1
|
Boyang H, Yangyanqiu W, Wenting R, Chenxin Y, Jian C, Zhanbo Q, Yanjun Y, Qiang Y, Shuwen H. Application and progress of highcontent imaging in molecular biology. Biotechnol J 2023; 18:e2300170. [PMID: 37639283 DOI: 10.1002/biot.202300170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Humans have adopted many different methods to explore matter imaging, among which high content imaging (HCI) could conduct automated imaging analysis of cells while maintaining its structural and functional integrity. Meanwhile, as one of the most important research tools for diagnosing human diseases, HCI is widely used in the frontier of medical research, and its future application has attracted researchers' great interests. Here, the meaning of HCI was briefly explained, the history of optical imaging and the birth of HCI were described, and the experimental methods of HCI were described. Furthermore, the directions of the application of HCI were highlighted in five aspects: protein localization changes, gene identification, chemical and genetic analysis, microbiology, and drug discovery. Most importantly, some challenges and future directions of HCI were discussed, and the application and optimization of HCI were expected to be further explored.
Collapse
Affiliation(s)
- Hu Boyang
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Wang Yangyanqiu
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Rui Wenting
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Yan Chenxin
- Shulan International Medical School, Zhejiang Shuren University, Hangzhou, China
| | - Chu Jian
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
| | - Qu Zhanbo
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
| | - Yao Yanjun
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Yan Qiang
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Han Shuwen
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
| |
Collapse
|
2
|
Guo Z, Yang CT, Chien CC, Selth LA, Bagnaninchi PO, Thierry B. Optical Cellular Micromotion: A New Paradigm to Measure Tumor Cells Invasion within Gels Mimicking the 3D Tumor Environments. SMALL METHODS 2022; 6:e2200471. [PMID: 35764869 DOI: 10.1002/smtd.202200471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/22/2022] [Indexed: 06/15/2023]
Abstract
Measuring tumor cell invasiveness through 3D tissues, particularly at the single-cell level, can provide important mechanistic understanding and assist in identifying therapeutic targets of tumor invasion. However, current experimental approaches, including standard in vitro invasion assays, have limited physiological relevance and offer insufficient insight into the vast heterogeneity in tumor cell migration through tissues. To address these issues, here the concept of optical cellular micromotion is reported on, where digital holographic microscopy is used to map the optical nano- to submicrometer thickness fluctuations within single-cells. These fluctuations are driven by the dynamic movement of subcellular structures including the cytoskeleton and inherently associated with the biological processes involved in cell invasion within tissues. It is experimentally demonstrated that the optical cellular micromotion correlates with tumor cells motility and invasiveness both at the population and single-cell levels. In addition, the optical cellular micromotion significantly reduced upon treatment with migrastatic drugs that inhibit tumor cell invasion. These results demonstrate that micromotion measurements can rapidly and non-invasively determine the invasive behavior of single tumor cells within tissues, yielding a new and powerful tool to assess the efficacy of approaches targeting tumor cell invasiveness.
Collapse
Affiliation(s)
- Zhaobin Guo
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Chih-Tsung Yang
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Chia-Chi Chien
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Luke A Selth
- Flinders Health and Medical Research Institute and Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA, 5042, Australia
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Male Health and Wellbeing, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Pierre O Bagnaninchi
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes, SA, 5095, Australia
| |
Collapse
|
3
|
Gerckens M, Schorpp K, Pelizza F, Wögrath M, Reichau K, Ma H, Dworsky AM, Sengupta A, Stoleriu MG, Heinzelmann K, Merl-Pham J, Irmler M, Alsafadi HN, Trenkenschuh E, Sarnova L, Jirouskova M, Frieß W, Hauck SM, Beckers J, Kneidinger N, Behr J, Hilgendorff A, Hadian K, Lindner M, Königshoff M, Eickelberg O, Gregor M, Plettenburg O, Yildirim AÖ, Burgstaller G. Phenotypic drug screening in a human fibrosis model identified a novel class of antifibrotic therapeutics. SCIENCE ADVANCES 2021; 7:eabb3673. [PMID: 34936468 PMCID: PMC8694600 DOI: 10.1126/sciadv.abb3673] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Fibrogenic processes instigate fatal chronic diseases leading to organ failure and death. Underlying biological processes involve induced massive deposition of extracellular matrix (ECM) by aberrant fibroblasts. We subjected diseased primary human lung fibroblasts to an advanced three-dimensional phenotypic high-content assay and screened a repurposing drug library of small molecules for inhibiting ECM deposition. Fibrotic Pattern Detection by Artificial Intelligence identified tranilast as an effective inhibitor. Structure-activity relationship studies confirmed N-(2-butoxyphenyl)-3-(phenyl)acrylamides (N23Ps) as a novel and highly potent compound class. N23Ps suppressed myofibroblast transdifferentiation, ECM deposition, cellular contractility, and altered cell shapes, thus advocating a unique mode of action. Mechanistically, transcriptomics identified SMURF2 as a potential therapeutic target network. Antifibrotic activity of N23Ps was verified by proteomics in a human ex vivo tissue fibrosis disease model, suppressing profibrotic markers SERPINE1 and CXCL8. Conclusively, N23Ps are a novel class of highly potent compounds inhibiting organ fibrosis in patients.
Collapse
Affiliation(s)
- Michael Gerckens
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Francesco Pelizza
- Chemical and Process Engineering, Strathclyde University, Glasgow, Scotland, UK
| | - Melanie Wögrath
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
| | - Kora Reichau
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Leibniz Universität Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research (BMWZ), Hannover, Germany
| | - Huilong Ma
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Leibniz Universität Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research (BMWZ), Hannover, Germany
| | - Armando-Marco Dworsky
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
| | - Arunima Sengupta
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Mircea Gabriel Stoleriu
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Asklepios Fachkliniken Munich-Gauting, Munich, Germany
| | - Katharina Heinzelmann
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Juliane Merl-Pham
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Hani N. Alsafadi
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Wallenberg Center for Molecular Medicine (WCMM), Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Eduard Trenkenschuh
- Department of Pharmacy–Center for Drug Research, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximillians University of Munich, Munich, Germany
| | - Lenka Sarnova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa Jirouskova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Wolfgang Frieß
- Department of Pharmacy–Center for Drug Research, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximillians University of Munich, Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Chair of Experimental Genetics, Technische Universität München, 85354 Freising, Germany
| | - Nikolaus Kneidinger
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Department of Internal Medicine V, Ludwig-Maximillians University of Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jürgen Behr
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Asklepios Fachkliniken Munich-Gauting, Munich, Germany
- Department of Internal Medicine V, Ludwig-Maximillians University of Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Lindner
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Asklepios Fachkliniken Munich-Gauting, Munich, Germany
- Paracelsus Medical Private University, Salzburg, Austria
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oliver Plettenburg
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Leibniz Universität Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research (BMWZ), Hannover, Germany
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Ali Önder Yildirim
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerald Burgstaller
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, Munich, Germany
- Corresponding author.
| |
Collapse
|
4
|
Karakioulaki M, Papakonstantinou E, Stolz D. Extracellular matrix remodelling in COPD. Eur Respir Rev 2020; 29:29/158/190124. [PMID: 33208482 DOI: 10.1183/16000617.0124-2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/16/2020] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the lung plays several important roles in lung function, as it offers a low resistant pathway that allows the exchange of gases, provides compressive strength and elasticity that supports the fragile alveolar-capillary intersection, controls the binding of cells with growth factors and cell surface receptors and acts as a buffer against retention of water.COPD is a chronic inflammatory respiratory condition, characterised by various conditions that result in progressive airflow limitation. At any stage in the course of the disease, acute exacerbations of COPD may occur and lead to accelerated deterioration of pulmonary function. A key factor of COPD is airway remodelling, which refers to the serious alterations of the ECM affecting airway wall thickness, resistance and elasticity. Various studies have shown that serum biomarkers of ECM turnover are significantly associated with disease severity in patients with COPD and may serve as potential targets to control airway inflammation and remodelling in COPD. Unravelling the complete molecular composition of the ECM in the diseased lungs will help to identify novel biomarkers for disease progression and therapy.
