1
|
Eschenbruch J, Dreissen G, Springer R, Konrad J, Merkel R, Hoffmann B, Noetzel E. From Microspikes to Stress Fibers: Actin Remodeling in Breast Acini Drives Myosin II-Mediated Basement Membrane Invasion. Cells 2021; 10:cells10081979. [PMID: 34440749 PMCID: PMC8394122 DOI: 10.3390/cells10081979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
The cellular mechanisms of basement membrane (BM) invasion remain poorly understood. We investigated the invasion-promoting mechanisms of actin cytoskeleton reorganization in BM-covered MCF10A breast acini. High-resolution confocal microscopy has characterized actin cell protrusion formation and function in response to tumor-resembling ECM stiffness and soluble EGF stimulation. Traction force microscopy quantified the mechanical BM stresses that invasion-triggered acini exerted on the BM-ECM interface. We demonstrate that acini use non-proteolytic actin microspikes as functional precursors of elongated protrusions to initiate BM penetration and ECM probing. Further, these microspikes mechanically widened the collagen IV pores to anchor within the BM scaffold via force-transmitting focal adhesions. Pre-invasive basal cells located at the BM-ECM interface exhibited predominantly cortical actin networks and actin microspikes. In response to pro-invasive conditions, these microspikes accumulated and converted subsequently into highly contractile stress fibers. The phenotypical switch to stress fiber cells matched spatiotemporally with emerging high BM stresses that were driven by actomyosin II contractility. The activation of proteolytic invadopodia with MT1-MMP occurred at later BM invasion stages and only in cells already disseminating into the ECM. Our study demonstrates that BM pore-widening filopodia bridge mechanical ECM probing function and contractility-driven BM weakening. Finally, these EMT-related cytoskeletal adaptations are critical mechanisms inducing the invasive transition of benign breast acini.
Collapse
|
2
|
Brock EJ, Jackson RM, Boerner JL, Li Q, Tennis MA, Sloane BF, Mattingly RR. Sprouty4 negatively regulates ERK/MAPK signaling and the transition from in situ to invasive breast ductal carcinoma. PLoS One 2021; 16:e0252314. [PMID: 34048471 PMCID: PMC8162601 DOI: 10.1371/journal.pone.0252314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/13/2021] [Indexed: 12/16/2022] Open
Abstract
Breast ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive ductal carcinoma (IDC). It is still unclear which DCIS will become invasive and which will remain indolent. Patients often receive surgery and radiotherapy, but this early intervention has not produced substantial decreases in late-stage disease. Sprouty proteins are important regulators of ERK/MAPK signaling and have been studied in various cancers. We hypothesized that Sprouty4 is an endogenous inhibitor of ERK/MAPK signaling and that its loss/reduced expression is a mechanism by which DCIS lesions progress toward IDC, including triple-negative disease. Using immunohistochemistry, we found reduced Sprouty4 expression in IDC patient samples compared to DCIS, and that ERK/MAPK phosphorylation had an inverse relationship to Sprouty4 expression. These observations were reproduced using a 3D culture model of disease progression. Knockdown of Sprouty4 in MCF10.DCIS cells increased ERK/MAPK phosphorylation as well as their invasive capability, while overexpression of Sprouty4 in MCF10.CA1d IDC cells reduced ERK/MAPK phosphorylation, invasion, and the aggressive phenotype exhibited by these cells. Immunofluorescence experiments revealed reorganization of the actin cytoskeleton and relocation of E-cadherin back to the cell surface, consistent with the restoration of adherens junctions. To determine whether these effects were due to changes in ERK/MAPK signaling, MEK1/2 was pharmacologically inhibited in IDC cells. Nanomolar concentrations of MEK162/binimetinib restored an epithelial-like phenotype and reduced pericellular proteolysis, similar to Sprouty4 overexpression. From these data we conclude that Sprouty4 acts to control ERK/MAPK signaling in DCIS, thus limiting the progression of these premalignant breast lesions.
Collapse
MESH Headings
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Female
- Humans
- Immunoblotting
- Immunohistochemistry
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
Collapse
Affiliation(s)
- Ethan J. Brock
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United states of America
| | - Ryan M. Jackson
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United states of America
| | - Julie L. Boerner
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United states of America
| | - Quanwen Li
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United states of America
| | - Meredith A. Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United states of America
| | - Bonnie F. Sloane
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United states of America
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United states of America
| | - Raymond R. Mattingly
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United states of America
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United states of America
| |
Collapse
|
3
|
Cell Force-Driven Basement Membrane Disruption Fuels EGF- and Stiffness-Induced Invasive Cell Dissemination from Benign Breast Gland Acini. Int J Mol Sci 2021; 22:ijms22083962. [PMID: 33921304 PMCID: PMC8070162 DOI: 10.3390/ijms22083962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/30/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Local basement membrane (BM) disruption marks the initial step of breast cancer invasion. The activation mechanisms of force-driven BM-weakening remain elusive. We studied the mechanical response of MCF10A-derived human breast cell acini with BMs of tuneable maturation to physical and soluble tumour-like extracellular matrix (ECM) cues. Traction force microscopy (TFM) and elastic resonator interference stress microscopy (ERISM) were used to quantify pro-invasive BM stress and protrusive forces. Substrate stiffening and mechanically impaired BM scaffolds induced the invasive transition of benign acini synergistically. Robust BM scaffolds attenuated this invasive response. Additional oncogenic EGFR activation compromised the BMs’ barrier function, fuelling invasion speed and incidence. Mechanistically, EGFR-PI3-Kinase downstream signalling modulated both MMP- and force-driven BM-weakening processes. We show that breast acini form non-proteolytic and BM-piercing filopodia for continuous matrix mechanosensation, which significantly push and pull on the BM and ECM under pro-invasive conditions. Invasion-triggered acini further shear and compress their BM by contractility-based stresses that were significantly increased (3.7-fold) compared to non-invasive conditions. Overall, the highest amplitudes of protrusive and contractile forces accompanied the highest invasiveness. This work provides a mechanistic concept for tumour ECM-induced mechanically misbalanced breast glands fuelling force-driven BM disruption. Finally, this could facilitate early cell dissemination from pre-invasive lesions to metastasize eventually.
Collapse
|
4
|
Dura G, Crespo-Cuadrado M, Waller H, Peters DT, Ferreira AM, Lakey JH, Fulton DA. Hydrogels of engineered bacterial fimbriae can finely tune 2D human cell culture. Biomater Sci 2021; 9:2542-2552. [PMID: 33571331 DOI: 10.1039/d0bm01966f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Demand continues to grow for biomimetic materials able to create well-defined environments for modulating the behaviour of living cells in culture. Here, we describe hydrogels based upon the polymeric bacterial fimbriae protein capsular antigen fragment 1 (Caf1) that presents tunable biological properties for enhanced tissue cell culture applications. We demonstrate how Caf1 hydrogels can regulate cellular functions such as spreading, proliferation and matrix deposition of human dermal fibroblast cells (hDFBs). Caf1 hydrogels exploring a range of mechanical properties were prepared using copolymers featuring controlled compositions of inert wild-type Caf1 subunits and a mutant subunit displaying the RGDS peptide motif. The hydrogels showed excellent cytocompatibility with hDFBs and the ability to modulate both cell morphology and matrix deposition. Interestingly, Caf1 hydrogels displaying faster stress relaxation were demonstrated to show the highest metabolic activities of growing cells in comparison with other Caf1 hydrogel formulations. The stiffest Caf1 hydrogel impacted cellular morphology, inducing alignment of the cells. This work is significant as it clearly indicates that Caf1-based hydrogels offer tuneable biochemical and mechanical substrates conditions suitable for cell culture applications.
Collapse
Affiliation(s)
- Gema Dura
- Chemical Nanoscience Laboratory, Chemistry-School of Natural and Environmental Sciences, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK.
| | | | | | | | | | | | | |
Collapse
|
5
|
Abstract
Viral infections lead to the death of more than a million people each year around the world, both directly and indirectly. Viruses interfere with many cell functions, particularly critical pathways for cell death, by affecting various intracellular mediators. MicroRNAs (miRNAs) are a major example of these mediators because they are involved in many (if not most) cellular mechanisms. Virus-regulated miRNAs have been implicated in three cell death pathways, namely, apoptosis, autophagy, and anoikis. Several molecules (e.g., BECN1 and B cell lymphoma 2 [BCL2] family members) are involved in both apoptosis and autophagy, while activation of anoikis leads to cell death similar to apoptosis. These mechanistic similarities suggest that common regulators, including some miRNAs (e.g., miR-21 and miR-192), are involved in different cell death pathways. Because the balance between cell proliferation and cell death is pivotal to the homeostasis of the human body, miRNAs that regulate cell death pathways have drawn much attention from researchers. miR-21 is regulated by several viruses and can affect both apoptosis and anoikis via modulating various targets, such as PDCD4, PTEN, interleukin (IL)-12, Maspin, and Fas-L. miR-34 can be downregulated by viral infection and has different effects on apoptosis, depending on the type of virus and/or host cell. The present review summarizes the existing knowledge on virus-regulated miRNAs involved in the modulation of cell death pathways. Understanding the mechanisms for virus-mediated regulation of cell death pathways could provide valuable information to improve the diagnosis and treatment of many viral diseases.
Collapse
|
6
|
The Tumor Microenvironment of Primitive and Metastatic Breast Cancer: Implications for Novel Therapeutic Strategies. Int J Mol Sci 2020; 21:ijms21218102. [PMID: 33143050 PMCID: PMC7662409 DOI: 10.3390/ijms21218102] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer evolves thanks to a dense and close interaction with the surrounding tumor microenvironment (TME). Fibroblasts, leukocytes, blood and lymphatic endothelial cells and extracellular matrix are the constituents of this entity, and they synergistically play a pivotal role in all of the stages of breast cancer development, from its onset to its metastatic spread. Moreover, it has been widely demonstrated that variations to the TME can correspond to prognosis variations. Breast cancer not only modulates the transformation of the environment within the mammary gland, but the same process is observed in metastases as well. In this minireview, we describe the features of TME within the primitive breast cancer, throughout its evolution and spread into the main metastatic sites.
