1
|
Tolentino MAK, Seyedzadeh MH, Peres NG, Du EY, Zhu L, Gaus K, Goyette J, Gooding JJ. Polyethylene Glycol-Based Hydrogel as a 3D Extracellular Matrix Mimic for Cytotoxic T Lymphocytes. J Biomed Mater Res A 2024. [PMID: 39429059 DOI: 10.1002/jbm.a.37811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024]
Abstract
Three-dimensional (3D) in vitro models enable us to understand cell behavior that is a better reflection of what occurs in vivo than 2D in vitro models. As a result, developing 3D models for extracellular matrix (ECM) has been growing exponentially. Most of the efforts for these 3D models are geared toward understanding cancer cells. An intricate network of cells that engages with cancer cells and can kill them are the immune cells, particularly cytotoxic T lymphocytes (CTLs). However, limited reports are available for 3D ECM mimics to understand CTL dynamics. Currently, we lack ECM mimetic hydrogels for immune cells, with sufficient control over variables, such as stiffness, to fully understand CTL dynamics in vitro. Here, we developed PEG-based hydrogels as ECM mimics for CTLs. The ECM mimics are targeted to mimic the stiffness of soft tissues where CTLs reside, migrate, and deliver their function. To understand cell-material interaction, we determined the porosity, biocompatibility, and stiffness of the ECM mimics. The ECM mimics have median pore sizes of 10.7 and 13.3 μm, close to the average nucleus size of CTLs (~8.6 μm), and good biocompatibility to facilitate cell migration. The stiffness of the ECM mimics corresponds to biologically relevant microenvironments such as lungs and kidneys. Using time-lapse fluorescence microscopy, 3D cell migration was imaged and measured. CTLs migrated faster in softer ECM mimic with larger pores, consistent with previous studies in collagen (the gold standard ECM mimic for CTLs). The work herein demonstrates that the PEG-based ECM mimic can serve as an in vitro tool to elucidate the cell dynamics of CTLs. Thus, this study opens possibilities to study the mechanics of CTLs in well-defined ECM mimic conditions in vitro.
Collapse
Affiliation(s)
- M A Kristine Tolentino
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Mir Hadi Seyedzadeh
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Newton Gil Peres
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Eric Yiwei Du
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lin Zhu
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Katharina Gaus
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jesse Goyette
- European Molecular Biology Laboratory Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - J Justin Gooding
- School of Chemistry and Australian Centre of NanoMedicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
2
|
Li Y, Zhu N, Ahmed M, Urbina J, Huang TY, Wong PK. Three-dimensional computer vision for exploring heterogeneity in collective Cancer Invasion. Sci Rep 2024; 14:23560. [PMID: 39384895 PMCID: PMC11464497 DOI: 10.1038/s41598-024-73688-y] [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: 03/11/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
Collective cancer invasion exhibits a hierarchical structure characterized by leader-follower organization. Dynamic gene expression analysis of invading cells using nanobiosensors within 3D microenvironments provides a valuable means to explore the regulation of leader cells during collective cancer invasion. Nonetheless, the analysis of time-lapse, multimodal images that capture the intricacies of complex invading structures and gene expression profiles in 3D tumor spheroids poses a significant technological challenge. Here, we present a computer vision-based workflow that streamlines the identification of protrusions and detached clusters from 3D tumor spheroids. This methodology not only discerns invading multicellular structures and quantifies their physical properties, but also captures gene expression patterns associated with these invasive mechanisms using an intracellular nanobiosensor. Consequently, it empowers a systematic exploration of the genotypic and phenotypic heterogeneities inherent in cancer invasion. To illustrate the effectiveness of this approach, we applied it to the analysis of a long noncoding RNA, MALAT1, in tumor spheroids derived from patients with muscle-invasive bladder cancer. Our investigation delved into the heterogeneity of cancer invasion and its relationship to MALAT1 expression. Overall, this workflow represents a valuable tool for gaining insights into the complexities of cancer invasion.
Collapse
Affiliation(s)
- Yanlin Li
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA, 16801, USA
| | - Ninghao Zhu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Mona Ahmed
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Julio Urbina
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA, 16801, USA
| | - Tai-Yin Huang
- Department of Physics, The Pennsylvania State University Lehigh Valley, Center Valley, PA, 18034, USA
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
3
|
Gagnon KA, Huang J, Hix OT, Hui VW, Hinds A, Bullitt E, Eyckmans J, Kotton DN, Chen CS. Multicompartment duct platform to study epithelial-endothelial crosstalk associated with lung adenocarcinoma. APL Bioeng 2024; 8:026126. [PMID: 38911024 PMCID: PMC11191334 DOI: 10.1063/5.0207228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
Previous lung-on-chip devices have facilitated significant advances in our understanding of lung biology and pathology. Here, we describe a novel lung-on-a-chip model in which human induced pluripotent stem cell-derived alveolar epithelial type II cells (iAT2s) form polarized duct-like lumens alongside engineered perfused vessels lined with human umbilical vein endothelium, all within a 3D, physiologically relevant microenvironment. Using this model, we investigated the morphologic and signaling consequences of the KRASG12D mutation, a commonly identified oncogene in human lung adenocarcinoma (LUAD). We show that expression of the mutant KRASG12D isoform in iAT2s leads to a hyperproliferative response and morphologic dysregulation in the epithelial monolayer. Interestingly, the mutant epithelia also drive an angiogenic response in the adjacent vasculature that is mediated by enhanced secretion of the pro-angiogenic factor soluble uPAR. These results demonstrate the functionality of a multi-cellular in vitro platform capable of modeling mutation-specific behavioral and signaling changes associated with lung adenocarcinoma.