Collapse
Affiliation(s)
- Meropi Karakioulaki
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland.,Dept of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Daiana Stolz
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| |
Collapse
|
5
|
Gels for Live Analysis of Compartmentalized Environments (GLAnCE): A tissue model to probe tumour phenotypes at tumour-stroma interfaces. Biomaterials 2020; 228:119572. [DOI: 10.1016/j.biomaterials.2019.119572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/19/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022]
|
6
|
Celie KB, Toyoda Y, Dong X, Morrison KA, Zhang P, Asanbe O, Jin JL, Hooper RC, Zanotelli MR, Kaymakcalan O, Bender RJ, Spector JA. Microstructured hydrogel scaffolds containing differential density interfaces promote rapid cellular invasion and vascularization. Acta Biomater 2019; 91:144-158. [PMID: 31004845 DOI: 10.1016/j.actbio.2019.04.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Insufficient vascularization of currently available clinical biomaterials has limited their application to optimal wound beds. We designed a hydrogel scaffold with a unique internal microstructure of differential collagen densities to induce cellular invasion and neovascularization. METHODS Microsphere scaffolds (MSS) were fabricated by encasing 1% (w/v) type 1 collagen microspheres 50-150 μm in diameter in 0.3% collagen bulk. 1% and 0.3% monophase collagen scaffolds and Integra® disks served as controls. Mechanical characterization as well as in vitro and in vivo invasion assays were performed. Cell number and depth of invasion were analyzed using Imaris™. Cell identity was assessed immunohistochemically. RESULTS In vitro, MSS exhibited significantly greater average depth of cellular invasion than Integra® and monophase collagen controls. MSS also demonstrated significantly higher cell counts than controls. In vivo, MSS revealed significantly more cellular invasion spanning the entire scaffold depth at 14 days than Integra®. CD31+ expressing luminal structures suggestive of neovasculature were seen within MSS at 7 days and were more prevalent after 14 days. Multiphoton microscopy of MSS demonstrated erythrocytes within luminal structures after 14 days. CONCLUSION By harnessing simple architectural cues to induce cellular migration, MSS holds great potential for clinical translation as the next generation dermal replacement product. STATEMENT OF SIGNIFICANCE Large skin wounds require tissue engineered dermal substitutes in order to promote healing. Currently available dermal replacement products do not always adequately incorporate into the body, especially in complex wounds, due to poor neovascularization. In this paper, we present a hydrogel with an innovative microarchitecture that is composed of dense type I collagen microspheres suspended in a less-dense collagen bulk. We show that cell invasion into the scaffold is driven solely by mechanical cues inherent within this differential density interface, and that this induces robust vascular cell invasion both in vitro and in a rodent model. Our hydrogel performs favorably compared to the current clinical gold standard, Integra®. We believe this hydrogel scaffold may be the first of the next generation of dermal replacement products.
Collapse
Affiliation(s)
- Karel-Bart Celie
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Yoshiko Toyoda
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Xue Dong
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Kerry A Morrison
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Peipei Zhang
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Ope Asanbe
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Julia L Jin
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Rachel C Hooper
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 121A Weill Hall, Ithaca, NY 14853, United States
| | - Omer Kaymakcalan
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Ryan J Bender
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States
| | - Jason A Spector
- Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical Center, 1300 York, Room A-821, New York, NY 10021, United States; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 121A Weill Hall, Ithaca, NY 14853, United States.
| |
Collapse
|
7
|
Gladilin E, Ohse S, Boerries M, Busch H, Xu C, Schneider M, Meister M, Eils R. TGFβ-induced cytoskeletal remodeling mediates elevation of cell stiffness and invasiveness in NSCLC. Sci Rep 2019; 9:7667. [PMID: 31113982 PMCID: PMC6529472 DOI: 10.1038/s41598-019-43409-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/25/2019] [Indexed: 02/07/2023] Open
Abstract
Importance of growth factor (GF) signaling in cancer progression is widely acknowledged. Transforming growth factor beta (TGFβ) is known to play a key role in epithelial-to-mesenchymal transition (EMT) and metastatic cell transformation that are characterized by alterations in cell mechanical architecture and behavior towards a more robust and motile single cell phenotype. However, mechanisms mediating cancer type specific enhancement of cell mechanical phenotype in response to TGFβ remain poorly understood. Here, we combine high-throughput mechanical cell phenotyping, microarray analysis and gene-silencing to dissect cytoskeletal mediators of TGFβ-induced changes in mechanical properties of on-small-cell lung carcinoma (NSCLC) cells. Our experimental results show that elevation of rigidity and invasiveness of TGFβ-stimulated NSCLC cells correlates with upregulation of several cytoskeletal and motor proteins including vimentin, a canonical marker of EMT, and less-known unconventional myosins. Selective probing of gene-silenced cells lead to identification of unconventional myosin MYH15 as a novel mediator of elevated cell rigidity and invasiveness in TGFβ-stimulated NSCLC cells. Our experimental results provide insights into TGFβ-induced cytoskeletal remodeling of NSCLC cells and suggest that mediators of elevated cell stiffness and migratory activity such as unconventional cytoskeletal and motor proteins may represent promising pharmaceutical targets for restraining invasive spread of lung cancer.
Collapse
Affiliation(s)
- E Gladilin
- German Cancer Research Center, Div. Bioinformatics and Omics Data Analytics, Mathematikon - Berliner Str. 41, 69120, Heidelberg, Germany. .,University Heidelberg, BioQuant, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany. .,Leibniz Institute of Plant Genetics and Crop Plant Research, OT Gatersleben Corrensstrasse 3, 06466, Seeland, Germany.