Collapse
|
7
|
Karve K, Netherton S, Deng L, Bonni A, Bonni S. Regulation of epithelial-mesenchymal transition and organoid morphogenesis by a novel TGFβ-TCF7L2 isoform-specific signaling pathway. Cell Death Dis 2020; 11:704. [PMID: 32843642 PMCID: PMC7447769 DOI: 10.1038/s41419-020-02905-z] [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: 12/16/2019] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022]
Abstract
Alternative splicing contributes to diversification of gene function, yet consequences of splicing on functions of specific gene products is poorly understood. The major transcription factor TCF7L2 undergoes alternative splicing but the biological significance of TCF7L2 isoforms has remained largely to be elucidated. Here, we find that the TCF7L2 E-isoforms maintain, whereas the M and S isoforms disrupt morphogenesis of 3D-epithelial cell-derived organoids via regulation of epithelial-mesenchymal transition (EMT). Remarkably, TCF7L2E2 antagonizes, whereas TCF7L2M2/S2 promotes EMT-like effects in epithelial cells induced by transforming growth factor beta (TGFβ) signaling. In addition, we find TGFβ signaling reduces the proportion of TCF7L2E to TCF7L2M/S protein in cells undergoing EMT. We also find that TCF7L2 operates via TGFβ-Smad3 signaling to regulate EMT. Collectively, our findings unveil novel isoform-specific functions for the major transcription factor TCF7L2 and provide novel links between TCF7L2 and TGFβ signaling in the control of EMT-like responses and epithelial tissue morphogenesis.
Collapse
Affiliation(s)
- Kunal Karve
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Stuart Netherton
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lili Deng
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shirin Bonni
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
8
|
Uncovering mutation-specific morphogenic phenotypes and paracrine-mediated vessel dysfunction in a biomimetic vascularized mammary duct platform. Nat Commun 2020; 11:3377. [PMID: 32632100 PMCID: PMC7338408 DOI: 10.1038/s41467-020-17102-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 06/05/2020] [Indexed: 12/15/2022] Open
Abstract
The mammary gland is a highly vascularized tissue capable of expansion and regression during development and disease. To enable mechanistic insight into the coordinated morphogenic crosstalk between the epithelium and vasculature, we introduce a 3D microfluidic platform that juxtaposes a human mammary duct in proximity to a perfused endothelial vessel. Both compartments recapitulate stable architectural features of native tissue and the ability to undergo distinct forms of branching morphogenesis. Modeling HER2/ERBB2 amplification or activating PIK3CA(H1047R) mutation each produces ductal changes observed in invasive progression, yet with striking morphogenic and behavioral differences. Interestingly, PI3KαH1047R ducts also elicit increased permeability and structural disorganization of the endothelium, and we identify the distinct secretion of IL-6 as the paracrine cause of PI3KαH1047R-associated vascular dysfunction. These results demonstrate the functionality of a model system that facilitates the dissection of 3D morphogenic behaviors and bidirectional signaling between mammary epithelium and endothelium during homeostasis and pathogenesis.
Collapse
|
9
|
3D multicellular models to study the regulation and roles of acid-base transporters in breast cancer. Biochem Soc Trans 2019; 47:1689-1700. [PMID: 31803922 DOI: 10.1042/bst20190131] [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: 09/05/2019] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022]
Abstract
As a result of elevated metabolic rates and net acid extrusion in the rapidly proliferating cancer cells, solid tumours are characterized by a highly acidic microenvironment, while cancer cell intracellular pH is normal or even alkaline. Two-dimensional (2D) cell monocultures, which have been used extensively in breast cancer research for decades, cannot precisely recapitulate the rich environment and complex processes occurring in tumours in vivo. The use of such models can consequently be misleading or non-predictive for clinical applications. Models mimicking the tumour microenvironment are particularly pivotal for studying tumour pH homeostasis, which is profoundly affected by the diffusion-limited conditions in the tumour. To advance the understanding of the mechanisms and consequences of dysregulated acid-base homeostasis in breast cancer, clinically relevant models that incorporate the unique microenvironment of these tumours are required. The development of three-dimensional (3D) cell cultures has provided new tools for basic research and pre-clinical approaches, allowing the culture of breast cancer cells under conditions that closely resemble tumour growth in a living organism. Here we provide an overview of the main 3D techniques relevant for breast cancer cell culture. We discuss the advantages and limitations of the classical 3D models as well as recent advances in 3D culture techniques, focusing on how these culture methods have been used to study acid-base transport in breast cancer. Finally, we outline future directions of 3D culture technology and their relevance for studies of acid-base transport.
Collapse
|
10
|
Connor Y, Tekleab Y, Tekleab S, Nandakumar S, Bharat D, Sengupta S. A mathematical model of tumor-endothelial interactions in a 3D co-culture. Sci Rep 2019; 9:8429. [PMID: 31182723 PMCID: PMC6557844 DOI: 10.1038/s41598-019-44713-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 05/23/2019] [Indexed: 11/09/2022] Open
Abstract
Intravasation and extravasation of cancer cells through blood/lymph vessel endothelium are essential steps during metastasis. Successful invasion requires coordinated tumor-endothelial crosstalk, utilizing mechanochemical signaling to direct cytoskeletal rearrangement for transmigration of cancer cells. However, mechanisms underlying physical interactions are difficult to observe due to the lack of experimental models easily combined with theoretical models that better elucidate these pathways. We have previously demonstrated that an engineered 3D in vitro endothelial-epithelial co-culture system can be used to isolate both molecular and physical tumor-endothelial interactions in a platform that is easily modeled, quantified, and probed for experimental investigation. Using this platform with mathematical modeling, we show that breast metastatic cells display unique behavior with the endothelium, exhibiting a 3.2-fold increase in interaction with the endothelium and a 61-fold increase in elongation compared to normal breast epithelial cells. Our mathematical model suggests energetic favorability for cellular deformation prior to breeching endothelial junctions, expending less energy as compared to undeformed cells, which is consistent with the observed phenotype. Finally, we show experimentally that pharmacological inhibition of the cytoskeleton can disrupt the elongatation and alignment of metastatic cells with endothelial tubes, reverting to a less invasive phenotype.
Collapse
Affiliation(s)
- Yamicia Connor
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, 02139, USA.,Brigham and Women's Hospital, Department of Medicine, Boston, MA, 02115, USA.,Harvard Medical School, Health Sciences & Technology, Boston, MA, 02115, USA.,Beth Israel Deaconess Medical Center, Department of Medicine, Boston, MA, 02215, USA
| | - Yonatan Tekleab
- Massachusetts Institute of Technology, Department of Aeronautics and Astronautics, Cambridge, MA, 02139, USA
| | - Sarah Tekleab
- Brigham and Women's Hospital, Department of Medicine, Boston, MA, 02115, USA
| | - Shyama Nandakumar
- Brigham and Women's Hospital, Department of Medicine, Boston, MA, 02115, USA
| | - Divya Bharat
- Brigham and Women's Hospital, Department of Medicine, Boston, MA, 02115, USA
| | - Shiladitya Sengupta
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, 02139, USA. .,Brigham and Women's Hospital, Department of Medicine, Boston, MA, 02115, USA. .,Harvard Medical School, Health Sciences & Technology, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Florian S, Iwamoto Y, Coughlin M, Weissleder R, Mitchison TJ. A human organoid system that self-organizes to recapitulate growth and differentiation of a benign mammary tumor. Proc Natl Acad Sci U S A 2019; 116:11444-11453. [PMID: 31101720 PMCID: PMC6561274 DOI: 10.1073/pnas.1702372116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As 3D culture has become central to investigation of tissue biology, mammary epithelial organoids have emerged as powerful tools for investigation of epithelial cell polarization and carcinogenesis. However, most current protocols start from single cells suspended in Matrigel, which can also restrict cell differentiation and behavior. Here, we show that the noncancerous mammary cell line HMT-3522 S1, when allowed to spontaneously form cell aggregates ("spheroids") in medium without Matrigel, switches to a collective growth mode that recapitulates many attributes of "usual ductal hyperplasia" (UDH), a common benign mammary lesion. Interestingly, these spheroids undergo a complex maturation process reminiscent of embryonic development: solid-cell cords form their own basement membrane, grow on the surface of initially homogeneous cell aggregates, and form asymmetric lumina lined by two distinct cell types that express basal and luminal cytokeratins. This sequence of events provides a cellular mechanism that explains how the characteristic crescent-shaped, asymmetrical lumina form in UDH. Our results suggest that HMT-3522 S1 spheroids are useful as an in vitro model system to study UDH biology, glandular lumen formation, and stem cell biology of the mammary gland.
Collapse
Affiliation(s)
- Stefan Florian
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115;
- Institute of Pathology, Charité University Hospital, 10117 Berlin, Germany
| | - Yoshiko Iwamoto
- Center for Systems Biology, Richard B. Simches Research Center, Massachusetts General Hospital, Boston, MA 02114
| | - Margaret Coughlin
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - Ralph Weissleder
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
- Center for Systems Biology, Richard B. Simches Research Center, Massachusetts General Hospital, Boston, MA 02114
| | | |
Collapse
|
12
|
Siregar P, Julen N, Hufnagl P, Mutter G. A general framework dedicated to computational morphogenesis Part I - Constitutive equations. Biosystems 2018; 173:298-313. [PMID: 30005999 DOI: 10.1016/j.biosystems.2018.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/30/2018] [Accepted: 07/05/2018] [Indexed: 01/14/2023]
Abstract
In order to understand living organisms, considerable experimental efforts and resources have been devoted to correlate genes and their expressions with cell, tissue, organ and whole organisms' phenotypes. This data driven approach to knowledge discovery has led to many breakthrough in our understanding of healthy and diseased states, and is paving the way to improve the diagnosis and treatment of diseases. Complementary to this data-driven approach, computational models of biological systems based on first principles have been developed in order to deepen our understanding of the multi-scale dynamics that drives normal and pathological biological functions. In this paper we describe the biological, physical and mathematical concepts that led to the design of a Computational Morphogenesis (CM) platform baptized Generic Modeling and Simulating Platform (GMSP). Its role is to generate realistic 3D multi-scale biological tissues from virtual stem cells and the intended target applications include in virtuo studies of normal and abnormal tissue (re)generation as well as the development of complex diseases such as carcinogenesis. At all space-scales of interest, biological agents interact with each other via biochemical, bioelectrical, and mechanical fields that operate in concert during embryogenesis, growth and adult life. The spatio-temporal dependencies of these fields can be modeled by physics-based constitutive equations that we propose to examine in relation to the landmark biological events that occur during embryogenesis.