Collapse
Affiliation(s)
| | | | | | - Veronica W. Hui
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, USA
| | - Anne Hinds
- The Pulmonary Center and Department of Medicine, Boston University Chobian & Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Esther Bullitt
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobian & Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
4
|
Ildiz ES, Gvozdenovic A, Kovacs WJ, Aceto N. Travelling under pressure - hypoxia and shear stress in the metastatic journey. Clin Exp Metastasis 2023; 40:375-394. [PMID: 37490147 PMCID: PMC10495280 DOI: 10.1007/s10585-023-10224-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023]
Abstract
Cancer cell invasion, intravasation and survival in the bloodstream are early steps of the metastatic process, pivotal to enabling the spread of cancer to distant tissues. Circulating tumor cells (CTCs) represent a highly selected subpopulation of cancer cells that tamed these critical steps, and a better understanding of their biology and driving molecular principles may facilitate the development of novel tools to prevent metastasis. Here, we describe key research advances in this field, aiming at describing early metastasis-related processes such as collective invasion, shedding, and survival of CTCs in the bloodstream, paying particular attention to microenvironmental factors like hypoxia and mechanical stress, considered as important influencers of the metastatic journey.
Collapse
Affiliation(s)
- Ece Su Ildiz
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Werner J Kovacs
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland.
| |
Collapse
|
5
|
Nitti P, Narayanan A, Pellegrino R, Villani S, Madaghiele M, Demitri C. Cell-Tissue Interaction: The Biomimetic Approach to Design Tissue Engineered Biomaterials. Bioengineering (Basel) 2023; 10:1122. [PMID: 37892852 PMCID: PMC10604880 DOI: 10.3390/bioengineering10101122] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
The advancement achieved in Tissue Engineering is based on a careful and in-depth study of cell-tissue interactions. The choice of a specific biomaterial in Tissue Engineering is fundamental, as it represents an interface for adherent cells in the creation of a microenvironment suitable for cell growth and differentiation. The knowledge of the biochemical and biophysical properties of the extracellular matrix is a useful tool for the optimization of polymeric scaffolds. This review aims to analyse the chemical, physical, and biological parameters on which are possible to act in Tissue Engineering for the optimization of polymeric scaffolds and the most recent progress presented in this field, including the novelty in the modification of the scaffolds' bulk and surface from a chemical and physical point of view to improve cell-biomaterial interaction. Moreover, we underline how understanding the impact of scaffolds on cell fate is of paramount importance for the successful advancement of Tissue Engineering. Finally, we conclude by reporting the future perspectives in this field in continuous development.
Collapse
Affiliation(s)
- Paola Nitti
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (A.N.); (R.P.); (S.V.); (M.M.); (C.D.)
| | | | | | | | | | | |
Collapse
|
6
|
Kim M, Panagiotakopoulou M, Chen C, Ruiz SB, Ganesh K, Tammela T, Heller DA. Micro-engineering and nano-engineering approaches to investigate tumour ecosystems. Nat Rev Cancer 2023; 23:581-599. [PMID: 37353679 PMCID: PMC10528361 DOI: 10.1038/s41568-023-00593-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/25/2023]
Abstract
The interactions among tumour cells, the tumour microenvironment (TME) and non-tumour tissues are of interest to many cancer researchers. Micro-engineering approaches and nanotechnologies are under extensive exploration for modelling these interactions and measuring them in situ and in vivo to investigate therapeutic vulnerabilities in cancer and extend a systemic view of tumour ecosystems. Here we highlight the greatest opportunities for improving the understanding of tumour ecosystems using microfluidic devices, bioprinting or organ-on-a-chip approaches. We also discuss the potential of nanosensors that can transmit information from within the TME or elsewhere in the body to address scientific and clinical questions about changes in chemical gradients, enzymatic activities, metabolic and immune profiles of the TME and circulating analytes. This Review aims to connect the cancer biology and engineering communities, presenting biomedical technologies that may expand the methodologies of the former, while inspiring the latter to develop approaches for interrogating cancer ecosystems.