| | - S Ohse
- University of Freiburg, Institute of Molecular Medicine and Cell Research (IMMZ), Stefan-Meier-Str. 17, 79104, Freiburg, Germany
| | - M Boerries
- University of Freiburg, Institute of Molecular Medicine and Cell Research (IMMZ), Stefan-Meier-Str. 17, 79104, Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department for Biometry, Epidemiology and Medical Bioinformatics and Comprehensive Cancer Center Freiburg (CCCF), University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Breisacherstrasse 153, 79110, Freiburg, Germany
| | - H Busch
- University of Freiburg, Institute of Molecular Medicine and Cell Research (IMMZ), Stefan-Meier-Str. 17, 79104, Freiburg, Germany.,University of Lübeck, Institute of Experimental Dermatology, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - C Xu
- Thoraxklinik at Heidelberg University Hospital, Amalienstr. 5, 69126, Heidelberg, Germany
| | - M Schneider
- Thoraxklinik at Heidelberg University Hospital, Amalienstr. 5, 69126, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - M Meister
- Thoraxklinik at Heidelberg University Hospital, Amalienstr. 5, 69126, Heidelberg, Germany
| | - R Eils
- Center for Digital Health, Berlin Institute of Health, and Charité Universitätsmedizin Berlin, Kapelle-Ufer 2, 10117, Berlin, Germany.,Health Data Science Unit, Heidelberg University Hospital, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| |
Collapse
|
8
|
Melnik S, Dvornikov D, Müller-Decker K, Depner S, Stannek P, Meister M, Warth A, Thomas M, Muley T, Risch A, Plass C, Klingmüller U, Niehrs C, Glinka A. Cancer cell specific inhibition of Wnt/β-catenin signaling by forced intracellular acidification. Cell Discov 2018; 4:37. [PMID: 29977599 PMCID: PMC6028397 DOI: 10.1038/s41421-018-0033-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 01/02/2023] Open
Abstract
Use of the diabetes type II drug Metformin is associated with a moderately lowered risk of cancer incidence in numerous tumor entities. Studying the molecular changes associated with the tumor-suppressive action of Metformin we found that the oncogene SOX4, which is upregulated in solid tumors and associated with poor prognosis, was induced by Wnt/β-catenin signaling and blocked by Metformin. Wnt signaling inhibition by Metformin was surprisingly specific for cancer cells. Unraveling the underlying specificity, we identified Metformin and other Mitochondrial Complex I (MCI) inhibitors as inducers of intracellular acidification in cancer cells. We demonstrated that acidification triggers the unfolded protein response to induce the global transcriptional repressor DDIT3, known to block Wnt signaling. Moreover, our results suggest that intracellular acidification universally inhibits Wnt signaling. Based on these findings, we combined MCI inhibitors with H+ ionophores, to escalate cancer cells into intracellular hyper-acidification and ATP depletion. This treatment lowered intracellular pH both in vitro and in a mouse xenograft tumor model, depleted cellular ATP, blocked Wnt signaling, downregulated SOX4, and strongly decreased stemness and viability of cancer cells. Importantly, the inhibition of Wnt signaling occurred downstream of β-catenin, encouraging applications in treatment of cancers caused by APC and β-catenin mutations.
Collapse
Affiliation(s)
- Svitlana Melnik
- 1Division of Epigenetics and Cancer Risks Factors, German Cancer Research Center, Heidelberg, D-69120 Germany.,2DNA vectors, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Dmytro Dvornikov
- 3Division of Systems Biology and Signal Transduction, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Karin Müller-Decker
- 5Tumor Models Unit, Center for Preclinical Research, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Sofia Depner
- 3Division of Systems Biology and Signal Transduction, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Peter Stannek
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Michael Meister
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,7Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, D-69126 Germany
| | - Arne Warth
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,8Institute of Pathology, Heidelberg University Hospital, Heidelberg, 69120 Germany
| | - Michael Thomas
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,7Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, D-69126 Germany
| | - Tomas Muley
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,7Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, D-69126 Germany
| | - Angela Risch
- 1Division of Epigenetics and Cancer Risks Factors, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,9Department of Molecular Biology, University of Salzburg, Salzburg, 5020 Austria.,Cancer Cluster Salzburg, Salzburg, 5020 Austria
| | - Christoph Plass
- 1Division of Epigenetics and Cancer Risks Factors, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Ursula Klingmüller
- 3Division of Systems Biology and Signal Transduction, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, D-69120 Germany.,11Institute of Molecular Biology (IMB), Mainz, 55128 Germany
| | - Andrey Glinka
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, D-69120 Germany
| |
Collapse
|
9
|
Expression ratio of the TGFβ-inducible gene MYO10 is prognostic for overall survival of squamous cell lung cancer patients and predicts chemotherapy response. Sci Rep 2018; 8:9517. [PMID: 29934580 PMCID: PMC6015003 DOI: 10.1038/s41598-018-27912-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/13/2018] [Indexed: 12/30/2022] Open
Abstract
In lung cancer a deregulation of Transforming Growth Factor-β (TGFβ) signaling has been observed. Yet, the impact of TGFβ in squamous cell carcinoma of the lung (LUSC) remained to be determined. We combined phenotypic and transcriptome-wide studies and showed that the stimulation of the LUSC cell line SK-MES1 with TGFβ results in an increase of migratory invasive properties. The analysis of the dynamics of gene expression by next-generation sequencing revealed that TGFβ stimulation orchestrates the upregulation of numerous motility- and actin cytoskeleton-related genes. Among these the non-muscle myosin 10 (MYO10) showed the highest upregulation in a LUSC patient cohort of the Cancer Genome Atlas (TCGA). Knockdown of MYO10 abrogated TGFβ-induced collagen gel invasion of SK-MES1 cells. The analysis of MYO10 mRNA expression in paired tissues of 151 LUSC patients with corresponding 80-month clinical follow-up data showed that the mRNA expression ratio of MYO10 in tumor and tumor-free tissue is prognostic for overall survival of LUSC patients and predictive for the response of these patients to adjuvant chemotherapy. Thus, MYO10 represents a new clinical biomarker for this aggressive disease and due to its role in cellular motility and invasion could serve as a potential molecular target for therapeutic interventions in patients with LUSC.
Collapse
|
10
|
Burgstaller G, Sengupta A, Vierkotten S, Preissler G, Lindner M, Behr J, Königshoff M, Eickelberg O. Distinct niches within the extracellular matrix dictate fibroblast function in (cell free) 3D lung tissue cultures. Am J Physiol Lung Cell Mol Physiol 2018; 314:L708-L723. [DOI: 10.1152/ajplung.00408.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cues from the extracellular matrix (ECM) and their functional interplay with cells play pivotal roles for development, tissue repair, and disease. However, the precise nature of this interplay remains elusive. We used an innovative 3D cell culture ECM model by decellularizing 300-µm-thick ex vivo lung tissue scaffolds (d3D-LTCs) derived from diseased and healthy mouse lungs, which widely mimics the native (patho)physiological in vivo ECM microenvironment. We successfully repopulated all d3D-LTCs with primary human and murine fibroblasts, and moreover, we demonstrated that the cells also populated the innermost core regions of the d3D-LTCs in a real 3D fashion. The engrafted fibroblasts revealed a striking functional plasticity, depending on their localization in distinct ECM niches of the d3D-LTCs, affecting the cells’ tissue engraftment, cellular migration rates, cell morphologies, and protein expression and phosphorylation levels. Surprisingly, we also observed fibroblasts that were homing to the lung scaffold’s interstitium as well as fibroblasts that were invading fibrotic areas. To date, the functional nature and even the existence of 3D cell matrix adhesions in vivo as well as in 3D culture models is still unclear and controversial. Here, we show that attachment of fibroblasts to the d3D-LTCs evidently occurred via focal adhesions, thus advocating for a relevant functional role in vivo. Furthermore, we found that protein levels of talin, paxillin, and zyxin and phosphorylation levels of paxillin Y118, as well as the migration-relevant small GTPases RhoA, Rac, and CDC42, were significantly reduced compared with their attachment to 2D plastic dishes. In summary, our results strikingly indicate that inherent physical or compositional characteristics of the ECM act as instructive cues altering the functional behavior of engrafted cells. Thus, d3D-LTCs might aid to obtain more realistic data in vitro, with a high relevance for drug discovery and mechanistic studies alike.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Arunima Sengupta
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Sarah Vierkotten
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerhard Preissler
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Michael Lindner
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Asklepios Fachkliniken München-Gauting, Munich, Germany
| | - Jürgen Behr
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Asklepios Fachkliniken München-Gauting, Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Division of Respiratory Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| | - Oliver Eickelberg
- Division of Respiratory Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| |
Collapse
|
11
|
Kenney RM, Lloyd CC, Whitman NA, Lockett MR. 3D cellular invasion platforms: how do paper-based cultures stack up? Chem Commun (Camb) 2018. [PMID: 28621775 DOI: 10.1039/c7cc02357j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cellular invasion is the gateway to metastasis, which is the leading cause of cancer-related deaths. Invasion is driven by a number of chemical and mechanical stresses that arise in the tumor microenvironment. In vitro assays are needed for the systematic study of cancer progress. To be truly predictive, these assays must generate tissue-like environments that can be experimentally controlled and manipulated. While two-dimensional (2D) monolayer cultures are easily assembled and evaluated, they lack the extracellular components needed to assess invasion. Three-dimensional (3D) cultures are better suited for invasion studies because they generate cellular phenotypes that are more representative of those found in vivo. This feature article provides an overview of four invasion platforms. We focus on paper-based cultures, an emerging 3D culture platform capable of generating tissue-like structures and quantifying cellular invasion. Paper-based cultures are as easily assembled and analyzed as monolayers, but provide an experimentally powerful platform capable of supporting: co-cultures and representative extracellular environments; experimentally controlled gradients; readouts capable of quantifying, discerning, and separating cells based on their invasiveness. With a series of examples we highlight the potential of paper-based cultures, and discuss how they stack up against other invasion platforms.