Collapse
Affiliation(s)
| | | | - Peter Hufnagl
- Department of Digital Pathology and IT, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - George Mutter
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
13
|
Avivar-Valderas A, McEwen R, Taheri-Ghahfarokhi A, Carnevalli LS, Hardaker EL, Maresca M, Hudson K, Harrington EA, Cruzalegui F. Functional significance of co-occurring mutations in PIK3CA and MAP3K1 in breast cancer. Oncotarget 2018; 9:21444-21458. [PMID: 29765551 PMCID: PMC5940413 DOI: 10.18632/oncotarget.25118] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/22/2018] [Indexed: 12/30/2022] Open
Abstract
The PI3Kα signaling pathway is frequently hyper-activated in breast cancer (BrCa), as a result of mutations/amplifications in oncogenes (e.g. HER2), decreased function in tumor suppressors (e.g. PTEN) or activating mutations in key components of the pathway. In particular, activating mutations of PIK3CA (~45%) are frequently found in luminal A BrCa samples. Genomic studies have uncovered inactivating mutations in MAP3K1 (13-20%) and MAP2K4 (~8%), two upstream kinases of the JNK apoptotic pathway in luminal A BrCa samples. Further, simultaneous mutation of PIK3CA and MAP3K1 are found in ~11% of mutant PIK3CA tumors. How these two alterations may cooperate to elicit tumorigenesis and impact the sensitivity to PI3K and AKT inhibitors is currently unknown. Using CRISPR gene editing we have genetically disrupted MAP3K1 expression in mutant PIK3CA cell lines to specifically create in vitro models reflecting the mutational status of PIK3CA and MAP3K1 in BrCa patients. MAP3K1 deficient cell lines exhibited ~2.4-fold increased proliferation rate and decreased sensitivity to PI3Kα/δ(AZD8835) and AKT (AZD5363) inhibitors (~2.61 and ~5.23-fold IC50 increases, respectively) compared with parental control cell lines. In addition, mechanistic analysis revealed that MAP3K1 disruption enhances AKT phosphorylation and downstream signaling and reduces sensitivity to AZD5363-mediated pathway inhibition. This appears to be a consequence of deficient MAP3K1-JNK signaling increasing IRS1 stability and therefore promoting IRS1 binding to p85, resulting in enhanced PI3Kα activity. Using 3D-MCF10A-PI3KαH1047R models, we found that MAP3K1 depletion increased overall acinar volume and counteracted AZD5363-mediated reduction of acinar growth due to enhanced proliferation and reduced apoptosis. Furthermore, in vivo efficacy studies revealed that MAP3K1-deficient MCF7 tumors were less sensitive to AKT inhibitor treatment, compared with parental MCF7 tumors. Our study provides mechanistic and in vivo evidence indicating a role for MAP3K1 as a tumor suppressor gene at least in the context of PIK3CA-mutant backgrounds. Further, our work predicts that MAP3K1 mutational status may be considered as a predictive biomarker for efficacy in PI3K pathway inhibitor trials.
Collapse
Affiliation(s)
- Alvaro Avivar-Valderas
- Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.,Current address: TiGenix, Parque Tecnológico de Madrid, Tres Cantos, Madrid, Spain
| | - Robert McEwen
- Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Amir Taheri-Ghahfarokhi
- Translational Genomics, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | | | - Marcello Maresca
- Translational Genomics, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Kevin Hudson
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.,Current address: 2TheNth, Adelphi Mill, Bollington, Macclesfield, UK
| | | | - Francisco Cruzalegui
- Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.,Current address: Pierre Fabre R&D Centre, Toulouse, France
| |
Collapse
|
14
|
DUOX1 Silencing in Mammary Cell Alters the Response to Genotoxic Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3570526. [PMID: 29849884 PMCID: PMC5933011 DOI: 10.1155/2018/3570526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/15/2018] [Indexed: 11/23/2022]
Abstract
DUOX1 is an H2O2-generating enzyme related to a wide range of biological features, such as hormone synthesis, host defense, cellular proliferation, and fertilization. DUOX1 is frequently downregulated in lung and liver cancers, suggesting a tumor suppressor role for this enzyme. Here, we show that DUOX1 expression is decreased in breast cancer cell lines and also in breast cancers when compared to the nontumor counterpart. In order to address the role of DUOX1 in breast cells, we stably knocked down the expression of DUOX1 in nontumor mammary cells (MCF12A) with shRNA. This led to higher cell proliferation rates and decreased migration and adhesion properties, which are typical features for transformed cells. After genotoxic stress induced by doxorubicin, DUOX1-silenced cells showed reduced IL-6 and IL-8 secretion and increased apoptosis levels. Furthermore, the cell proliferation rate was higher in DUOX1-silenced cells after doxorubicin medication in comparison to control cells. In conclusion, we demonstrate here that DUOX1 is silenced in breast cancer, which seems to be involved in breast carcinogenesis.
Collapse
|
15
|
Sweeney MF, Sonnenschein C, Soto AM. Characterization of MCF-12A cell phenotype, response to estrogens, and growth in 3D. Cancer Cell Int 2018; 18:43. [PMID: 29559854 PMCID: PMC5859508 DOI: 10.1186/s12935-018-0534-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/05/2018] [Indexed: 01/28/2023] Open
Abstract
Background Three-dimensional cultures of mammary epithelial cells allow for biologically-relevant studies of the development of the mammary gland in rodents and humans under normal and pathological conditions, like carcinogenesis. Under these conditions, mammotropic hormones play significant roles in tissue morphogenesis. Therefore, a system that recreates the normal, hormonally responsive epithelium would be a valuable tool to study the normal state and its transition to carcinogenesis. MCF-12A cells have been claimed to be non-tumorigenic mammary epithelial cells with reported sensitivity to estrogens. In this study, we aimed at characterizing MCF-12A cells for use in a hormone-responsive 3D culture system to determine their usefulness as a tool to identify normal and abnormal microenvironmental cues. Methods MCF-12A cells were single-cell cloned in order to investigate their heterogeneous makeup. The parental cells were then treated with estradiol to investigate proliferative and transcriptional responses through the estrogen receptor alpha. Finally, parental cells and epithelial-like cell-derived clones were seeded in rat-tail collagen I to profile the morphogenesis of multicellular 3D structures. The resultant structures were then analyzed using unsupervised morphometric analysis. Results MCF-12A cells consist of epithelial-like colonies which shed elongated, freely growing cells on the colony's edges. The cells express E-cadherin as well as mesenchymal vimentin but do not express markers associated with myoepithelial cells or fibroblasts. Treatment with estradiol does not affect either the proliferation rate or the induction of gene expression in MCF-12A cells. Parental MCF-12A cells form acini, solid spheres and elongated branching ducts when grown in rat-tail collagen type I matrix, the geometries and distribution of which are altered following the removal of fibroblast-like cells. Conclusions MCF-12A cells are a heterogeneous pseudo-epithelial cell line capable of forming a variety of multicellular structures in 3D culture. We found no indication that the cells display estrogen-responsive characteristics, thus refuting previous studies which reported estrogen responsiveness. We report that MCF-12A cells are not suited for use in studies in which differential behaviors of "normal" and "cancerous" estrogen-responsive cells are to be compared.
Collapse
Affiliation(s)
- Michael F Sweeney
- 1Sackler School of Graduate Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Carlos Sonnenschein
- 2Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Ana M Soto
- 2Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| |
Collapse
|
16
|
Liu X, Sun L, Gursel DB, Cheng C, Huang S, Rademaker AW, Khan SA, Yin J, Kiyokawa H. The non-canonical ubiquitin activating enzyme UBA6 suppresses epithelial-mesenchymal transition of mammary epithelial cells. Oncotarget 2017; 8:87480-87493. [PMID: 29152096 PMCID: PMC5675648 DOI: 10.18632/oncotarget.20900] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/17/2017] [Indexed: 12/23/2022] Open
Abstract
Ubiquitination plays critical roles in the regulation of oncoproteins and tumor suppressors during carcinogenesis. The two ubiquitin activating enzymes (E1) in human genome, UBA1 and UBA6, initiate ubiquitination by ATP-dependent activation of ubiquitin. Recent evidence suggests that UBA1 and UBA6 play partially overlapped yet distinct roles in controlling the proteome. Here we demonstrate that ubiquitination pathways initiated specifically by UBA6 set a suppressive barrier against critical steps of mammary carcinogenesis such as loss of polarity, anoikis resistance and epithelial-mesenchymal transition (EMT). Mammary epithelial MCF-10A cells expressing shRNA against UBA6 fail in establishing cell cycle arrest in response to detachment from extracellular matrix, confluency with fully engaged cell-cell contact or growth factor deprivation. Moreover, UBA6-deficient MCF-10A cells undergo spontaneous EMT under growth factor deprivation and exhibit accelerated kinetics of TGF-β-induced EMT. The Rho-GTPase CDC42 is one of the specific targets of UBA6-initiated ubiquitination and plays a key role in the function of UBA6 in controlling epithelial homeostasis, since a CDC42 inhibitor, ML141, rescues UBA6-deficient cells from the EMT phenotype. Immunohistochemical analysis of human breast cancer tissues demonstrates that 38% of invasive carcinomas express low or undetectable expression of UBA6, suggesting that downregulation of this non-canonical E1 plays a role in breast cancer development.
Collapse
Affiliation(s)
- Xianpeng Liu
- Department of Pharmacology, Northwestern University, Chicago, Illinois 60611, USA
| | - Limin Sun
- Department of Pharmacology, Northwestern University, Chicago, Illinois 60611, USA
| | - Demirkan B Gursel
- Department of Pathology, Northwestern University, Chicago, Illinois 60611, USA
| | - Chonghui Cheng
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA.,Current/Present address: Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sui Huang
- Department of Cell and Molecular Biology, Northwestern University, Chicago, Illinois 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA
| | - Alfred W Rademaker
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA
| | - Seema A Khan
- Department of Surgery, Northwestern University, Chicago, Illinois 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA
| | - Jun Yin
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, USA
| | - Hiroaki Kiyokawa
- Department of Pharmacology, Northwestern University, Chicago, Illinois 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
17
|
Seifert A, Posern G. Tightly controlled MRTF-A activity regulates epithelial differentiation during formation of mammary acini. Breast Cancer Res 2017; 19:68. [PMID: 28592291 PMCID: PMC5463372 DOI: 10.1186/s13058-017-0860-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/25/2017] [Indexed: 01/06/2023] Open
Abstract
Background Myocardin-related transcription factors (MRTF) A and B link actin dynamics and mechanotransduction to gene expression. In mice, MRTF-A is involved in mammary gland differentiation, but its role in human mammary epithelial cells remains unclear. Methods Three-dimensional cultures of human mammary epithelial MCF10A cells were used to model acinar morphogenesis. Stable MRTF-A knockdown, MRTF-A/B rescue and MRTF-A/B overexpression was established to characterize the functional role during morphogenesis using confocal microscopy and expression analysis. Breast cancer patient databases were analyzed for MRTF-A expression. Results We showed that a precise temporal control of MRTFs is required for normal morphogenesis of MCF10A mammary acini. MRTF transcriptional activity, but not their protein amounts, is transiently induced during 3D acini formation. MRTF-A knockdown dramatically reduces acini size and prevents lumen formation. These effects are rescued by re-expression of MRTF-A, and partially by MRTF-B. Conversely, overexpression of MRTF-A and MRTF-B increases acini size, resulting in irregular spheroids without lumen and defective apico-basal polarity. These phenotypes correlate with deregulated expression of cell cycle inhibitors p21/Waf1, p27/Kip1 and altered phosphorylation of retinoblastoma protein. In MRTF overexpressing spheroids, proliferation and apoptosis are simultaneously increased at late stages, whilst neither occurs in control acini. MRTFs interfere with anoikis of the inner cells and cause an integrin switch from α6 to α5, repression of E-cadherin and induction of mesenchymal markers vimentin, Snai2 and Zeb1. Moreover, MRTF-overexpressing spheroids are insensitive to alteration in matrix stiffness. In two breast cancer cohorts, high expression of MRTF-A and known target genes was associated with decreased patient survival. Conclusion MRTF-A is required for proliferation and formation of mammary acini from luminal epithelial cells. Conversely, elevated MRTF activity results in pre-malignant spheroid formation due to defective proliferation, polarity loss and epithelial-mesenchymal transition. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0860-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anja Seifert
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany.