Collapse
Affiliation(s)
- Mijin Kim
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
| | | | - Chen Chen
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, Sloan Kettering Institute, New York, NY, USA
| | - Stephen B Ruiz
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Tuomas Tammela
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
| | - Daniel A Heller
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Liang D, Liu L, Zhao Y, Luo Z, He Y, Li Y, Tang S, Tang J, Chen N. Targeting extracellular matrix through phytochemicals: a promising approach of multi-step actions on the treatment and prevention of cancer. Front Pharmacol 2023; 14:1186712. [PMID: 37560476 PMCID: PMC10407561 DOI: 10.3389/fphar.2023.1186712] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Extracellular matrix (ECM) plays a pivotal and dynamic role in the construction of tumor microenvironment (TME), becoming the focus in cancer research and treatment. Multiple cell signaling in ECM remodeling contribute to uncontrolled proliferation, metastasis, immune evasion and drug resistance of cancer. Targeting trilogy of ECM remodeling could be a new strategy during the early-, middle-, advanced-stages of cancer and overcoming drug resistance. Currently nearly 60% of the alternative anticancer drugs are derived from natural products or active ingredients or structural analogs isolated from plants. According to the characteristics of ECM, this manuscript proposes three phases of whole-process management of cancer, including prevention of cancer development in the early stage of cancer (Phase I); prevent the metastasis of tumor in the middle stage of cancer (Phase II); provide a novel method in the use of immunotherapy for advanced cancer (Phase III), and present novel insights on the contribution of natural products use as innovative strategies to exert anticancer effects by targeting components in ECM. Herein, we focus on trilogy of ECM remodeling and the interaction among ECM, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), and sort out the intervention effects of natural products on the ECM and related targets in the tumor progression, provide a reference for the development of new drugs against tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Dan Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunjie Zhao
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Zhenyi Luo
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Yadi He
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nianzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Zhu N, Ahmed M, Li Y, Liao JC, Wong PK. Long noncoding RNA MALAT1 is dynamically regulated in leader cells during collective cancer invasion. Proc Natl Acad Sci U S A 2023; 120:e2305410120. [PMID: 37364126 PMCID: PMC10319025 DOI: 10.1073/pnas.2305410120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/13/2023] [Indexed: 06/28/2023] Open
Abstract
Cancer cells collectively invade using a leader-follower organization, but the regulation of leader cells during this dynamic process is poorly understood. Using a dual double-stranded locked nucleic acid (LNA) nanobiosensor that tracks long noncoding RNA (lncRNA) dynamics in live single cells, we monitored the spatiotemporal distribution of lncRNA during collective cancer invasion. We show that the lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) is dynamically regulated in the invading fronts of cancer cells and patient-derived spheroids. MALAT1 transcripts exhibit distinct abundance, diffusivity, and distribution between leader and follower cells. MALAT1 expression increases when a cancer cell becomes a leader and decreases when the collective migration process stops. Transient knockdown of MALAT1 prevents the formation of leader cells and abolishes the invasion of cancer cells. Taken together, our single-cell analysis suggests that MALAT1 is dynamically regulated in leader cells during collective cancer invasion.
Collapse
Affiliation(s)
- Ninghao Zhu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA16802
| | - Mona Ahmed
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA16802
| | - Yanlin Li
- Department of Electrical Engineering, The Pennsylvania State University, University Park, PA16802
| | - Joseph C. Liao
- Department of Urology, Stanford University School of Medicine, Stanford, CA94305
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA16802
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA16802
- Department of Surgery, The Pennsylvania State University, University Park, PA17033
| |
Collapse
|
9
|
Ellis K, Wood R. The Comparative Invasiveness of Endometriotic Cell Lines to Breast and Endometrial Cancer Cell Lines. Biomolecules 2023; 13:1003. [PMID: 37371583 DOI: 10.3390/biom13061003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Endometriosis is an invasive condition that affects 10% of women (and people assigned as female at birth) worldwide. The purpose of this study was to characterize the relative invasiveness of three available endometriotic cell lines (EEC12Z, iEc-ESCs, tHESCs) to cancer cell lines (MDA-MB-231, SW1353 and EM-E6/E7/TERT) and assess whether the relative invasiveness was consistent across different invasion assays. All cell lines were subjected to transwell, spheroid drop, and spheroid-gel invasion assays, and stained for vimentin, cytokeratin, E-Cadherin and N-Cadherin to assess changes in expression. In all assays, endometriotic cell lines showed comparable invasiveness to the cancer cell lines used in this study, with no significant differences in invasiveness identified. EEC12Z cells that had invaded within the assay periods showed declines in E-Cadherin expression compared to cells that had not invaded within the assay period, without significant changes in N-Cadherin expression, which may support the hypothesis that an epithelial-to-mesenchymal transition is an influence on the invasiveness shown by this peritoneal endometriosis cell line.