Collapse
Affiliation(s)
- Rachael M Kenney
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, NC 27599-3290, USA.
| | | | | | | |
Collapse
|
12
|
Voura EB, Montalvo MJ, Dela Roca KT, Fisher JM, Defamie V, Narala SR, Khokha R, Mulligan ME, Evans CA. Planarians as models of cadmium-induced neoplasia provide measurable benchmarks for mechanistic studies. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2017; 142:544-554. [PMID: 28482323 DOI: 10.1016/j.ecoenv.2017.04.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/02/2017] [Accepted: 04/19/2017] [Indexed: 06/07/2023]
Abstract
Bioassays of planarian neoplasia highlight the potential of these organisms as useful standards to assess whether environmental toxins such as cadmium promote tumorigenesis. These studies complement other investigations into the exceptional healing and regeneration of planarians - processes that are driven by a population of active stem cells, or neoblasts, which are likely transformed during planarian tumor growth. Our goal was to determine if planarian tumorigenesis assays are amenable to mechanistic studies of cadmium carcinogenesis. To that end we demonstrate, by examining both counts of cell populations by size, and instances of mitosis, that the activity of the stem cell population can be monitored. We also provide evidence that specific biomodulators can affect the potential of planarian neoplastic growth, in that an inhibitor of metalloproteinases effectively blocked the development of the lesions. From these results, we infer that neoblast activity does respond to cadmium-induced tumor growth, and that metalloproteinases are required for the progression of cancer in the planarian.
Collapse
Affiliation(s)
- Evelyn B Voura
- School of Science, Technology and Health Studies, Morrisville State College, 80 Eaton Street, Morrisville, New York 13408, USA.
| | - Melissa J Montalvo
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| | - Kevin T Dela Roca
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| | - Julia M Fisher
- Colgate University, 13 Oak Drive, Hamilton, New York 13346, USA
| | - Virginie Defamie
- Ontario Cancer Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Swami R Narala
- Ontario Cancer Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Rama Khokha
- Ontario Cancer Institute, University Health Network, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Margaret E Mulligan
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| | - Colleen A Evans
- Department of Math and Science, Dominican College, 470 Western Highway South, Orangeburg, New York 10962, USA
| |
Collapse
|
13
|
Burgstaller G, Oehrle B, Gerckens M, White ES, Schiller HB, Eickelberg O. The instructive extracellular matrix of the lung: basic composition and alterations in chronic lung disease. Eur Respir J 2017; 50:50/1/1601805. [PMID: 28679607 DOI: 10.1183/13993003.01805-2016] [Citation(s) in RCA: 305] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/29/2017] [Indexed: 12/13/2022]
Abstract
The pulmonary extracellular matrix (ECM) determines the tissue architecture of the lung, and provides mechanical stability and elastic recoil, which are essential for physiological lung function. Biochemical and biomechanical signals initiated by the ECM direct cellular function and differentiation, and thus play a decisive role in lung development, tissue remodelling processes and maintenance of adult homeostasis. Recent proteomic studies have demonstrated that at least 150 different ECM proteins, glycosaminoglycans and modifying enzymes are expressed in the lung, and these assemble into intricate composite biomaterials. These highly insoluble assemblies of interacting ECM proteins and their glycan modifications can act as a solid phase-binding interface for hundreds of secreted proteins, which creates an information-rich signalling template for cell function and differentiation. Dynamic changes within the ECM that occur upon injury or with ageing are associated with several chronic lung diseases. In this review, we summarise the available data about the structure and function of the pulmonary ECM, and highlight changes that occur in idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension (PAH), chronic obstructive pulmonary disease (COPD), asthma and lung cancer. We discuss potential mechanisms of ECM remodelling and modification, which we believe are relevant for future diagnosis and treatment of chronic lung disease.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Bettina Oehrle
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Michael Gerckens
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Herbert B Schiller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Oliver Eickelberg
- Division of Respiratory Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
| |
Collapse
|
14
|
Parlato M, Molenda J, Murphy WL. Specific recruitment of circulating angiogenic cells using biomaterials as filters. Acta Biomater 2017; 56:65-79. [PMID: 28373084 DOI: 10.1016/j.actbio.2017.03.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/03/2017] [Accepted: 03/28/2017] [Indexed: 02/08/2023]
Abstract
Endogenous recruitment of circulating angiogenic cells (CACs) is an emerging strategy to induce angiogenesis within a defect site, and multiple recent strategies have deployed soluble protein releasing biomaterials for this purpose. However, the way in which the design of biomaterials affects CAC recruitment and invasion are poorly understood. Here we used an enhanced-throughput cell invasion assay to systematically examine the effects of biomaterial design on CAC recruitment. The screens co-optimized hydrogel presentation of a stromal-derived factor-1α (SDF-1α) gradient, hydrogel degradability, and hydrogel stiffness for maximal CAC invasion. We also examined the specificity of this invasion by assessing dermal fibroblast, mesenchymal stem cell, and lymphocyte invasion individually and in co-culture with CACs to identify hydrogels specific to CAC invasion. These screens suggested a subset of MMP-degradable hydrogels presenting a specific range of SDF-1α gradient slopes that induced specific invasion of CACs, and we posit that the design parameters of this subset of hydrogels may serve as instructive templates for the future design of biomaterials to specifically recruit CACs. We also posit that this design concept may be applied more broadly in that it may be possible to utilize these specific subsets of biomaterials as "filters" to control which types of cell populations invade into and populate the biomaterial. STATEMENT OF SIGNIFICANCE The recruitment of specific cell types for cell-based therapies in vivo is of great interest to the regenerative medicine community. Circulating angiogenic cells (CACs), CD133+ cells derived from the blood stream, are of particular interest for induction of angiogenesis in ischemic tissues, and recent studies utilizing soluble-factor releasing biomaterials to recruit these cells in vivo show great promise. However, these studies are largely "proof of concept" and are not systematic in nature. Thus, little is currently known about how biomaterial design affects the recruitment of CACs. In the present work, we use a high throughput cell invasion screening platform to systematically examine the effects of biomaterial design on circulating angiogenic cell (CAC) recruitment, and we successfully screened 263 conditions at 3 replicates each. Our results identify a particular subset of conditions that robustly recruit CACs. Additionally, we found that these conditions also specifically recruited CACs and excluded the other tested cells types of dermal fibroblasts, mesenchymal stem cells, and lymphocytes. This suggests an intriguing new role for biomaterials as "filters" to control the types of cells that invade and populate that biomaterial.