| |
Collapse
|
18
|
Kamal M, Holliday DL, Morrison EE, Speirs V, Toomes C, Bell SM. Loss of CSMD1 expression disrupts mammary duct formation while enhancing proliferation, migration and invasion. Oncol Rep 2017; 38:283-292. [PMID: 28534981 DOI: 10.3892/or.2017.5656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
The CUB and sushi multiple domains 1 (CSMD1) gene maps to chromosome 8p23, a region deleted in many cancers. Loss of CSMD1 expression is associated with poor prognosis in breast cancer suggesting that it acts as a tumour suppressor in this cancer. However, the function of CSMD1 is largely unknown. Herein, we investigated CSMD1 functions in cell line models. CSMD1 expression was suppressed in MCF10A and LNCaP cells using short hairpin RNA. Functional assays were performed focusing on the 'normal' MCF10A cell line. Suppression of CSMD1 significantly increased the proliferation, cell migration and invasiveness of MCF10A cells compared to shcontrols. shCSMD1 cells also showed significantly reduced adhesion to Matrigel and fibronectin. In a three-dimensional Matrigel model of MCF10A cells, reduced CSMD1 expression resulted in the development of larger and more poorly differentiated breast acini-like structures that displayed impaired lumen formation. Loss of CSMD1 expression disrupts a model of mammary duct formation while enhancing proliferation, migration and invasion. Our data suggest that CSMD1 is involved in the suppression of a transformed phenotype.
Collapse
Affiliation(s)
- Mohamed Kamal
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Deborah L Holliday
- Leeds Institute of Cancer and Pathology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Ewan E Morrison
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Carmel Toomes
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Sandra M Bell
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
19
|
Bray LJ, Werner C. Evaluation of Three-Dimensional in Vitro Models to Study Tumor Angiogenesis. ACS Biomater Sci Eng 2017; 4:337-346. [DOI: 10.1021/acsbiomaterials.7b00139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Laura J. Bray
- Institute
of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059 Queensland Australia
- Mater
Research Institute - University of Queensland (MRI-UQ), Translational Research Institute, 37 Kent Street, Woolloongabba 4102, QLD Australia
| | - Carsten Werner
- Leibniz
Institute of Polymer Research Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, 01069 Dresden, Saxony, Germany
- Center
for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Saxony, Germany
| |
Collapse
|
20
|
Zhu X, Ding X. Study on a 3D Hydrogel-Based Culture Model for Characterizing Growth of Fibroblasts under Viral Infection and Drug Treatment. SLAS DISCOVERY 2017; 22:626-634. [PMID: 28340537 DOI: 10.1177/2472555217701247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Three-dimensional (3D) in vitro tissue models provide an approach for the systematic, repetitive, and quantitative study of drugs. In this study, we constructed an in vitro 3D acrylated hyaluronic acid (AHA) hydrogel model encapsulating fibroblasts, performed long-period 3D culture, and tested cellular topological changes and proliferation variation in the presence of herpes simplex virus-1 (HSV-1) as an infecting virus and acyclovir (ACV) as the treatment drug. The AHA hydrogels were formed by using Michael addition chemistry of bis-cysteine containing MMP-degradable cross-linker onto AHA prefunctionalized with cell adhesion peptides (RGD). Cellular structures of 3T3 fibroblasts in hydrogel presented different morphological evolution processes and proliferation rates between different groups, including HSV-1 treated alone, ACV treated alone, HSV-1 and ACV cotreated, and control samples. In AHA hydrogel, ACV blocked HSV-1 infection/replication on fibroblasts. Yet, the proliferation of ACV-treated fibroblasts was slower than that of the control group. A significantly longer period was required for cells in 3D AHA gel to regain a healthy status when compared with cells in two-dimensional (2D) culture. This hydrogel-based 3D culture model potentially lays a foundation for analyzing the response of self-organized 3D tissues to viruses and drugs in a way that is closer to nature.
Collapse
Affiliation(s)
- Xiaolu Zhu
- 1 College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu, PR China.,3 Mechanical and Aerospace Engineering Department, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xianting Ding
- 2 Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, PR China
| |
Collapse
|
21
|
Kriston-Vizi J, Flotow H. Getting the whole picture: High content screening using three-dimensional cellular model systems and whole animal assays. Cytometry A 2016; 91:152-159. [PMID: 27403779 DOI: 10.1002/cyto.a.22907] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/09/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Phenotypic or High Content Screening (HCS) is becoming more widely used for primary screening campaigns in drug discovery. Currently the vast majority of HCS campaigns are using cell lines grown in well-established monolayer cultures (2D tissue culture). There is widespread recognition that the more biologically relevant 3D tissue culture technologies such as spheroids and organoids and even whole animal assays will eventually be run as primary HCS. Upgrading the IT infrastructure to cope with the increase in data volumes requires investments in hardware (and software) and this will be manageable. However, the main bottleneck for the effective adoption and use of 3D tissue culture and whole animal assays in HCS is anticipated to be the development of software for the analysis of 3D images. In this review we summarize the current state of the available software and how they may be applied to analyzing 3D images obtained from a HCS campaign. © 2016 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Janos Kriston-Vizi
- Bioinformatics Image Core, MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Horst Flotow
- HDC GmbH, Byk Gulden Strasse 2, Konstanz, Germany
| |
Collapse
|
22
|
ZEEBERG KATRINE, CARDONE ROSAANGELA, GRECO MARIARAFFAELLA, SACCOMANO MARA, NØHR-NIELSEN ASBJØRN, ALVES FRAUKE, PEDERSEN STINEFALSIG, RESHKIN STEPHANJOEL. Assessment of different 3D culture systems to study tumor phenotype and chemosensitivity in pancreatic ductal adenocarcinoma. Int J Oncol 2016; 49:243-52. [DOI: 10.3892/ijo.2016.3513] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/13/2016] [Indexed: 11/05/2022] Open
|
23
|
Bioengineered silk scaffolds in 3D tissue modeling with focus on mammary tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 59:1168-1180. [DOI: 10.1016/j.msec.2015.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/04/2015] [Accepted: 10/02/2015] [Indexed: 02/07/2023]
|
24
|
Gaiko-Shcherbak A, Fabris G, Dreissen G, Merkel R, Hoffmann B, Noetzel E. The Acinar Cage: Basement Membranes Determine Molecule Exchange and Mechanical Stability of Human Breast Cell Acini. PLoS One 2015; 10:e0145174. [PMID: 26674091 PMCID: PMC4684506 DOI: 10.1371/journal.pone.0145174] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/30/2015] [Indexed: 11/19/2022] Open
Abstract
The biophysical properties of the basement membrane that surrounds human breast glands are poorly understood, but are thought to be decisive for normal organ function and malignancy. Here, we characterize the breast gland basement membrane with a focus on molecule permeation and mechanical stability, both crucial for organ function. We used well-established and nature-mimicking MCF10A acini as 3D cell model for human breast glands, with ether low- or highly-developed basement membrane scaffolds. Semi-quantitative dextran tracer (3 to 40 kDa) experiments allowed us to investigate the basement membrane scaffold as a molecule diffusion barrier in human breast acini in vitro. We demonstrated that molecule permeation correlated positively with macromolecule size and intriguingly also with basement membrane development state, revealing a pore size of at least 9 nm. Notably, an intact collagen IV mesh proved to be essential for this permeation function. Furthermore, we performed ultra-sensitive atomic force microscopy to quantify the response of native breast acini and of decellularized basement membrane shells against mechanical indentation. We found a clear correlation between increasing acinar force resistance and basement membrane formation stage. Most important native acini with highly-developed basement membranes as well as cell-free basement membrane shells could both withstand physiologically relevant loads (≤ 20 nN) without loss of structural integrity. In contrast, low-developed basement membranes were significantly softer and more fragile. In conclusion, our study emphasizes the key role of the basement membrane as conductor of acinar molecule influx and mechanical stability of human breast glands, which are fundamental for normal organ function.
Collapse
Affiliation(s)
- Aljona Gaiko-Shcherbak
- Institute of Complex Systems ICS7: Biomechanics, Forschungszentrum Jülich, Jülich, Germany
| | - Gloria Fabris
- Institute of Complex Systems ICS7: Biomechanics, Forschungszentrum Jülich, Jülich, Germany
| | - Georg Dreissen
- Institute of Complex Systems ICS7: Biomechanics, Forschungszentrum Jülich, Jülich, Germany
| | - Rudolf Merkel
- Institute of Complex Systems ICS7: Biomechanics, Forschungszentrum Jülich, Jülich, Germany
| | - Bernd Hoffmann
- Institute of Complex Systems ICS7: Biomechanics, Forschungszentrum Jülich, Jülich, Germany
| | - Erik Noetzel
- Institute of Complex Systems ICS7: Biomechanics, Forschungszentrum Jülich, Jülich, Germany
- * E-mail:
| |
Collapse
|
25
|
Booth ME, Nash CE, Roberts NP, Magee DR, Treanor D, Hanby AM, Speirs V. 3-D Tissue Modelling and Virtual Pathology as New Approaches to Study Ductal Carcinoma In Situ. Altern Lab Anim 2015; 43:377-83. [DOI: 10.1177/026119291504300605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Widespread screening mammography programmes mean that ductal carcinoma in situ (DCIS), a pre-invasive breast lesion, is now more frequently diagnosed. However, not all diagnosed DCIS lesions progress to invasive breast cancer, which presents a dilemma for clinicians. As such, there is much interest in studying DCIS in the laboratory, in order to help understand more about its biology and determine the characteristics of those that progress to invasion. Greater knowledge would lead to targeted and better DCIS treatment. Here, we outline some of the models available to study DCIS, with a particular focus on animal-free systems.