Collapse
Affiliation(s)
- Katherine Ellis
- Department of Chemical and Process Engineering, University of Canterbury, Christchurch 8041, New Zealand
- Endometriosis New Zealand, Christchurch 8041, New Zealand
| | - Rachael Wood
- Department of Chemical and Process Engineering, University of Canterbury, Christchurch 8041, New Zealand
- The Biomolecular Interaction Centre, University of Canterbury, Christchurch 8041, New Zealand
| |
Collapse
|
10
|
Stöberl S, Balles M, Kellerer T, Rädler JO. Photolithographic microfabrication of hydrogel clefts for cell invasion studies. LAB ON A CHIP 2023; 23:1886-1895. [PMID: 36867426 DOI: 10.1039/d2lc01105k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Invasion of migrating cells into surrounding tissue plays a key role in cancer metastasis and immune response. In order to assess invasiveness, most in vitro invasion assays measure the degree to which cells migrate between microchambers that provide a chemoattractant gradient across a polymeric membrane with defined pores. However, in real tissue cells experience soft, mechanically deformable microenvironments. Here we introduce RGD-functionalized hydrogel structures that present pressurized clefts for invasive migration of cells between reservoirs maintaining a chemotactic gradient. Using UV-photolithography, equally spaced blocks of polyethylene glycol-norbornene (PEG-NB) hydrogels are formed, which subsequently swell and close the interjacent gaps. The swelling ratio and final contours of the hydrogel blocks were determined using confocal microscopy confirming a swelling induced closure of the structures. The velocity profile of cancer cells transmigrating through the clefts, which we name 'sponge clamp', is found to depend on the elastic modulus as well as the gap size between the swollen blocks. The 'sponge clamp' discriminates the invasiveness of two distinct cell lines, MDA-MB-231 and HT-1080. The approach provides soft 3D-microstructures mimicking invasion conditions in extracellular matrix.
Collapse
Affiliation(s)
- Stefan Stöberl
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| | - Miriam Balles
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| | - Thomas Kellerer
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
- Department of Applied Science and Mechatronics, University of Applied Science, Munich, Germany
| | - Joachim O Rädler
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| |
Collapse
|
11
|
Chlastáková A, Kaščáková B, Kotál J, Langhansová H, Kotsyfakis M, Kutá Smatanová I, Tirloni L, Chmelař J. Iripin-1, a new anti-inflammatory tick serpin, inhibits leukocyte recruitment in vivo while altering the levels of chemokines and adhesion molecules. Front Immunol 2023; 14:1116324. [PMID: 36756125 PMCID: PMC9901544 DOI: 10.3389/fimmu.2023.1116324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Serpins are widely distributed and functionally diverse inhibitors of serine proteases. Ticks secrete serpins with anti-coagulation, anti-inflammatory, and immunomodulatory activities via their saliva into the feeding cavity to modulate host's hemostatic and immune reaction initiated by the insertion of tick's mouthparts into skin. The suppression of the host's immune response not only allows ticks to feed on a host for several days but also creates favorable conditions for the transmission of tick-borne pathogens. Herein we present the functional and structural characterization of Iripin-1 (Ixodes ricinus serpin-1), whose expression was detected in the salivary glands of the tick Ixodes ricinus, a European vector of tick-borne encephalitis and Lyme disease. Of 16 selected serine proteases, Iripin-1 inhibited primarily trypsin and further exhibited weaker inhibitory activity against kallikrein, matriptase, and plasmin. In the mouse model of acute peritonitis, Iripin-1 enhanced the production of the anti-inflammatory cytokine IL-10 and chemokines involved in neutrophil and monocyte recruitment, including MCP-1/CCL2, a potent histamine-releasing factor. Despite increased chemokine levels, the migration of neutrophils and monocytes to inflamed peritoneal cavities was significantly attenuated following Iripin-1 administration. Based on the results of in vitro experiments, immune cell recruitment might be inhibited due to Iripin-1-mediated reduction of the expression of chemokine receptors in neutrophils and adhesion molecules in endothelial cells. Decreased activity of serine proteases in the presence of Iripin-1 could further impede cell migration to the site of inflammation. Finally, we determined the tertiary structure of native Iripin-1 at 2.10 Å resolution by employing the X-ray crystallography technique. In conclusion, our data indicate that Iripin-1 facilitates I. ricinus feeding by attenuating the host's inflammatory response at the tick attachment site.