Collapse
|
15
|
Ahluwalia N, Grasberger PE, Mugo BM, Feghali-Bostwick C, Pardo A, Selman M, Lagares D, Tager AM. Fibrogenic Lung Injury Induces Non-Cell-Autonomous Fibroblast Invasion. Am J Respir Cell Mol Biol 2017; 54:831-42. [PMID: 26600305 DOI: 10.1165/rcmb.2015-0040oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pathologic accumulation of fibroblasts in pulmonary fibrosis appears to depend on their invasion through basement membranes and extracellular matrices. Fibroblasts from the fibrotic lungs of patients with idiopathic pulmonary fibrosis (IPF) have been demonstrated to acquire a phenotype characterized by increased cell-autonomous invasion. Here, we investigated whether fibroblast invasion is further stimulated by soluble mediators induced by lung injury. We found that bronchoalveolar lavage fluids from bleomycin-challenged mice or patients with IPF contain mediators that dramatically increase the matrix invasion of primary lung fibroblasts. Further characterization of this non-cell-autonomous fibroblast invasion suggested that the mediators driving this process are produced locally after lung injury and are preferentially produced by fibrogenic (e.g., bleomycin-induced) rather than nonfibrogenic (e.g., LPS-induced) lung injury. Comparison of invasion and migration induced by a series of fibroblast-active mediators indicated that these two forms of fibroblast movement are directed by distinct sets of stimuli. Finally, knockdown of multiple different membrane receptors, including platelet-derived growth factor receptor-β, lysophosphatidic acid 1, epidermal growth factor receptor, and fibroblast growth factor receptor 2, mitigated the non-cell-autonomous fibroblast invasion induced by bronchoalveolar lavage from bleomycin-injured mice, suggesting that multiple different mediators drive fibroblast invasion in pulmonary fibrosis. The magnitude of this mediator-driven fibroblast invasion suggests that its inhibition could be a novel therapeutic strategy for pulmonary fibrosis. Further elaboration of the molecular mechanisms that drive non-cell-autonomous fibroblast invasion consequently may provide a rich set of novel drug targets for the treatment of IPF and other fibrotic lung diseases.
Collapse
Affiliation(s)
- Neil Ahluwalia
- 1 Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Paula E Grasberger
- 1 Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Brian M Mugo
- 1 Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Carol Feghali-Bostwick
- 2 Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Annie Pardo
- 3 Universidad Nacional Autónoma de México, Mexico City, Mexico; and
| | - Moisés Selman
- 4 Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - David Lagares
- 1 Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrew M Tager
- 1 Division of Pulmonary and Critical Care Medicine and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Surolia R, Li FJ, Wang Z, Li H, Liu G, Zhou Y, Luckhardt T, Bae S, Liu RM, Rangarajan S, de Andrade J, Thannickal VJ, Antony VB. 3D pulmospheres serve as a personalized and predictive multicellular model for assessment of antifibrotic drugs. JCI Insight 2017; 2:e91377. [PMID: 28138565 DOI: 10.1172/jci.insight.91377] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal progressive fibrotic lung disease characterized by the presence of invasive myofibroblasts in the lung. Currently, there are only two FDA-approved drugs (pirfenidone and nintedanib) for the treatment of IPF. There are no defined criteria to guide specific drug therapy. New methodologies are needed not only to predict personalized drug therapy, but also to screen novel molecules that are on the horizon for treatment of IPF. We have developed a model system that exploits the invasive phenotype of IPF lung tissue. This ex vivo 3D model uses lung tissue from patients to develop pulmospheres. Pulmospheres are 3D spheroids composed of cells derived exclusively from primary lung biopsies and inclusive of lung cell types reflective of those in situ, in the patient. We tested the pulmospheres of 20 subjects with IPF and 9 control subjects to evaluate the responsiveness of individual patients to antifibrotic drugs. Clinical parameters and outcomes were also followed in the same patients. Our results suggest that pulmospheres simulate the microenvironment in the lung and serve as a personalized and predictive model for assessing responsiveness to antifibrotic drugs in patients with IPF.
Collapse
Affiliation(s)
- Ranu Surolia
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Fu Jun Li
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Zheng Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Huashi Li
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Yong Zhou
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Tracy Luckhardt
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Sejong Bae
- Division of Preventative Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | | | - Joao de Andrade
- Division of Pulmonary, Allergy, and Critical Care Medicine and.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine and.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Veena B Antony
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| |
Collapse
|
17
|
Marwitz S, Depner S, Dvornikov D, Merkle R, Szczygieł M, Müller-Decker K, Lucarelli P, Wäsch M, Mairbäurl H, Rabe KF, Kugler C, Vollmer E, Reck M, Scheufele S, Kröger M, Ammerpohl O, Siebert R, Goldmann T, Klingmüller U. Downregulation of the TGFβ Pseudoreceptor BAMBI in Non-Small Cell Lung Cancer Enhances TGFβ Signaling and Invasion. Cancer Res 2016; 76:3785-801. [PMID: 27197161 DOI: 10.1158/0008-5472.can-15-1326] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 03/11/2016] [Indexed: 11/16/2022]
Abstract
Non-small cell lung cancer (NSCLC) is characterized by early metastasis and has the highest mortality rate among all solid tumors, with the majority of patients diagnosed at an advanced stage where curative therapeutic options are lacking. In this study, we identify a targetable mechanism involving TGFβ elevation that orchestrates tumor progression in this disease. Substantial activation of this pathway was detected in human lung cancer tissues with concomitant downregulation of BAMBI, a negative regulator of the TGFβ signaling pathway. Alterations of epithelial-to-mesenchymal transition (EMT) marker expression were observed in lung cancer samples compared with tumor-free tissues. Distinct alterations in the DNA methylation of the gene regions encoding TGFβ pathway components were detected in NSCLC samples compared with tumor-free lung tissues. In particular, epigenetic silencing of BAMBI was identified as a hallmark of NSCLC. Reconstitution of BAMBI expression in NSCLC cells resulted in a marked reduction of TGFβ-induced EMT, migration, and invasion in vitro, along with reduced tumor burden and tumor growth in vivo In conclusion, our results demonstrate how BAMBI downregulation drives the invasiveness of NSCLC, highlighting TGFβ signaling as a candidate therapeutic target in this setting. Cancer Res; 76(13); 3785-801. ©2016 AACR.