Collapse
Affiliation(s)
- Mary E. Booth
- Leeds Institute of Cancer and Pathology, Leeds, UK
- Joint first authors
| | - Claire E. Nash
- Leeds Institute of Cancer and Pathology, Leeds, UK
- Joint first authors
- Current address: The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | | | | | - Darren Treanor
- Leeds Institute of Cancer and Pathology, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | |
Collapse
|
26
|
Helicobacter pylori CagA Suppresses Apoptosis through Activation of AKT in a Nontransformed Epithelial Cell Model of Glandular Acini Formation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:761501. [PMID: 26557697 PMCID: PMC4628739 DOI: 10.1155/2015/761501] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 02/07/2023]
Abstract
H. pylori infection is the most important environmental risk to develop gastric cancer, mainly through its virulence factor CagA. In vitro models of CagA function have demonstrated a phosphoprotein activity targeting multiple cellular signaling pathways, while cagA transgenic mice develop carcinomas of the gastrointestinal tract, supporting oncogenic functions. However, it is still not completely clear how CagA alters cellular processes associated with carcinogenic events. In this study, we evaluated the capacity of H. pylori CagA positive and negative strains to alter nontransformed MCF-10A glandular acini formation. We found that CagA positive strains inhibited lumen formation arguing for an evasion of apoptosis activity of central acini cells. In agreement, CagA positive strains induced a cell survival activity that correlated with phosphorylation of AKT and of proapoptotic proteins BIM and BAD. Anoikis is a specific type of apoptosis characterized by AKT and BIM activation and it is the mechanism responsible for lumen formation of MCF-10A acini in vitro and mammary glands in vivo. Anoikis resistance is also a common mechanism of invading tumor cells. Our data support that CagA positive strains signaling function targets the AKT and BIM signaling pathway and this could contribute to its oncogenic activity through anoikis evasion.
Collapse
|
27
|
Kurup A, Ravindranath S, Tran T, Keating M, Gascard P, Valdevit L, Tlsty TD, Botvinick EL. Novel insights from 3D models: the pivotal role of physical symmetry in epithelial organization. Sci Rep 2015; 5:15153. [PMID: 26472542 PMCID: PMC4608012 DOI: 10.1038/srep15153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 09/15/2015] [Indexed: 12/19/2022] Open
Abstract
3D tissue culture models are utilized to study breast cancer and other pathologies because they better capture the complexity of in vivo tissue architecture compared to 2D models. However, to mimic the in vivo environment, the mechanics and geometry of the ECM must also be considered. Here, we studied the mechanical environment created in two 3D models, the overlay protocol (OP) and embedded protocol (EP). Mammary epithelial acini features were compared using OP or EP under conditions known to alter acinus organization, i.e. collagen crosslinking and/or ErbB2 receptor activation. Finite element analysis and active microrheology demonstrated that OP creates a physically asymmetric environment with non-uniform mechanical stresses in radial and circumferential directions. Further contrasting with EP, acini in OP displayed cooperation between ErbB2 signalling and matrix crosslinking. These differences in acini phenotype observed between OP and EP highlight the functional impact of physical symmetry in 3D tissue culture models.
Collapse
Affiliation(s)
- Abhishek Kurup
- University of California Irvine, Department of Biomedical Engineering, Irvine, USA
| | - Shreyas Ravindranath
- University of California Irvine, Department of Biomedical Engineering, Irvine, USA
| | - Tim Tran
- University of California Irvine, Department of Biomedical Engineering, Irvine, USA
| | - Mark Keating
- University of California Irvine, Department of Biomedical Engineering, Irvine, USA
| | - Philippe Gascard
- University of California San Francisco, Department of Pathology, San Francisco, USA
| | - Lorenzo Valdevit
- University of California Irvine, Department of Mechanical and Aerospace Engineering, Irvine, USA
| | - Thea D Tlsty
- University of California San Francisco, Department of Pathology, San Francisco, USA
| | - Elliot L Botvinick
- University of California Irvine, Department of Biomedical Engineering, Irvine, USA.,University of California Irvine, Department of Surgery, Irvine, USA
| |
Collapse
|
28
|
Yu X, Li Z. Long non-coding RNA HOTAIR: A novel oncogene (Review). Mol Med Rep 2015; 12:5611-8. [PMID: 26238267 DOI: 10.3892/mmr.2015.4161] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 04/20/2015] [Indexed: 11/05/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been found to be pervasively transcribed in the genome and are critical regulators of the epigenome. Increasing evidence suggests that lncRNAs are aberrantly expressed in several types of human cancer and that they are important in the initiation, development and metastasis of human cancer. Previous studies have revealed that HOX transcript antisense intergenic RNA (HOTAIR) was frequently upregulated in various types of cancer, including breast cancer, esophageal cancer, lung cancer and gastric cancer. In addition, patients with high expression levels of HOTAIR have a significantly poorer prognosis, compared with those with low levels of expression. HOTAIR is involved in the control of cell apoptosis, growth, metastasis, angiogenesis, DNA repair and tumor cells metabolism. The present review provides an overview of the current knowledge concerning the role of HOTAIR in tumor development and progression.
Collapse
Affiliation(s)
- Xin Yu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, P.R. China
| | - Zheng Li
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
29
|
Dolega ME, Abeille F, Picollet-D'hahan N, Gidrol X. Controlled 3D culture in Matrigel microbeads to analyze clonal acinar development. Biomaterials 2015; 52:347-57. [DOI: 10.1016/j.biomaterials.2015.02.042] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/31/2015] [Accepted: 02/06/2015] [Indexed: 01/01/2023]
|
30
|
Eukaryotic Translation Initiation Factor 4E Is a Feed-Forward Translational Coactivator of Transforming Growth Factor β Early Protransforming Events in Breast Epithelial Cells. Mol Cell Biol 2015; 35:2597-609. [PMID: 25986608 DOI: 10.1128/mcb.00324-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/12/2015] [Indexed: 12/18/2022] Open
Abstract
Eukaryotic translation initiation factor 4E (eIF4E) is overexpressed early in breast cancers in association with disease progression and reduced survival. Much remains to be understood regarding the role of eIF4E in human cancer. We determined, using immortalized human breast epithelial cells, that elevated expression of eIF4E translationally activates the transforming growth factor β (TGF-β) pathway, promoting cell invasion, a loss of cell polarity, increased cell survival, and other hallmarks of early neoplasia. Overexpression of eIF4E is shown to facilitate the selective translation of integrin β1 mRNA, which drives the translationally controlled assembly of a TGF-β receptor signaling complex containing α3β1 integrins, β-catenin, TGF-β receptor I, E-cadherin, and phosphorylated Smad2/3. This receptor complex acutely sensitizes nonmalignant breast epithelial cells to activation by typically substimulatory levels of activated TGF-β. TGF-β can promote cellular differentiation or invasion and transformation. As a translational coactivator of TGF-β, eIF4E confers selective mRNA translation, reprogramming nonmalignant cells to an invasive phenotype by reducing the set point for stimulation by activated TGF-β. Overexpression of eIF4E may be a proinvasive facilitator of TGF-β activity.
Collapse
|
31
|
Kim BJ, Zhao S, Bunaciu RP, Yen A, Wu M. A 3D in situ cell counter reveals that breast tumor cell (MDA-MB-231) proliferation rate is reduced by the collagen matrix density. Biotechnol Prog 2015; 31:990-996. [PMID: 25683564 DOI: 10.1002/btpr.2062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/15/2015] [Indexed: 11/08/2022]
Abstract
Many cell types require the biophysical and biochemical cues within the 3D extracellular matrix (ECM) to exhibit their true physiologically relevant behavior. As a result, cell culture platforms have been evolving from traditional 2D petridish plates into 3D biomatrices, and there is a need for developing analytic tools to characterize 3D cell culture. The existing cell counting method, using a hemocytometer or coulter counter, requires that cells are suspended in fluids prior to counting. This poses a challenge for 3D cell culture as cells are embedded in a 3D biomatrix. We use a facile 3D cell counting method that overcomes this limitation and allows for in situ cell counting in a 3D cell culture using equipment that is commonly available in a biology lab. Using a breast tumor cell line, MDA-MB-231, as a model system, we demonstrated that MDA-MB-231 cells (1) grow slower within a 3D collagen matrix than on a 2D substrate for an extended growth time (a week) with a comparable, initial cell-to-cell distance, (2) their cell growth rate decreases with the increase of collagen concentration, showing a linear growth rate rather than an exponential growth rate. Further work using flow cytometry showed that the observed growth rate reduction was consistent with the retardation of the transition to S (synthesis) phase in the cell cycle. This work demonstrates the validity of the 3D cell counting method and the importance of cell-ECM interactions in cell proliferation.
Collapse
Affiliation(s)
- Beum Jun Kim
- Dept. of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853
| | - Shuting Zhao
- Dept. of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853
| | - Rodica P Bunaciu
- Dept. of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Andrew Yen
- Dept. of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Mingming Wu
- Dept. of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853
| |
Collapse
|
32
|
Tissue-engineered 3D tumor angiogenesis models: potential technologies for anti-cancer drug discovery. Adv Drug Deliv Rev 2014; 79-80:30-9. [PMID: 24819220 DOI: 10.1016/j.addr.2014.05.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/14/2014] [Accepted: 05/02/2014] [Indexed: 01/06/2023]
Abstract
Angiogenesis is indispensable for solid tumor expansion, and thus it has become a major target of cancer research and anti-cancer therapies. Deciphering the arcane actions of various cell populations during tumor angiogenesis requires sophisticated research models, which could capture the dynamics and complexity of the process. There is a continuous need for improvement of existing research models, which engages interdisciplinary approaches of tissue engineering with life sciences. Tireless efforts to develop a new model to study tumor angiogenesis result in innovative solutions, which bring us one step closer to decipher the dubious nature of cancer. This review aims to overview the recent developments, current limitations and future challenges in three-dimensional tissue-engineered models for the study of tumor angiogenesis and for the purpose of elucidating novel targets aimed at anti-cancer drug discovery.