Collapse
Affiliation(s)
- Adéla Chlastáková
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia,Laboratory of Molecular Biology of Ticks, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | - Barbora Kaščáková
- Laboratory of Structural Chemistry, Institute of Chemistry, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Jan Kotál
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Helena Langhansová
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Michail Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | - Ivana Kutá Smatanová
- Laboratory of Structural Chemistry, Institute of Chemistry, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Jindřich Chmelař
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia,*Correspondence: Jindřich Chmelař,
| |
Collapse
|
12
|
Proestaki M, Sarkar M, Burkel BM, Ponik SM, Notbohm J. Effect of hyaluronic acid on microscale deformations of collagen gels. J Mech Behav Biomed Mater 2022; 135:105465. [PMID: 36154991 PMCID: PMC9575965 DOI: 10.1016/j.jmbbm.2022.105465] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/05/2022] [Accepted: 09/10/2022] [Indexed: 11/18/2022]
Abstract
As fibrous collagen is the most abundant protein in mammalian tissues, gels of collagen fibers have been extensively used as an extracellular matrix scaffold to study how cells sense and respond to cues from their microenvironment. Other components of native tissues, such as glycosaminoglycans like hyaluronic acid, can affect cell behavior in part by changing the mechanical properties of the collagen gel. Prior studies have quantified the effects of hyaluronic acid on the mechanical properties of collagen gels in experiments of uniform shear or compression at the macroscale. However, there remains a lack of experimental studies of how hyaluronic acid changes the mechanical properties of collagen gels at the scale of a cell. Here, we studied how addition of hyaluronic acid to gels of collagen fibers affects the local field of displacements in response to contractile loads applied on length scales similar to those of a contracting cell. Using spherical poly(N-isopropylacrylamide) particles, which contract when heated, we induced displacement in gels of collagen and collagen with hyaluronic acid. Displacement fields were quantified using a combination of confocal microscopy and digital image correlation. Results showed that hyaluronic acid suppressed the distance over which displacements propagated, suggesting that it caused the network to become more linear. Additionally, hyaluronic acid had no statistical effect on heterogeneity of the displacement fields, but it did make the gels more elastic by substantially reducing the magnitude of permanent deformations. Lastly, we examined the effect of hyaluronic acid on fiber remodeling due to localized forces and found that hyaluronic acid partially - but not fully - inhibited remodeling. This result is consistent with prior studies suggesting that fiber remodeling is associated with a phase transition resulting from an instability caused by nonlinearity of the collagen gel.
Collapse
Affiliation(s)
- Maria Proestaki
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Mainak Sarkar
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian M Burkel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
13
|
Bayly-Jones C, Lupton CJ, Fritz C, Venugopal H, Ramsbeck D, Wermann M, Jäger C, de Marco A, Schilling S, Schlenzig D, Whisstock JC. Helical ultrastructure of the metalloprotease meprin α in complex with a small molecule inhibitor. Nat Commun 2022; 13:6178. [PMID: 36261433 PMCID: PMC9581967 DOI: 10.1038/s41467-022-33893-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
The zinc-dependent metalloprotease meprin α is predominantly expressed in the brush border membrane of proximal tubules in the kidney and enterocytes in the small intestine and colon. In normal tissue homeostasis meprin α performs key roles in inflammation, immunity, and extracellular matrix remodelling. Dysregulated meprin α is associated with acute kidney injury, sepsis, urinary tract infection, metastatic colorectal carcinoma, and inflammatory bowel disease. Accordingly, meprin α is the target of drug discovery programs. In contrast to meprin β, meprin α is secreted into the extracellular space, whereupon it oligomerises to form giant assemblies and is the largest extracellular protease identified to date (~6 MDa). Here, using cryo-electron microscopy, we determine the high-resolution structure of the zymogen and mature form of meprin α, as well as the structure of the active form in complex with a prototype small molecule inhibitor and human fetuin-B. Our data reveal that meprin α forms a giant, flexible, left-handed helical assembly of roughly 22 nm in diameter. We find that oligomerisation improves proteolytic and thermal stability but does not impact substrate specificity or enzymatic activity. Furthermore, structural comparison with meprin β reveal unique features of the active site of meprin α, and helical assembly more broadly.
Collapse
Affiliation(s)
- Charles Bayly-Jones
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Christopher J Lupton
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Claudia Fritz
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Hariprasad Venugopal
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, 3800, VIC, Australia
| | - Daniel Ramsbeck
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Michael Wermann
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | | | - Alex de Marco
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Stephan Schilling
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
- Hochschule Anhalt, University of Applied Sciences, Köthen, Germany
| | - Dagmar Schlenzig
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany.
| | - James C Whisstock
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia.
- EMBL Australia, Monash University, Melbourne, VIC, 3800, Australia.