Collapse
Affiliation(s)
- Sebastian Marwitz
- Pathology of the University Hospital of Lübeck and the Leibniz Research Center Borstel, Borstel, Germany. Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany
| | - Sofia Depner
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. BIOQUANT, University of Heidelberg, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Dmytro Dvornikov
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Ruth Merkle
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. BIOQUANT, University of Heidelberg, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Magdalena Szczygieł
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | | | - Philippe Lucarelli
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Marvin Wäsch
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Heimo Mairbäurl
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany. Medical Clinic VII, Sports Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Klaus F Rabe
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. LungenClinic Groβhansdorf, Groβhansdorf, Germany. Christian Albrechts University Kiel, Kiel, Germany
| | - Christian Kugler
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. LungenClinic Groβhansdorf, Groβhansdorf, Germany
| | - Ekkehard Vollmer
- Pathology of the University Hospital of Lübeck and the Leibniz Research Center Borstel, Borstel, Germany. Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany
| | - Martin Reck
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. LungenClinic Groβhansdorf, Groβhansdorf, Germany
| | - Swetlana Scheufele
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. Institute of Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Maren Kröger
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. Institute of Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ole Ammerpohl
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. Institute of Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Reiner Siebert
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany. Institute of Human Genetics, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Torsten Goldmann
- Pathology of the University Hospital of Lübeck and the Leibniz Research Center Borstel, Borstel, Germany. Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Groβhansdorf, Germany
| | - Ursula Klingmüller
- Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany. BIOQUANT, University of Heidelberg, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.
| |
Collapse
|
18
|
Fraietta I, Gasparri F. The development of high-content screening (HCS) technology and its importance to drug discovery. Expert Opin Drug Discov 2016; 11:501-14. [PMID: 26971542 DOI: 10.1517/17460441.2016.1165203] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION High-content screening (HCS) was introduced about twenty years ago as a promising analytical approach to facilitate some critical aspects of drug discovery. Its application has spread progressively within the pharmaceutical industry and academia to the point that it today represents a fundamental tool in supporting drug discovery and development. AREAS COVERED Here, the authors review some of significant progress in the HCS field in terms of biological models and assay readouts. They highlight the importance of high-content screening in drug discovery, as testified by its numerous applications in a variety of therapeutic areas: oncology, infective diseases, cardiovascular and neurodegenerative diseases. They also dissect the role of HCS technology in different phases of the drug discovery pipeline: target identification, primary compound screening, secondary assays, mechanism of action studies and in vitro toxicology. EXPERT OPINION Recent advances in cellular assay technologies, such as the introduction of three-dimensional (3D) cultures, induced pluripotent stem cells (iPSCs) and genome editing technologies (e.g., CRISPR/Cas9), have tremendously expanded the potential of high-content assays to contribute to the drug discovery process. Increasingly predictive cellular models and readouts, together with the development of more sophisticated and affordable HCS readers, will further consolidate the role of HCS technology in drug discovery.
Collapse
Affiliation(s)
- Ivan Fraietta
- a Department of Biology , Nerviano Medical Sciences S.r.l ., Nerviano , Milano , Italy
| | - Fabio Gasparri
- a Department of Biology , Nerviano Medical Sciences S.r.l ., Nerviano , Milano , Italy
| |
Collapse
|
19
|
Bartmann J, Frankenberger M, Neurohr C, Eickelberg O, Noessner E, von Wulffen W. A novel role of MMP-13 for murine DC function: its inhibition dampens T-cell activation. Int Immunol 2016; 28:473-487. [PMID: 26921214 DOI: 10.1093/intimm/dxw008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/19/2016] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) have been shown to express matrix metalloproteinase 13 (MMP-13), but little is known about its specific function in DCs and its role in inflammatory conditions. In the present study, we describe a novel role of MMP-13 in regulating the immunostimulatory function of murine DCs through moderating MHC-I surface presentation, endocytosis and cytokine/chemokine secretion. MMP-13 expression was confirmed in bone marrow-derived DCs at both the mRNA and the protein level and, furthermore, at the activity level. Remarkably, LPS treatment strongly enhanced MMP-13 mRNA expression as well as MMP-13 activity, indicating an important role of MMP-13 in inflammatory processes. Functionally, MMP-13 inhibition did not influence the DC migratory capacity, while endocytosis of ovalbumin was significantly decreased. Inhibition of MMP-13 lowered the capability of murine DCs to activate CD8+ T cells, apparently through reducing MHC-I surface presentation. Decreased surface expression of CD11c on DCs, as well as changes in the DC cytokine/chemokine profile after MMP-13 inhibition, emphasizes the influence of MMP-13 on DC function. Moreover, T-cell-targeting cytokines such as IL-12, IL-23 and IL-6 were significantly reduced. Collectively, our data reveal a novel involvement of MMP-13 in regulating DC immunobiology through moderating MHC-I surface presentation, endocytosis and cytokine/chemokine secretion. Furthermore, the reduced MHC-I surface presentation by DCs resulted in a poor CD8+ T-cell response in vitro This novel finding indicates that MMP-13 might be a promising target for therapeutic intervention in inflammatory diseases.
Collapse
Affiliation(s)
- Juliane Bartmann
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich, Asklepios Kliniken Gauting and Helmholtz Zentrum München, Member of the German Center for Lung Research, 81377 Munich, Germany
| | - Marion Frankenberger
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich, Asklepios Kliniken Gauting and Helmholtz Zentrum München, Member of the German Center for Lung Research, 81377 Munich, Germany
| | - Claus Neurohr
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich, Asklepios Kliniken Gauting and Helmholtz Zentrum München, Member of the German Center for Lung Research, 81377 Munich, Germany Department of Pneumology, Klinikum der Universität München-Großhadern, 81377 Munich, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich, Asklepios Kliniken Gauting and Helmholtz Zentrum München, Member of the German Center for Lung Research, 81377 Munich, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, Helmholtz Zentrum München, 81377 Munich, Germany
| | - Werner von Wulffen
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich, Asklepios Kliniken Gauting and Helmholtz Zentrum München, Member of the German Center for Lung Research, 81377 Munich, Germany Department of Respiratory Diseases, Klinik Augustinum München, 81375 Munich, Germany
| |
Collapse
|
20
|
Abstract
The impressive advances in the generation and interpretation of functional omics data have greatly contributed to a better understanding of the (patho-)physiology of many biological systems and led to a massive increase in the number of specific targets and phenotypes to investigate in both basic and applied research. The obvious complexity revealed by these studies represents a major challenge to the research community and asks for improved target characterisation strategies with the help of reliable, high-quality assays. Thus, the use of living cells has become an integral part of many research activities because the cellular context more closely represents target-specific interrelations and activity patterns. Although still predominant, the use of traditional two-dimensional (2D) monolayer cell culture models has been gradually complemented by studies based on three-dimensional (3D) spheroid (Sutherland 1988) and other 3D tissue culture systems (Santos et al. 2012; Matsusaki et al. 2014) in an attempt to employ model systems more closely representing the microenvironment of cells in the body. Hence, quite a variety of state-of-the-art cell culture models are available for the generation of novel chemical probes or the identification of starting points for drug development in translational research and pharma drug discovery. In order to cope with these information-rich formats and their increasing technical complexity, cell-based assay development has become a scientific research topic in its own right and is used to ensure the provision of significant, reliable and high-quality data outlasting any discussions related to the current "irreproducibility epidemic" (Dolgin 2014; Prinz et al. 2011; Schatz 2014). At the same time the use of cells in microplate assay formats has become state of the art and greatly facilitates rigorous cell-based assay development by providing the researcher with the opportunity to address the multitude of factors affecting the actual assay results in a systematic fashion and a timely manner. This microplate-based assay development strategy should result in the setting up of more robust and reliable test systems that ensure and increase the confidence in the statistical significance of the actual data generated. And, although assay miniaturisation is essential in order to achieve this, most, if not all, cell-based assays can be easily reformatted and adapted to be used in this format in a straightforward manner. This synopsis aims at summarising valuable, general observations made when implementing a diverse set of functional cellular in vitro assays at Bayer Pharma AG without claiming to deeply review all of the literature available in each and every detail. In addition, phenotypic assays (Moffat et al. 2014) or label-free detection methods (Minor 2008) are not discussed. Although this essay tries to cover the most relevant technological developments in the field, it nevertheless may express personal preferences and peculiarities of the author's approach to state-of-the-art cell-based assay development. For additional reviews covering the actual field, see Wunder et al. (2008) and Michelini et al. (2010).