Collapse
|
33
|
Chambers KF, Mosaad EMO, Russell PJ, Clements JA, Doran MR. 3D Cultures of prostate cancer cells cultured in a novel high-throughput culture platform are more resistant to chemotherapeutics compared to cells cultured in monolayer. PLoS One 2014; 9:e111029. [PMID: 25380249 PMCID: PMC4224379 DOI: 10.1371/journal.pone.0111029] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/26/2014] [Indexed: 11/25/2022] Open
Abstract
Despite monolayer cultures being widely used for cancer drug development and testing, 2D cultures tend to be hypersensitive to chemotherapy and are relatively poor predictors of whether a drug will provide clinical benefit. Whilst generally more complicated, three dimensional (3D) culture systems often better recapitulate true cancer architecture and provide a more accurate drug response. As a step towards making 3D cancer cultures more accessible, we have developed a microwell platform and surface modification protocol to enable high throughput manufacture of 3D cancer aggregates. Herein we use this novel system to characterize prostate cancer cell microaggregates, including growth kinetics and drug sensitivity. Our results indicate that prostate cancer cells are viable in this system, however some non-cancerous prostate cell lines are not. This system allows us to consistently control for the presence or absence of an apoptotic core in the 3D cancer microaggregates. Similar to tumor tissues, the 3D microaggregates display poor polarity. Critically the response of 3D microaggregates to the chemotherapeutic drug, docetaxel, is more consistent with in vivo results than the equivalent 2D controls. Cumulatively, our results demonstrate that these prostate cancer microaggregates better recapitulate the morphology of prostate tumors compared to 2D and can be used for high-throughput drug testing.
Collapse
Affiliation(s)
- Karen F. Chambers
- Stem Cell Therapies Laboratory, Queensland University of Technology at the Translational Research Institute, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre, Translational Research Institute, Brisbane, Queensland, Australia
| | - Eman M. O. Mosaad
- Stem Cell Therapies Laboratory, Queensland University of Technology at the Translational Research Institute, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre, Translational Research Institute, Brisbane, Queensland, Australia
| | - Pamela J. Russell
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre, Translational Research Institute, Brisbane, Queensland, Australia
| | - Judith A. Clements
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre, Translational Research Institute, Brisbane, Queensland, Australia
| | - Michael R. Doran
- Stem Cell Therapies Laboratory, Queensland University of Technology at the Translational Research Institute, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre, Translational Research Institute, Brisbane, Queensland, Australia
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
34
|
Casbas-Hernandez P, D'Arcy M, Roman-Perez E, Brauer HA, McNaughton K, Miller SM, Chhetri RK, Oldenburg AL, Fleming JM, Amos KD, Makowski L, Troester MA. Role of HGF in epithelial-stromal cell interactions during progression from benign breast disease to ductal carcinoma in situ. Breast Cancer Res 2014; 15:R82. [PMID: 24025166 PMCID: PMC3978616 DOI: 10.1186/bcr3476] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 07/16/2013] [Indexed: 12/17/2022] Open
Abstract
Introduction Basal-like and luminal breast cancers have distinct stromal–epithelial interactions, which play a role in progression to invasive cancer. However, little is known about how stromal–epithelial interactions evolve in benign and pre-invasive lesions. Methods To study epithelial–stromal interactions in basal-like breast cancer progression, we cocultured reduction mammoplasty fibroblasts with the isogenic MCF10 series of cell lines (representing benign/normal, atypical hyperplasia, and ductal carcinoma in situ). We used gene expression microarrays to identify pathways induced by coculture in premalignant cells (MCF10DCIS) compared with normal and benign cells (MCF10A and MCF10AT1). Relevant pathways were then evaluated in vivo for associations with basal-like subtype and were targeted in vitro to evaluate effects on morphogenesis. Results Our results show that premalignant MCF10DCIS cells express characteristic gene expression patterns of invasive basal-like microenvironments. Furthermore, while hepatocyte growth factor (HGF) secretion is upregulated (relative to normal, MCF10A levels) when fibroblasts are cocultured with either atypical (MCF10AT1) or premalignant (MCF10DCIS) cells, only MCF10DCIS cells upregulated the HGF receptor MET. In three-dimensional cultures, upregulation of HGF/MET in MCF10DCIS cells induced morphological changes suggestive of invasive potential, and these changes were reversed by antibody-based blocking of HGF signaling. These results are relevant to in vivo progression because high expression of a novel MCF10DCIS-derived HGF signature was correlated with the basal-like subtype, with approximately 86% of basal-like cancers highly expressing the HGF signature, and because high expression of HGF signature was associated with poor survival. Conclusions Coordinated and complementary changes in HGF/MET expression occur in epithelium and stroma during progression of pre-invasive basal-like lesions. These results suggest that targeting stroma-derived HGF signaling in early carcinogenesis may block progression of basal-like precursor lesions.
Collapse
|
35
|
MacMillan CD, Leong HS, Dales DW, Robertson AE, Lewis JD, Chambers AF, Tuck AB. Stage of breast cancer progression influences cellular response to activation of the WNT/planar cell polarity pathway. Sci Rep 2014; 4:6315. [PMID: 25204426 PMCID: PMC4159636 DOI: 10.1038/srep06315] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/11/2014] [Indexed: 12/20/2022] Open
Abstract
Planar cell polarity (PCP) signaling has been shown in different studies to either promote or inhibit the malignancy of breast cancer. Using the 21T cell lines, which were derived from an individual patient and represent distinct stages of progression, we show that the prototypical PCP ligand, WNT5A, is expressed highest in 21MT-1 cells (invasive mammary carcinoma) and lowest in 21PT (atypical ductal hyperplasia) and 21NT (ductal carcinoma in situ) cells. Overexpression of WNT5A decreased spherical colony formation and increased invasion and in vivo extravasation only in 21NT cells; whereas overexpression increased migration of both 21PT and 21NT cells. WNT5A overexpression also increased RHOA expression of both cell lines and subsequent RHOA knockdown blocked WNT5A-induced migration, but only partially blocked WNT5A-induced invasion of 21NT cells. PCP can signal through VANGL1 to modulate AP-1 target genes (e.g. MMP3) and induce invasion. VANGL1 knockdown inhibited WNT5A-induced invasion of 21NT cells, but had no effect on WNT5A-induced migration of either 21PT or 21NT cells. WNT5A-induced MMP3 expression was seen only in 21NT cells, an effect that was VANGL1 dependent, but independent of AP-1. We thus provide evidence that PCP signaling can act in a context dependent manner to promote breast cancer progression.
Collapse
Affiliation(s)
- Connor D MacMillan
- 1] Department of Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [2] London Regional Cancer Program, London Health Sciences Centre, London, ON [3] Department of Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - Hon S Leong
- 1] London Regional Cancer Program, London Health Sciences Centre, London, ON [2] Department of Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - David W Dales
- London Regional Cancer Program, London Health Sciences Centre, London, ON
| | - Amy E Robertson
- 1] London Regional Cancer Program, London Health Sciences Centre, London, ON [2] Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - John D Lewis
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB
| | - Ann F Chambers
- 1] Department of Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [2] London Regional Cancer Program, London Health Sciences Centre, London, ON [3] Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [4] Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| | - Alan B Tuck
- 1] Department of Pathology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON [2] London Regional Cancer Program, London Health Sciences Centre, London, ON [3] Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON
| |
Collapse
|
36
|
Dolega ME, Wagh J, Gerbaud S, Kermarrec F, Alcaraz JP, Martin DK, Gidrol X, Picollet-D’hahan N. Facile bench-top fabrication of enclosed circular microchannels provides 3D confined structure for growth of prostate epithelial cells. PLoS One 2014; 9:e99416. [PMID: 24945245 PMCID: PMC4063722 DOI: 10.1371/journal.pone.0099416] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/14/2014] [Indexed: 12/15/2022] Open
Abstract
We present a simple bench-top method to fabricate enclosed circular channels for biological experiments. Fabricating the channels takes less than 2 hours by using glass capillaries of various diameters (from 100 µm up to 400 µm) as a mould in PDMS. The inner surface of microchannels prepared in this way was coated with a thin membrane of either Matrigel or a layer-by-layer polyelectrolyte to control cellular adhesion. The microchannels were then used as scaffolds for 3D-confined epithelial cell culture. To show that our device can be used with several epithelial cell types from exocrine glandular tissues, we performed our biological studies on adherent epithelial prostate cells (non-malignant RWPE-1 and invasive PC3) and also on breast (non-malignant MCF10A) cells We observed that in static conditions cells adhere and proliferate to form a confluent layer in channels of 150 µm in diameter and larger, whereas cellular viability decreases with decreasing diameter of the channel. Matrigel and PSS (poly (sodium 4-styrenesulphonate)) promote cell adhesion, whereas the cell proliferation rate was reduced on the PAH (poly (allylamine hydrochloride))-terminated surface. Moreover infusing channels with a continuous flow did not induce any cellular detachment. Our system is designed to simply grow cells in a microchannel structure and could be easily fabricated in any biological laboratory. It offers opportunities to grow epithelial cells that support the formation of a light. This system could be eventually used, for example, to collect cellular secretions, or study cell responses to graduated hypoxia conditions, to chemicals (drugs, siRNA, …) and/or physiological shear stress.
Collapse
Affiliation(s)
- Monika E. Dolega
- Univ. Grenoble Alpes, iRTSV-BGE, Grenoble, France
- CEA, iRTSV-BGE, Grenoble, France
- INSERM, BGE, Grenoble, France
| | - Jayesh Wagh
- Univ. Grenoble Alpes, iRTSV-BGE, Grenoble, France
- CEA, iRTSV-BGE, Grenoble, France
- INSERM, BGE, Grenoble, France
| | - Sophie Gerbaud
- Univ. Grenoble Alpes, iRTSV-BGE, Grenoble, France
- CEA, iRTSV-BGE, Grenoble, France
- INSERM, BGE, Grenoble, France
| | - Frederique Kermarrec
- Univ. Grenoble Alpes, iRTSV-BGE, Grenoble, France
- CEA, iRTSV-BGE, Grenoble, France
- INSERM, BGE, Grenoble, France
| | | | - Donald K. Martin
- UJF-Grenoble 1, CNRS, TIMC-IMAG UMR 5525 (SyNaBi), Grenoble, France
| | - Xavier Gidrol
- Univ. Grenoble Alpes, iRTSV-BGE, Grenoble, France
- CEA, iRTSV-BGE, Grenoble, France
- INSERM, BGE, Grenoble, France
| | - Nathalie Picollet-D’hahan
- Univ. Grenoble Alpes, iRTSV-BGE, Grenoble, France
- CEA, iRTSV-BGE, Grenoble, France
- INSERM, BGE, Grenoble, France
- * E-mail:
| |
Collapse
|
37
|
Hernandez-Gordillo V, Chmielewski J. Mimicking the extracellular matrix with functionalized, metal-assembled collagen peptide scaffolds. Biomaterials 2014; 35:7363-73. [PMID: 24933513 DOI: 10.1016/j.biomaterials.2014.05.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/08/2014] [Indexed: 12/27/2022]
Abstract
Natural and synthetic three-dimensional (3-D) scaffolds that mimic the microenvironment of the extracellular matrix (ECM), with growth factor storage/release and the display of cell adhesion signals, offer numerous advantages for regenerative medicine and in vitro morphogenesis and oncogenesis modeling. Here we report the design of collagen mimetic peptides (CMPs) that assemble into a highly crosslinked 3-D matrix in response to metal ion stimuli, that may be functionalized with His-tagged cargoes, such as green fluorescent protein (GFP-His8) and human epidermal growth factor (hEGF-His6). The bound hEGF-His6 was found to gradually release from the matrix in vitro and induce cell proliferation in the EGF-dependent cell line MCF10A. The additional incorporation of a cell adhesion sequence (RGDS) at the N-terminus of the CMP creates an environment that facilitated the organization of matrix-encapsulated MCF10A cells into spheroid structures, thus mimicking the ECM environment.