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
14
|
Modeling ATP-mediated endothelial cell elongation on line patterns. Biomech Model Mechanobiol 2022; 21:1531-1548. [PMID: 35902488 PMCID: PMC9626447 DOI: 10.1007/s10237-022-01604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/24/2022] [Indexed: 11/08/2022]
Abstract
Endothelial cell (EC) migration is crucial for a wide range of processes including vascular wound healing, tumor angiogenesis, and the development of viable endovascular implants. We have previously demonstrated that ECs cultured on 15-μm wide adhesive line patterns exhibit three distinct migration phenotypes: (a) “running” cells that are polarized and migrate continuously and persistently on the adhesive lines with possible spontaneous directional changes, (b) “undecided” cells that are highly elongated and exhibit periodic changes in the direction of their polarization while maintaining minimal net migration, and (c) “tumbling-like” cells that migrate persistently for a certain amount of time but then stop and round up for a few hours before spreading again and resuming migration. Importantly, the three migration patterns are associated with distinct profiles of cell length. Because of the impact of adenosine triphosphate (ATP) on cytoskeletal organization and cell polarization, we hypothesize that the observed differences in EC length among the three different migration phenotypes are driven by differences in intracellular ATP levels. In the present work, we develop a mathematical model that incorporates the interactions between cell length, cytoskeletal (F-actin) organization, and intracellular ATP concentration. An optimization procedure is used to obtain the model parameter values that best fit the experimental data on EC lengths. The results indicate that a minimalist model based on differences in intracellular ATP levels is capable of capturing the different cell length profiles observed experimentally.
Collapse
|
15
|
Fu C, Wang J, Pallikkuth S, Ding Y, Chen J, Wren JD, Yang Y, Wong KK, Kameyama H, Jayaraman M, Munshi A, Tanaka T, Lidke KA, Zhang XA. EWI2 prevents EGFR from clustering and endocytosis to reduce tumor cell movement and proliferation. Cell Mol Life Sci 2022; 79:389. [PMID: 35773608 PMCID: PMC10428948 DOI: 10.1007/s00018-022-04417-9] [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: 02/18/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 12/01/2022]
Abstract
EWI2 is a transmembrane immunoglobulin superfamily (IgSF) protein that physically associates with tetraspanins and integrins. It inhibits cancer cells by influencing the interactions among membrane molecules including the tetraspanins and integrins. The present study revealed that, upon EWI2 silencing or ablation, the elevated movement and proliferation of cancer cells in vitro and increased cancer metastatic potential and malignancy in vivo are associated with (i) increases in clustering, endocytosis, and then activation of EGFR and (ii) enhancement of Erk MAP kinase signaling. These changes in signaling make cancer cells (i) undergo partial epithelial-to-mesenchymal (EMT) for more tumor progression and (ii) proliferate faster for better tumor formation. Inhibition of EGFR or Erk kinase can abrogate the cancer cell phenotypes resulting from EWI2 removal. Thus, to inhibit cancer cells, EWI2 prevents EGFR from clustering and endocytosis to restrain its activation and signaling.
Collapse
Affiliation(s)
- Chenying Fu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Jie Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - Yingjun Ding
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Junxiong Chen
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - Yuchao Yang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | | | | | - Anupama Munshi
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Takemi Tanaka
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - Xin A Zhang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA.
| |
Collapse
|
16
|
Canet-Jourdan C, Pagès DL, Nguyen-Vigouroux C, Cartry J, Zajac O, Desterke C, Lopez JB, Gutierrez-Mateyron E, Signolle N, Adam J, Raingeaud J, Polrot M, Gonin P, Mathieu JRR, Souquere S, Pierron G, Gelli M, Dartigues P, Ducreux M, Barresi V, Jaulin F. Patient-derived organoids identify an apico-basolateral polarity switch associated with survival in colorectal cancer. J Cell Sci 2022; 135:276070. [PMID: 35703098 DOI: 10.1242/jcs.259256] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
The metastatic progression of cancer remains a major issue in patient treatment. Yet, the molecular and cellular mechanisms underlying this process remains unclear. Here, we use primary explants and organoids from patients harboring mucinous colorectal carcinoma (MUC CRC), a poor prognosis histological form of digestive cancers, to study the architecture, invasive behavior and chemoresistance of tumor cell intermediates. We report that these tumors maintain a robust apico-basolateral polarity as they spread in the peritumoral stroma or organotypic collagen-I gels. We identified two distinct topologies: MUC CRCs either display a conventional "apical-in" polarity or, more frequently, harbor an inverted "apical-out" topology. Transcriptomic analyses combined with interference experiments on organoids showed that TGFb and focal adhesion signaling pathways are the main drivers of polarity orientation. Finally, this apical-out topology is associated with increased resistance to chemotherapeutic treatments in organoids and decreased patient survival in the clinic. Thus, patient-derived organoids have the potential to bridge histological, cellular and molecular analyses to decrypt onco-morphogenic programs and stratify cancer patients.