Collapse
Affiliation(s)
- Gernot Langer
- Bayer Healthcare, Bayer Pharma AG, Lead Discovery Berlin - Screening, Müllerstr. 178, 13353, Berlin, Germany.
| |
Collapse
|
21
|
A novel 3D high-content assay identifies compounds that prevent fibroblast invasion into tissue surrogates. Exp Cell Res 2015; 339:35-43. [PMID: 26475730 DOI: 10.1016/j.yexcr.2015.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 09/29/2015] [Accepted: 10/02/2015] [Indexed: 11/23/2022]
Abstract
Invasion processes underlie or accompany several pathological processes but only a limited number of high-throughput capable phenotypic models exist to test anti-invasive compounds in vitro. We here evaluated 3D co-cultures as a high-content phenotypic screening system for fibrotic invasive processes. 3D multicellular spheroids were used as living tissue surrogates in co-culture with fluorescently labeled lung fibroblasts to monitor invasion processes by automated microscopy. This setup was used to screen a compound library containing 480 known bioactive substances. Identified hits prevented fibroblast invasion and could be subdivided into two hit classes. First, Prostaglandins were shown to prevent fibroblast invasion, most likely mediated by the prostaglandin EP2 receptor and generation of cAMP. Additionally, Rho-associated protein kinase (ROCK) inhibitors prevented fibroblast invasion, possibly by inactivation of myosin II. Importantly, both Prostaglandins and ROCK inhibitors are potential treatment options shown to be effective in in vitro and in vivo models of fibrotic diseases. This validates the presented novel phenotypic screening approach for the evaluation of potential inhibitors and the identification of novel compounds with activity in diseases that are associated with fibroblast invasion.
Collapse
|
22
|
Masci VL, Taddei AR, Gambellini G, Giorgi F, Fausto AM. Ultrastructural investigation on fibroblast interaction with collagen scaffold. J Biomed Mater Res A 2015; 104:272-82. [PMID: 26375405 DOI: 10.1002/jbm.a.35563] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/25/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
Collagen-based scaffolds are used as temporary or permanent coverings to help wound healing. Under natural conditions, wound healing is affected by such factors as cell types, growth factors and several components of the extracellular matrix. Due to the complexity of the cell-to-matrix interaction, many cell based mechanisms regulating wound healing in vivo are not yet properly understood. However, the whole process can be partially simulated in vitro to determine how cells interact with the collagen scaffold in relation to such features as physico-chemical properties, matrix architecture and fiber stability. Under these conditions, cell migration into the collagen matrix can be easily assessed and causally correlated with these features. In this study, we aimed at providing a structural analysis of how NIH3T3 fibroblasts migrate and proliferate in vitro when seeded on a native type-I collagen scaffold. To this end, samples were collected at regular time intervals and analyzed by light microscopy (LM), scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Through this experimental approach we demonstrate that collagen is gradually frayed into progressively thinner fibrils as fibroblasts migrate into the matrix, embrace the collagen fibers with long filopodia and form large intracellular vacuoles. A key role in this process is also played by microvesicles shed from the fibroblast plasma membrane and spread over long distances inside the collagen matrix. These observations indicate that a native type-I equine collagen provides favorable conditions for simulating collagen processing in vitro and eventually for unraveling the mechanisms controlling cell uptake and intracellular degradation.
Collapse
Affiliation(s)
- Valentina Laghezza Masci
- Department for Innovation in Biological, Agrifood and Forestry Systems, Tuscia University, Viterbo, 01100, Italy
| | - Anna Rita Taddei
- Section of Electron Microscopy, Great Equipment Center, Tuscia University, Viterbo, 01100, Italy
| | - Gabriella Gambellini
- Section of Electron Microscopy, Great Equipment Center, Tuscia University, Viterbo, 01100, Italy
| | | | - Anna Maria Fausto
- Department for Innovation in Biological, Agrifood and Forestry Systems, Tuscia University, Viterbo, 01100, Italy
| |
Collapse
|
23
|
Validated prediction of pro-invasive growth factors using a transcriptome-wide invasion signature derived from a complex 3D invasion assay. Sci Rep 2015; 5:12673. [PMID: 26243655 PMCID: PMC4525140 DOI: 10.1038/srep12673] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/03/2015] [Indexed: 12/28/2022] Open
Abstract
The invasion of activated fibroblasts represents a key pathomechanism in fibrotic diseases, carcinogenesis and metastasis. Invading fibroblasts contribute to fibrotic extracellular matrix (ECM) formation and the initiation, progression, or resistance of cancer. To construct transcriptome-wide signatures of fibroblast invasion, we used a multiplex phenotypic 3D invasion assay using lung fibroblasts. Microarray-based gene expression profiles of invading and non-invading fibroblasts demonstrated that 1,049 genes were differentially regulated (>1.5-fold). Unbiased pathway analysis (Ingenuity) identified significant enrichment for the functional clusters 'invasion of cells', 'idiopathic pulmonary fibrosis', and 'metastasis'. Matrix metalloprotease 13 (MMP13), transforming growth factor (TGF)-β1, Caveolin (Cav) 1, Phosphatase and Tensin Homolog (Pten), and secreted frizzled-related protein (Sfrp) 1 were among the highest regulated genes, confirmed by qRT-PCR and Western Blotting. We next performed in silico analysis (Ingenuity Pathway Analysis) to predict mediators that induced fibroblast invasion. Of these, TGFβ1, epidermal growth factor (EGF), fibroblast growth factor (FGF) 2, and platelet-derived growth factor (PDGF)-BB were tested in our 3D invasion assay and found to significantly induce invasion, thus validating the transcriptome profile. Accordingly, our transcriptomic invasion signature describes the invading fibroblast phenotype in unprecedented detail and provides a tool for future functional studies of cell invasion and therapeutic modulation thereof using complex phenotypic assays.
Collapse
|
24
|
Wiesner C, Le-Cabec V, El Azzouzi K, Maridonneau-Parini I, Linder S. Podosomes in space: macrophage migration and matrix degradation in 2D and 3D settings. Cell Adh Migr 2015; 8:179-91. [PMID: 24713854 DOI: 10.4161/cam.28116] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Migration of macrophages is a key process for a variety of physiological functions, such as pathogen clearance or tissue homeostasis. However, it can also be part of pathological scenarios, as in the case of tumor-associated macrophages. This review presents an overview of the different migration modes macrophages can adopt, depending on the physical and chemical properties of specific environments, and the constraints they impose upon cells. We discuss the importance of these environmental and also of cellular parameters, as well as their relative impact on macrophage migration and on the formation of matrix-lytic podosomes in 2D and 3D. Moreover, we present an overview of routinely used and also newly developed assays for the study of macrophage migration in both 2D and 3D contexts, their respective advantages and limitations, and also their potential to reliably mimic in vivo situations.