Collapse
Affiliation(s)
| | - Jean Chmielewski
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
38
|
Wittig R, Rosenholm JM, von Haartman E, Hemming J, Genze F, Bergman L, Simmet T, Lindén M, Sahlgren C. Active targeting of mesoporous silica drug carriers enhances γ-secretase inhibitor efficacy in an in vivo model for breast cancer. Nanomedicine (Lond) 2014; 9:971-87. [DOI: 10.2217/nnm.13.62] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: In this article, we use an alternative cancer model for the evaluation of nanotherapy, and assess the impact of surface functionalization and active targeting of mesoporous silica nanoparticles (MSNPs) on therapeutic efficacy in vivo. Materials & methods: We used the chorioallantoic membrane xenograft assay to investigate the biodistribution and therapeutic efficacy of folate versus polyethyleneimine-functionalized γ-secretase inhibitor-loaded MSNPs in breast and prostate tumor models. Results: γ-secretase inhibitor-loaded MSNPs inhibited tumor growth in breast and prostate cancer xenografts. Folate conjugation improved the therapeutic outcome in folic acid receptor-positive breast cancer, but not in prostate cancer lacking the receptor. Conclusion: The results demonstrate that therapeutic efficacy is linked to cellular uptake of MSNPs as opposed to tumor accumulation, and show that MSNP-based delivery of γ-secretase inhibitors is therapeutically effective in both breast and prostate cancer. In this article, we present a model system for a medium-to-high throughput, cost-effective, quantitative evaluation of nanoparticulate drug carriers. Original submitted 12 November 2012; Revised submitted 8 February 2013
Collapse
Affiliation(s)
- Rainer Wittig
- Institute for Laser Technologies in Medicine & Metrology at Ulm University, Helmholtzstrasse 12, D-89081 Ulm, Germany
| | - Jessica M Rosenholm
- Center for Functional Materials, Laboratory for Physical Chemistry, Department of Natural Sciences, Åbo Akademi University, FI-20500 Turku, Finland
| | - Eva von Haartman
- Center for Functional Materials, Laboratory for Physical Chemistry, Department of Natural Sciences, Åbo Akademi University, FI-20500 Turku, Finland
| | - Jarl Hemming
- Wood & Paper Chemistry, Department of Chemical Engineering, Åbo Akademi University, FI-20500 Turku, Finland
| | - Felicitas Genze
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Helmholtzstrasse 20, D-89081 Ulm, Germany
| | - Lotta Bergman
- Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Helmholtzstrasse 20, D-89081 Ulm, Germany
| | - Mika Lindén
- Inorganic Chemistry II, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | - Cecilia Sahlgren
- Department of Biomedical Engineering, Technical University of Eindhoven, 2612 Eindhoven, The Netherlands
| |
Collapse
|
39
|
Piccinini AM, Midwood KS. Illustrating the interplay between the extracellular matrix and microRNAs. Int J Exp Pathol 2014; 95:158-80. [PMID: 24761792 DOI: 10.1111/iep.12079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/18/2014] [Indexed: 12/19/2022] Open
Abstract
The discovery of cell surface receptors that bind to extracellular matrix (ECM) components marked a new era in biological research. Since then there has been an increasing appreciation of the importance of studying cells in the context of their extracellular environment. Cell behaviour is profoundly affected by the ECM, whose synthesis and turnover must be finely balanced in order to maintain normal function and prevent disease. In the last decade, microRNAs (miRNAs) have emerged as key regulators of ECM gene expression. As new technologies for the identification and validation of miRNA targets continue to be developed, a growing body of data supporting the role of miRNAs in regulating the ECM biology has arisen from a variety of cell and animal models along with clinical studies. However, more recent findings suggest an intriguing interplay between the ECM and miRNAs: not only can miRNAs control the composition of the ECM, but also the ECM can affect the expression of specific miRNAs. Here we discuss how miRNAs contribute to the synthesis, maintenance and remodelling of the ECM during development and disease. Furthermore, we bring to light evidence that points to a role for the ECM in regulating miRNA expression and function.
Collapse
Affiliation(s)
- Anna M Piccinini
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, UK
| | | |
Collapse
|
40
|
Arisi MF, Starker RA, Addya S, Huang Y, Fernandez SV. All trans-retinoic acid (ATRA) induces re-differentiation of early transformed breast epithelial cells. Int J Oncol 2014; 44:1831-42. [PMID: 24676586 PMCID: PMC4063534 DOI: 10.3892/ijo.2014.2354] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/03/2014] [Indexed: 02/03/2023] Open
Abstract
Retinoids have been used as potential chemotherapeutic or chemopreventive agents because of their differentiative, anti-proliferative, pro-apoptotic and antioxidant properties. We investigated the effect of all trans-retinoic acid (ATRA) at different stages of the neoplastic transformation using an in vitro model of breast cancer progression. This model was previously developed by treating the MCF-10F human normal breast epithelial cells with high dose of estradiol and consists of four cell lines which show a progressive neoplastic transformation: MCF-10F, normal stage; trMCF, transformed MCF-10F; bsMCF, invasive stage; and caMCF, tumorigenic stage. In 3D cultures, MCF-10F cells form tubules resembling the structures in the normal mammary gland. After treatment with estradiol, these cells formed tubules and spherical masses which are indicative of transformation. Cells that only formed spherical masses in collagen were isolated (trMCF clone 11) and treated with ATRA. After treatment with 10 or 1 µM ATRA, the trMCF clone 11 cells showed tubules in collagen; 10 and 43% of the structures were tubules in cells treated with 10 and 1 µM ATRA, respectively. Gene expression studies showed that 207 genes upregulated in transformed trMCF clone 11 cells were downregulated after 1 µM ATRA treatment to levels comparable to those found in the normal breast epithelial cells MCF-10F. Furthermore, 236 genes that were downregulated in trMCF clone 11 were upregulated after 1 µM ATRA treatment to similar levels shown in normal epithelial cells. These 443 genes defined a signature of the ATRA re-programming effect. Our results showed that 1 µM ATRA was able to re-differentiate transformed cells at early stages of the neoplastic process and antagonistically regulate breast cancer associated genes. The invasive and tumorigenic cells did not show any changes in morphology after ATRA treatment. These results suggest that ATRA could be used as a chemopreventive agent to inhibit the progression of premalignant lesions of the breast.
Collapse
Affiliation(s)
- Maria F Arisi
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Rebecca A Starker
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sankar Addya
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yong Huang
- Section of Gastroenterology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Sandra V Fernandez
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
41
|
Miz1 deficiency in the mammary gland causes a lactation defect by attenuated Stat5 expression and phosphorylation. PLoS One 2014; 9:e89187. [PMID: 24586582 PMCID: PMC3929623 DOI: 10.1371/journal.pone.0089187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/15/2014] [Indexed: 01/25/2023] Open
Abstract
Miz1 is a zinc finger transcription factor with an N-terminal POZ domain. Complexes with Myc, Bcl-6 or Gfi-1 repress expression of genes like Cdkn2b (p15Ink4) or Cdkn1a (p21Cip1). The role of Miz1 in normal mammary gland development has not been addressed so far. Conditional knockout of the Miz1 POZ domain in luminal cells during pregnancy caused a lactation defect with a transient reduction of glandular tissue, reduced proliferation and attenuated differentiation. This was recapitulated in vitro using mouse mammary gland derived HC11 cells. Further analysis revealed decreased Stat5 activity in Miz1ΔPOZ mammary glands and an attenuated expression of Stat5 targets. Gene expression of the Prolactin receptor (PrlR) and ErbB4, both critical for Stat5 phosphorylation (pStat5) or pStat5 nuclear translocation, was decreased in Miz1ΔPOZ females. Microarray, ChIP-Seq and gene set enrichment analysis revealed a down-regulation of Miz1 target genes being involved in vesicular transport processes. Our data suggest that deranged intracellular transport and localization of PrlR and ErbB4 disrupt the Stat5 signalling pathway in mutant glands and cause the observed lactation phenotype.
Collapse
|
42
|
Wu M, Swartz MA. Modeling tumor microenvironments in vitro. J Biomech Eng 2014; 136:021011. [PMID: 24402507 PMCID: PMC4023667 DOI: 10.1115/1.4026447] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/28/2013] [Accepted: 01/09/2014] [Indexed: 12/31/2022]
Abstract
Tumor progression depends critically upon the interactions between the tumor cells and their microenvironment. The tumor microenvironment is heterogeneous and dynamic; it consists of extracellular matrix, stromal cells, immune cells, progenitor cells, and blood and lymphatic vessels. The emerging fields of tissue engineering and microtechnologies have opened up new possibilities for engineering physiologically relevant and spatially well-defined microenvironments. These in vitro models allow specific manipulation of biophysical and biochemical parameters, such as chemical gradients, biomatrix stiffness, metabolic stress, and fluid flows; thus providing a means to study their roles in certain aspects of tumor progression such as cell proliferation, invasion, and crosstalk with other cell types. Challenges and perspectives for deconvolving the complexity of tumor microenvironments will be discussed. Emphasis will be given to in vitro models of tumor cell migration and invasion.
Collapse
|
43
|
Xu G, Yin F, Wu H, Hu X, Zheng L, Zhao J. In vitro ovarian cancer model based on three-dimensional agarose hydrogel. J Tissue Eng 2014; 5:2041731413520438. [PMID: 24551446 PMCID: PMC3924902 DOI: 10.1177/2041731413520438] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/16/2013] [Indexed: 12/17/2022] Open
Abstract
To establish a typical tumor model of ovarian cancer which may be more representative and reliable than traditional monolayer culture and pellet, agarose was used as cell vehicle to engineering tumor. Selection of agarose is based on its successful application in tissue engineering with both amenable mechanical and biological properties. In this study, ovarian cancer cell line SKOV3 was encapsulated in agarose hydrogel with cell aggregates and two-dimensional culture as controls. In vitro cell proliferation was assessed by MTT and cell viability was examined at time points of 2, 4, and 6 days. The expression of tumor malignancy markers including matrix metalloproteinase 2, matrix metalloproteinase 9, hypoxia-inducible factor-1α, and vascular endothelial growth factor–A was assessed by real-time polymerase chain reaction. The results showed that cells proliferated more rapidly in three-dimensional agarose culture than controls. Furthermore, upregulation of matrix metalloproteinase 9 and matrix metalloproteinase 2 activity and increased expression of vascular endothelial growth factor–A and hypoxia-inducible factor-1α were shown in agarose-engineered tumors. All the evidences demonstrated that agarose may provide a more favorable environment for cancer cell growth, mimicking the in vivo environment for tumor generation. The novel in vitro tumor model may be useful for the further investigation of anticancer therapeutics.