Collapse
Affiliation(s)
| | | | | | - Jérôme Cartry
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| | - Olivier Zajac
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005 Paris, France
| | | | | | | | - Nicolas Signolle
- INSERM Unit U981, Experimental Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Julien Adam
- INSERM Unit U981, Experimental Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Joel Raingeaud
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| | - Mélanie Polrot
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/ US 23, Gustave Roussy, Villejuif, F-94805, France
| | - Patrick Gonin
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/ US 23, Gustave Roussy, Villejuif, F-94805, France
| | | | | | | | - Maximiliano Gelli
- Department of Medical Oncology, Gustave Roussy, Villejuif, F-94805, France
| | - Peggy Dartigues
- Pathology Department, Gustave Roussy, Villejuif, F-94805, France
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Villejuif, F-94805, France.,Paris-Saclay University, Saint-Aubin, F-91190, France
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Verona 37129, Italia
| | - Fanny Jaulin
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| |
Collapse
|
17
|
Amini R, Bhatnagar A, Schlüßler R, Möllmert S, Guck J, Norden C. Amoeboid-like migration ensures correct horizontal cell layer formation in the developing vertebrate retina. eLife 2022; 11:e76408. [PMID: 35639083 PMCID: PMC9208757 DOI: 10.7554/elife.76408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
Migration of cells in the developing brain is integral for the establishment of neural circuits and function of the central nervous system. While migration modes during which neurons employ predetermined directional guidance of either preexisting neuronal processes or underlying cells have been well explored, less is known about how cells featuring multipolar morphology migrate in the dense environment of the developing brain. To address this, we here investigated multipolar migration of horizontal cells in the zebrafish retina. We found that these cells feature several hallmarks of amoeboid-like migration that enable them to tailor their movements to the spatial constraints of the crowded retina. These hallmarks include cell and nuclear shape changes, as well as persistent rearward polarization of stable F-actin. Interference with the organization of the developing retina by changing nuclear properties or overall tissue architecture hampers efficient horizontal cell migration and layer formation showing that cell-tissue interplay is crucial for this process. In view of the high proportion of multipolar migration phenomena observed in brain development, the here uncovered amoeboid-like migration mode might be conserved in other areas of the developing nervous system.
Collapse
Affiliation(s)
- Rana Amini
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Archit Bhatnagar
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Raimund Schlüßler
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Stephanie Möllmert
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und MedizinErlangenGermany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und MedizinErlangenGermany
- Physics of Life, Technische Universität DresdenDresdenGermany
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6OeirasPortugal
| |
Collapse
|
18
|
Thuault S, Ghossoub R, David G, Zimmermann P. A Journey on Extracellular Vesicles for Matrix Metalloproteinases: A Mechanistic Perspective. Front Cell Dev Biol 2022; 10:886381. [PMID: 35669514 PMCID: PMC9163832 DOI: 10.3389/fcell.2022.886381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are key players in matrix remodeling and their function has been particularly investigated in cancer biology. Indeed, through extracellular matrix (ECM) degradation and shedding of diverse cell surface macromolecules, they are implicated in different steps of tumor development, from local expansion by growth to tissue invasion and metastasis. Interestingly, MMPs are also components of extracellular vesicles (EVs). EVs are membrane-limited organelles that cells release in their extracellular environment. These "secreted" vesicles are now well accepted players in cell-to-cell communication. EVs have received a lot of interest in recent years as they are also envisioned as sources of biomarkers and as potentially outperforming vehicles for the delivery of therapeutics. Molecular machineries governing EV biogenesis, cargo loading and delivery to recipient cells are complex and still under intense investigation. In this review, we will summarize the state of the art of our knowledge about the molecular mechanisms implicated in MMP trafficking and secretion. We focus on MT1-MMP, a major effector of invasive cell behavior. We will also discuss how this knowledge is of interest for a better understanding of EV-loading of MMPs. Such knowledge might be of use to engineer novel strategies for cancer treatment. A better understanding of these mechanisms could also be used to design more efficient EV-based therapies.
Collapse
Affiliation(s)
- Sylvie Thuault
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Rania Ghossoub
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Guido David
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
- Department of Human Genetics, KU Leuven, University of Leuven, Leuven, Belgium
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
- Department of Human Genetics, KU Leuven, University of Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Abstract
A cardinal feature common to embryonic development and tissue reorganization, as well as to wound healing and cancer cell invasion, is collective cellular migration. During collective migratory events the phenomena of cell jamming and unjamming are increasingly recognized, and underlying mechanical, genomic, transcriptional, and signaling events are increasingly coming to light. In this brief perspective I propose a synthesis that brings together in a new way two key concepts. On the one hand, it has been suggested that the unjammed phase of the cellular collective evolved under a selective pressure favoring fluid-like migratory dynamics as would be required so as to accommodate episodes of tissue evolution, development, plasticity, and repair. Being dynamic, such an unjammed migratory phase is expected to be energetically expensive compared with the jammed non-migratory phase, which is presumed to have evolved under a selective pressure favoring a solid-like homeostatic regime that, by comparison, is energetically economical and mechanically stable. On the other hand, well before the discovery of cell jamming and unjamming Kauffman proposed the general biological principle that living systems exist in a solid regime near the edge of chaos, and that natural selection achieves and sustains such a poised state. Here I propose that, in certain systems at least, this poised solid-like state as predicted in the abstract by Kauffman is realized in the particular by the jammed regime just at the brink of unjamming.