Collapse
Affiliation(s)
- Christiane Wiesner
- Institute for Medical Microbiology; Virology and Hygiene; University Medical Center Eppendorf; Hamburg, Germany
| | - Véronique Le-Cabec
- CNRS UMR 5089; IPBS (Institut de Pharmacologie et de Biologie Structurale), BP64182, 205 route de Narbonne, 31077 Toulouse Cedex 04, France; Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France
| | - Karim El Azzouzi
- Institute for Medical Microbiology; Virology and Hygiene; University Medical Center Eppendorf; Hamburg, Germany
| | - Isabelle Maridonneau-Parini
- CNRS UMR 5089; IPBS (Institut de Pharmacologie et de Biologie Structurale), BP64182, 205 route de Narbonne, 31077 Toulouse Cedex 04, France; Université de Toulouse; UPS; IPBS; F-31077 Toulouse, France; These authors contributed equally to this work
| | - Stefan Linder
- Institute for Medical Microbiology; Virology and Hygiene; University Medical Center Eppendorf; Hamburg, Germany; These authors contributed equally to this work
| |
Collapse
|
25
|
Burgstaller G, Vierkotten S, Lindner M, Königshoff M, Eickelberg O. Multidimensional immunolabeling and 4D time-lapse imaging of vital ex vivo lung tissue. Am J Physiol Lung Cell Mol Physiol 2015; 309:L323-32. [PMID: 26092995 DOI: 10.1152/ajplung.00061.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/10/2015] [Indexed: 11/22/2022] Open
Abstract
During the last decades, the study of cell behavior was largely accomplished in uncoated or extracellular matrix (ECM)-coated plastic dishes. To date, considerable cell biological efforts have tried to model in vitro the natural microenvironment found in vivo. For the lung, explants cultured ex vivo as lung tissue cultures (LTCs) provide a three-dimensional (3D) tissue model containing all cells in their natural microenvironment. Techniques for assessing the dynamic live interaction between ECM and cellular tissue components, however, are still missing. Here, we describe specific multidimensional immunolabeling of living 3D-LTCs, derived from healthy and fibrotic mouse lungs, as well as patient-derived 3D-LTCs, and concomitant real-time four-dimensional multichannel imaging thereof. This approach allowed the evaluation of dynamic interactions between mesenchymal cells and macrophages with their ECM. Furthermore, fibroblasts transiently expressing focal adhesions markers incorporated into the 3D-LTCs, paving new ways for studying the dynamic interaction between cellular adhesions and their natural-derived ECM. A novel protein transfer technology (FuseIt/Ibidi) shuttled fluorescently labeled α-smooth muscle actin antibodies into the native cells of living 3D-LTCs, enabling live monitoring of α-smooth muscle actin-positive stress fibers in native tissue myofibroblasts residing in fibrotic lesions of 3D-LTCs. Finally, this technique can be applied to healthy and diseased human lung tissue, as well as to adherent cells in conventional two-dimensional cell culture. This novel method will provide valuable new insights into the dynamics of ECM (patho)biology, studying in detail the interaction between ECM and cellular tissue components in their natural microenvironment.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| | - Sarah Vierkotten
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| | - Michael Lindner
- Center for Thoracic Surgery, Asklepios Biobank for Lung Diseases, Comprehensive Pneumology Center, Asklepios Clinic Munich-Gauting, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany; and
| |
Collapse
|
26
|
Cvetković D, Goertzen CGF, Bhattacharya M. Quantification of breast cancer cell invasiveness using a three-dimensional (3D) model. J Vis Exp 2014. [PMID: 24961804 DOI: 10.3791/51341] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It is now well known that the cellular and tissue microenvironment are critical regulators influencing tumor initiation and progression. Moreover, the extracellular matrix (ECM) has been demonstrated to be a critical regulator of cell behavior in culture and homeostasis in vivo. The current approach of culturing cells on two-dimensional (2D), plastic surfaces results in the disturbance and loss of complex interactions between cells and their microenvironment. Through the use of three-dimensional (3D) culture assays, the conditions for cell-microenvironment interaction are established resembling the in vivo microenvironment. This article provides a detailed methodology to grow breast cancer cells in a 3D basement membrane protein matrix, exemplifying the potential of 3D culture in the assessment of cell invasion into the surrounding environment. In addition, we discuss how these 3D assays have the potential to examine the loss of signaling molecules that regulate epithelial morphology by immunostaining procedures. These studies aid to identify important mechanistic details into the processes regulating invasion, required for the spread of breast cancer.
Collapse
Affiliation(s)
- Donna Cvetković
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario
| | | | - Moshmi Bhattacharya
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario; Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario; Lawson Health Research Institute;
| |
Collapse
|
27
|
Conway JRW, Carragher NO, Timpson P. Developments in preclinical cancer imaging: innovating the discovery of therapeutics. Nat Rev Cancer 2014; 14:314-28. [PMID: 24739578 DOI: 10.1038/nrc3724] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Integrating biological imaging into early stages of the drug discovery process can provide invaluable readouts of drug activity within complex disease settings, such as cancer. Iterating this approach from initial lead compound identification in vitro to proof-of-principle in vivo analysis represents a key challenge in the drug discovery field. By embracing more complex and informative models in drug discovery, imaging can improve the fidelity and statistical robustness of preclinical cancer studies. In this Review, we highlight how combining advanced imaging with three-dimensional systems and intravital mouse models can provide more informative and disease-relevant platforms for cancer drug discovery.
Collapse
Affiliation(s)
- James R W Conway
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre Sydney, St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, New South Wales 2010, Sydney, Australia
| | - Neil O Carragher
- Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Paul Timpson
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre Sydney, St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, New South Wales 2010, Sydney, Australia
| |
Collapse
|
28
|
Weigelt B, Ghajar CM, Bissell MJ. The need for complex 3D culture models to unravel novel pathways and identify accurate biomarkers in breast cancer. Adv Drug Deliv Rev 2014; 69-70:42-51. [PMID: 24412474 PMCID: PMC4186247 DOI: 10.1016/j.addr.2014.01.001] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/27/2013] [Accepted: 01/03/2014] [Indexed: 12/11/2022]
Abstract
The recent cataloging of the genomic aberrations in breast cancer has revealed the diversity and complexity of the disease at the genetic level. To unravel the functional consequences of specific repertoires of mutations and copy number changes on signaling pathways in breast cancer, it is crucial to develop model systems that truly recapitulate the disease. Here we discuss the three-dimensional culture models currently being used or recently developed for the study of normal mammary epithelial cells and breast cancer, including primary tumors and dormancy. We discuss the insights gained from these models in regards to cell signaling and potential therapeutic strategies, and the challenges that need to be met for the generation of heterotypic breast cancer model systems that are amenable for high-throughput approaches.
Collapse
Affiliation(s)
- Britta Weigelt
- Department of Pathology, Memorial-Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | - Cyrus M Ghajar
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mina J Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|