Collapse
Affiliation(s)
- Guojie Xu
- Osteopathy Ward, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China ; Research Center for Regenerative Medicine, Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Fuqiang Yin
- The Medical and Scientific Research Center, Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Huayu Wu
- Department of Cell Biology & Genetics, School of Premedical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Xuefeng Hu
- Osteopathy Ward, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Li Zheng
- Research Center for Regenerative Medicine, Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China ; The Medical and Scientific Research Center, Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinming Zhao
- Osteopathy Ward, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China ; Research Center for Regenerative Medicine, Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
44
|
Abstract
Morphogenesis is the remarkable process by which cells self-assemble into complex tissues and organs that exhibit specialized form and function during embryological development. Many of the genes and chemical cues that mediate tissue and organ formation have been identified; however, these signals alone are not sufficient to explain how tissues and organs are constructed that exhibit their unique material properties and three-dimensional forms. Here, we review work that has revealed the central role that physical forces and extracellular matrix mechanics play in the control of cell fate switching, pattern formation, and tissue development in the embryo and how these same mechanical signals contribute to tissue homeostasis and developmental control throughout adult life.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115;
| | | | | |
Collapse
|
45
|
Label-free analysis of prostate acini-like 3D structures by lensfree imaging. Biosens Bioelectron 2013; 49:176-83. [DOI: 10.1016/j.bios.2013.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 11/22/2022]
|
46
|
Witkiewicz AK, Cox DW, Rivadeneira D, Ertel AE, Fortina P, Schwartz GF, Knudsen ES. The retinoblastoma tumor suppressor pathway modulates the invasiveness of ErbB2-positive breast cancer. Oncogene 2013; 33:3980-91. [PMID: 24121271 PMCID: PMC4150690 DOI: 10.1038/onc.2013.367] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/18/2013] [Accepted: 06/21/2013] [Indexed: 12/18/2022]
Abstract
The processes that control the progression of ductal carcinoma in situ (DCIS) to invasive breast cancer remain poorly understood. Epidermal growth factor receptor 2 (ErbB2) over expression is common in DCIS, as is disruption of the retinoblastoma tumor suppressor (RB) pathway. Here we examined the cooperative impact of ErbB2 and RB deregulation on facets of disease progression. Our studies demonstrate that RB deficiency altered the expression of key molecules needed for proper cellular organization and epithelial cell-cell adhesion as part of a program related to the epithelial to mesenchymal transition (EMT). An increase in the invasive potential of ErbB2 over expressing cells was observed upon RB depletion. Furthermore, stable knockdown of RB resulted in invasive lesions in orthotopic xenograft assays, compared to DCIS-like lesions developing from RB-proficient cells. Conversely, the invasive phenotype observed in ErbB2-positive cancer models was inhibited through CDK4/6 inhibition in an RB-dependent manner. Lastly, in a cohort of DCIS cases, we show that while elevated levels of ErbB2 are associated with increased risk of a subsequent DCIS recurrence, it is not associated with progression to invasive disease. In contrast, RB loss in ErbB2 positive DCIS cases was associated with increased risk for invasive breast cancer. Taken together, these data demonstrate a key role for the RB-pathway in invasion associated with breast tumor progression, and shed light on the key molecular events that promote the progression of DCIS to invasive disease.
Collapse
Affiliation(s)
- A K Witkiewicz
- 1] Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA [2] Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - D W Cox
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - D Rivadeneira
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - A E Ertel
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - P Fortina
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - G F Schwartz
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - E S Knudsen
- 1] Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA [2] Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
47
|
Loessner D, Kobel S, Clements JA, Lutolf MP, Hutmacher DW. Hydrogel Microwell Arrays Allow the Assessment of Protease-Associated Enhancement of Cancer Cell Aggregation and Survival. MICROARRAYS 2013; 2:208-27. [PMID: 27605189 PMCID: PMC5003461 DOI: 10.3390/microarrays2030208] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 07/31/2013] [Accepted: 08/13/2013] [Indexed: 01/03/2023]
Abstract
Current routine cell culture techniques are only poorly suited to capture the physiological complexity of tumor microenvironments, wherein tumor cell function is affected by intricate three-dimensional (3D), integrin-dependent cell-cell and cell-extracellular matrix (ECM) interactions. 3D cell cultures allow the investigation of cancer-associated proteases like kallikreins as they degrade ECM proteins and alter integrin signaling, promoting malignant cell behaviors. Here, we employed a hydrogel microwell array platform to probe using a high-throughput mode how ovarian cancer cell aggregates of defined size form and survive in response to the expression of kallikreins and treatment with paclitaxel, by performing microscopic, quantitative image, gene and protein analyses dependent on the varying microwell and aggregate sizes. Paclitaxel treatment increased aggregate formation and survival of kallikrein-expressing cancer cells and levels of integrins and integrin-related factors. Cancer cell aggregate formation was improved with increasing aggregate size, thereby reducing cell death and enhancing integrin expression upon paclitaxel treatment. Therefore, hydrogel microwell arrays are a powerful tool to screen the viability of cancer cell aggregates upon modulation of protease expression, integrin engagement and anti-cancer treatment providing a micro-scaled yet high-throughput technique to assess malignant progression and drug-resistance.
Collapse
Affiliation(s)
- Daniela Loessner
- Faculty of Health, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia.
| | - Stefan Kobel
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Building AI 3138, Station 15, CH-1015 Lausanne, Switzerland.
| | - Judith A Clements
- Faculty of Health, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia.
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Building AI 3138, Station 15, CH-1015 Lausanne, Switzerland.
| | - Dietmar W Hutmacher
- Faculty of Science and Engineering, IHBI, QUT, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia.
| |
Collapse
|
48
|
Kaur H, Mao S, Shah S, Gorski DH, Krawetz SA, Sloane BF, Mattingly RR. Next-generation sequencing: a powerful tool for the discovery of molecular markers in breast ductal carcinoma in situ. Expert Rev Mol Diagn 2013; 13:151-65. [PMID: 23477556 DOI: 10.1586/erm.13.4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mammographic screening leads to frequent biopsies and concomitant overdiagnosis of breast cancer, particularly ductal carcinoma in situ (DCIS). Some DCIS lesions rapidly progress to invasive carcinoma, whereas others remain indolent. Because we cannot yet predict which lesions will not progress, all DCIS is regarded as malignant, and many women are overtreated. Thus, there is a pressing need for a panel of molecular markers in addition to the current clinical and pathological factors to provide prognostic information. Genomic technologies such as microarrays have made major contributions to defining subtypes of breast cancer. Next-generation sequencing (NGS) modalities offer unprecedented depth of expression analysis through revealing transcriptional boundaries, mutations, rare transcripts and alternative splice variants. NGS approaches are just beginning to be applied to DCIS. Here, the authors review the applications and challenges of NGS in discovering novel potential therapeutic targets and candidate biomarkers in the premalignant progression of breast cancer.
Collapse
Affiliation(s)
- Hitchintan Kaur
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
ARF6-regulated endocytosis of growth factor receptors links cadherin-based adhesion to canonical Wnt signaling in epithelia. Mol Cell Biol 2013; 33:2963-75. [PMID: 23716594 DOI: 10.1128/mcb.01698-12] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Wnt signaling has an essential role in embryonic development as well as stem/progenitor cell renewal, and its aberrant activation is implicated in many diseases, including several cancers. β-Catenin is a critical component of Wnt-mediated transcriptional activation. Here we show that ARF6 activation during canonical Wnt signaling promotes the intracellular accumulation of β-catenin via a mechanism that involves the endocytosis of growth factor receptors and robust activation of extracellular signal-regulated kinase (ERK). ERK promotes casein kinase 2-mediated phosphorylation of α-catenin, leading to destabilization of the adherens junctions and a subsequent increase in cytoplasmic pools of active β-catenin and E-cadherin. ERK also phosphorylates LRP6 to amplify the Wnt transduction pathway. The aforementioned Wnt-ERK signaling pathway initiates lumen filling of epithelial cysts by promoting cell proliferation in three-dimensional cell cultures. This study elucidates a mechanism responsible for the switch in β-catenin functions in cell adhesion at the adherens junctions and Wnt-induced nuclear signaling.
Collapse
|
50
|
Zhuang Y, Wang X, Nguyen HT, Zhuo Y, Cui X, Fewell C, Flemington EK, Shan B. Induction of long intergenic non-coding RNA HOTAIR in lung cancer cells by type I collagen. J Hematol Oncol 2013; 6:35. [PMID: 23668363 PMCID: PMC3655931 DOI: 10.1186/1756-8722-6-35] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 05/07/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The tumor microenvironment is a crucial determinant in tumor progression. Interstitial extracellular matrix (ECM), such as type I collagen (Col-1), is aberrantly enriched in the tumor microenvironment and promotes tumor progression. Long intergenic non-coding RNAs (lincRNA) are a new family of regulatory RNAs that modulate fundamental cellular processes via diverse mechanisms. FINDINGS We investigated whether the expression of lincRNAs was regulated by the tumor promoting Col-1. In a three-dimensional organotypic culture model using the reconstituted basement membrane ECM Matrigel (rBM 3-D), supplementation of Col-1 disrupted acini, a differentiation feature of well-differentiated lung adenocarcinoma cells, and concurrently induced the expression of a tumor-promoting lincRNA, HOX transcript antisense RNA (HOTAIR). Induction of HOTAIR by Col-1 was diminished by a neutralizing antibody against the Col-1 receptor α2β1 integrin. Col-1 activates the expression of a reporter gene controlled by the human HOTAIR promoter. Moreover the expression of HOTAIR and Col-1 was concurrently up-regulated in human non-small cell lung cancer. CONCLUSIONS Our findings indicate that tumor-promoting Col-1 up-regulates the expression of HOTAIR in NSCLC cells. These initial results warrant further investigation of HOTAIR and other lincRNA genes in lung tumorigenesis.
Collapse
Affiliation(s)
- Yan Zhuang
- Department of Medicine and Pathology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|