. J Cell Physiol 2020; 235:5972-5984. [PMID: 31970784 DOI: 10.1002/jcp.29522] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022]
Abstract
Mechanical force across sutures is able to promote suture osteogenesis. Orthodontic clinics often use this biological characteristic of sutures to treat congenital cranio-maxillofacial malformations. However, the underlying mechanisms still remain poorly understood. Craniofacial sutures provide a special growth source and support primary sites of osteogenesis. Here, we isolated rat sagittal suture cells (rSAGs), which had mesenchymal stem cell characteristics and differentiating abilities. Cells were then subjected to mechanical tension (5% elongation, 0.5 Hz; sinusoidal waveforms) showing that mechanical tension could enhance osteogenic differentiation but hardly affect proliferation of rSAGs. Besides, mechanical tension could increase Rho-associated kinase (ROCK) expression and enhance transcriptional coactivator with PDZ-binding motif (TAZ) nuclear translocation. Inhibiting ROCK expression could suppress tension-induced osteogenesis and block tension-induced upregulation of nuclear TAZ. In addition, our results indicated that TAZ had direct combination sites with runt-related transcription factor 2 (Runx2) in rSAGs, and knock-downed TAZ simultaneously decreased the expression of Runx2 no matter with or without mechanical tension. In summary, our findings demonstrated that the multipotency of rSAGs in vitro could give rise to early osteogenic differentiation under mechanical tension, which was mediated by ROCK-TAZ signal axis.
Collapse
Affiliation(s)
- Wenlei Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Jialu Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Lian Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Haiyang Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yongchu Pan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hua Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Wei-Bing Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
146
|
Makhija EP, Espinosa-Hoyos D, Jagielska A, Van Vliet KJ. Mechanical regulation of oligodendrocyte biology. Neurosci Lett 2020; 717:134673. [PMID: 31838017 PMCID: PMC12023767 DOI: 10.1016/j.neulet.2019.134673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 12/27/2022]
Abstract
Oligodendrocytes (OL) are a subset of glial cells in the central nervous system (CNS) comprising the brain and spinal cord. The CNS environment is defined by complex biochemical and biophysical cues during development and response to injury or disease. In the last decade, significant progress has been made in understanding some of the key biophysical factors in the CNS that modulate OL biology, including their key role in myelination of neurons. Taken together, those studies offer translational implications for remyelination therapies, pharmacological research, identification of novel drug targets, and improvements in methods to generate human oligodendrocyte progenitor cells (OPCs) and OLs from donor stem cells in vitro. This review summarizes current knowledge of how various physical and mechanical cues affect OL biology and its implications for disease, therapeutic approaches, and generation of human OPCs and OLs.
Collapse
Affiliation(s)
- Ekta P Makhija
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Critical Analytics for Manufacturing Personalized-Medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, 138602, Singapore
| | - Daniela Espinosa-Hoyos
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Anna Jagielska
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| | - Krystyn J Van Vliet
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Critical Analytics for Manufacturing Personalized-Medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, 138602, Singapore; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| |
Collapse
|
147
|
Kim MH, Kino-Oka M. Bioengineering Considerations for a Nurturing Way to Enhance Scalable Expansion of Human Pluripotent Stem Cells. Biotechnol J 2020; 15:e1900314. [PMID: 31904180 DOI: 10.1002/biot.201900314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Understanding how defects in mechanotransduction affect cell-to-cell variability will add to the fundamental knowledge of human pluripotent stem cell (hPSC) culture, and may suggest new approaches for achieving a robust, reproducible, and scalable process that result in consistent product quality and yields. Here, the current state of the understanding of the fundamental mechanisms that govern the growth kinetics of hPSCs between static and dynamic cultures is reviewed, the factors causing fluctuations are identified, and culture strategies that might eliminate or minimize the occurrence of cell-to-cell variability arising from these fluctuations are discussed. The existing challenges in the development of hPSC expansion methods for enabling the transition from process development to large-scale production are addressed, a mandatory step for industrial and clinical applications of hPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
148
|
Bordeleau F, Wang W, Simmons A, Antonyak MA, Cerione RA, Reinhart-King CA. Tissue transglutaminase 2 regulates tumor cell tensional homeostasis by increasing contractility. J Cell Sci 2020; 133:jcs.231134. [PMID: 31822629 DOI: 10.1242/jcs.231134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/01/2019] [Indexed: 12/21/2022] Open
Abstract
Abnormal tensional cellular homeostasis is now considered a hallmark of cancer. Despite this, the origin of this abnormality remains unclear. In this work, we investigated the role of tissue transglutaminase 2 (TG2, also known as TGM2), a protein associated with poor prognosis and increased metastatic potential, and its relationship to the EGF receptor in the regulation of the mechanical state of tumor cells. Remarkably, we observed a TG2-mediated modulation of focal adhesion composition as well as stiffness-induced FAK activation, which was linked with a distinctive increase in cell contractility, in experiments using both pharmacological and shRNA-based approaches. Additionally, the increased contractility could be reproduced in non-malignant cells upon TG2 expression. Moreover, the increased cell contractility mediated by TG2 was largely due to the loss of EGFR-mediated inhibition of cell contractility. These findings establish intracellular TG2 as a regulator of cellular tensional homeostasis and suggest the existence of signaling switches that control the contribution of growth factor receptors in determining the mechanical state of a cell.
Collapse
Affiliation(s)
- Francois Bordeleau
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Faculty of Medecine, Université Laval, Québec G1R 3S3, Canada .,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Alysha Simmons
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
| | - Marc A Antonyak
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Richard A Cerione
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
149
|
Li J, Barbone PE, Smith ML, Stamenović D. Effect of correlation between traction forces on tensional homeostasis in clusters of endothelial cells and fibroblasts. J Biomech 2019; 100:109588. [PMID: 31902611 DOI: 10.1016/j.jbiomech.2019.109588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/06/2019] [Accepted: 12/22/2019] [Indexed: 10/25/2022]
Abstract
The ability of cells to maintain a constant level of cytoskeletal tension in response to external and internal disturbances is referred to as tensional homeostasis. It is essential for the normal physiological function of cells and tissues, and for protection against disease progression, including atherosclerosis and cancer. In previous studies, we defined tensional homeostasis as the ability of cells to maintain a consistent level of cytoskeletal tension with low temporal fluctuations. In those studies, we measured temporal fluctuations of cell-substrate traction forces in clusters of endothelial cells and of fibroblasts. We observed those temporal fluctuations to decrease with increasing cluster size in endothelial cells, but not in fibroblasts. We quantified temporal fluctuation, and thus homeostasis, through the coefficient of variation (CV) of the traction field; the lower the value of CV, the closer the cell is to the state of tensional homeostasis. This metric depends on correlation between individual traction forces. In this study, we analyzed the contribution of correlation between traction forces on traction field CV in clusters of endothelial cells and fibroblasts using experimental data that we had obtained previously. Results of our analysis showed that positive correlation between traction forces was detrimental to homeostasis, and that it was cell type-dependent.
Collapse
Affiliation(s)
- Juanyong Li
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, United States
| | - Paul E Barbone
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, United States
| | - Michael L Smith
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Dimitrije Stamenović
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States; Division of Material Science and Engineering, Boston University, Brookline, MA 02446, United States.
| |
Collapse
|
150
|
Vania V, Wang L, Tjakra M, Zhang T, Qiu J, Tan Y, Wang G. The interplay of signaling pathway in endothelial cells-matrix stiffness dependency with targeted-therapeutic drugs. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165645. [PMID: 31866415 DOI: 10.1016/j.bbadis.2019.165645] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/17/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVDs) have been one of the major causes of human deaths in the world. The study of CVDs has focused on cell chemotaxis for decades. With the advances in mechanobiology, accumulating evidence has demonstrated the influence of mechanical stimuli on arterial pathophysiology and endothelial dysfunction that is a hallmark of atherosclerosis development. An increasing number of drugs have been exploited to decrease the stiffness of vascular tissue for CVDs therapy. However, the underlying mechanisms have yet to be explored. This review aims to summarize how matrix stiffness mediates atherogenesis through various important signaling pathways in endothelial cells and cellular mechanophenotype, including RhoA/Rho-associated protein kinase (ROCK), mitogen-activated protein kinase (MAPK), and Hippo pathways. We also highlight the roles of putative mechanosensitive non-coding RNAs in matrix stiffness-mediated atherogenesis. Finally, we describe the usage of tunable hydrogel and its future strategy to improve our knowledge underlying matrix stiffness-mediated CVDs mechanism.
Collapse
Affiliation(s)
- Vicki Vania
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lu Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Marco Tjakra
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Tao Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|