Collapse
|
20
|
Shore T, Levi T, Kalifa R, Dreifuss A, Rekler D, Weinberg-Shukron A, Nevo Y, Bialistoky T, Moyal V, Gold MY, Leebhoff S, Zangen D, Deshpande G, Gerlitz O. Nucleoporin107 mediates female sexual differentiation via Dsx. eLife 2022; 11:72632. [PMID: 35311642 PMCID: PMC8975549 DOI: 10.7554/elife.72632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
We recently identified a missense mutation in Nucleoporin107 (Nup107; D447N) underlying XX-ovarian-dysgenesis, a rare disorder characterized by underdeveloped and dysfunctional ovaries. Modeling of the human mutation in Drosophila or specific knockdown of Nup107 in the gonadal soma resulted in ovarian-dysgenesis-like phenotypes. Transcriptomic analysis identified the somatic sex-determination gene doublesex (dsx) as a target of Nup107. Establishing Dsx as a primary relevant target of Nup107, either loss or gain of Dsx in the gonadal soma is sufficient to mimic or rescue the phenotypes induced by Nup107 loss. Importantly, the aberrant phenotypes induced by compromising either Nup107 or dsx are reminiscent of BMP signaling hyperactivation. Remarkably, in this context, the metalloprotease AdamTS-A, a transcriptional target of both Dsx and Nup107, is necessary for the calibration of BMP signaling. As modulation of BMP signaling is a conserved critical determinant of soma-germline interaction, the sex and tissue specific deployment of Dsx-F by Nup107 seems crucial for the maintenance of the homeostatic balance between the germ cells and somatic gonadal cells.
Collapse
Affiliation(s)
- Tikva Shore
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Tgst Levi
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Rachel Kalifa
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Amatzia Dreifuss
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Dina Rekler
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | | | - Yuval Nevo
- Bioinformatics Unit of the I-CORE Computation Center, The Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Tzofia Bialistoky
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Victoria Moyal
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Merav Yaffa Gold
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Shira Leebhoff
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - David Zangen
- Division of Pediatric Endocrinology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Offer Gerlitz
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
21
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
22
|
Chen H, Luo T, He S, Sa G. Regulatory mechanism of oral mucosal rete peg formation. J Mol Histol 2021; 52:859-868. [PMID: 34463917 DOI: 10.1007/s10735-021-10016-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/26/2021] [Indexed: 01/17/2023]
Abstract
Rete pegs are finger-like structures that are formed during the development and wound healing process of the skin and oral mucosa, and they provide better mechanical resistance and nutritional supply between the epithelium and dermis. An increasing number of studies have shown that rete pegs have physiological functions, such as resisting bacterial invasion, body fluid loss, and other harmful changes, which indicate that rete pegs are important structures in natural skin and oral mucosa. Although a great deal of progress has been made in scaffold materials and construction methods for tissue-engineered skin and oral mucosa in recent years, construction of the oral mucosa with functional rete pegs remains a major challenge. In this review, we summarized current research on the progress on formation of rete pegs in human oral mucosa as well as its molecular basis and regulatory mechanism, which might provide new ideas for functional construction of tissue-engineered skin and oral mucosa.
Collapse
Affiliation(s)
- Heng Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, People's Republic of China
| | - Tianhao Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, People's Republic of China
| | - Sangang He
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei, China.
| | - Guoliang Sa
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei, China.
| |
Collapse
|
23
|
Vilchez Mercedes SA, Bocci F, Levine H, Onuchic JN, Jolly MK, Wong PK. Decoding leader cells in collective cancer invasion. Nat Rev Cancer 2021; 21:592-604. [PMID: 34239104 DOI: 10.1038/s41568-021-00376-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/07/2023]
Abstract
Collective cancer invasion with leader-follower organization is increasingly recognized as a predominant mechanism in the metastatic cascade. Leader cells support cancer invasion by creating invasion tracks, sensing environmental cues and coordinating with follower cells biochemically and biomechanically. With the latest developments in experimental and computational models and analysis techniques, the range of specific traits and features of leader cells reported in the literature is rapidly expanding. Yet, despite their importance, there is no consensus on how leader cells arise or their essential characteristics. In this Perspective, we propose a framework for defining the essential aspects of leader cells and provide a unifying perspective on the varying cellular and molecular programmes that are adopted by each leader cell subtype to accomplish their functions. This Perspective can lead to more effective strategies to interdict a major contributor to metastatic capability.
Collapse
Affiliation(s)
| | - Federico Bocci
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Department of Physics, and Department of Bioengineering, Northeastern University, Boston, MA, USA.
| | - José N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
- Department of Physics and Astronomy, Department of Chemistry and Department of Biosciences, Rice University, Houston, TX, USA.
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA.
- Department of Mechanical Engineering and Department of Surgery, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
24
|
Kamionka EM, Qian B, Gross W, Bergmann F, Hackert T, Beretta CA, Dross N, Ryschich E. Collagen Organization Does Not Influence T-Cell Distribution in Stroma of Human Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13153648. [PMID: 34359549 PMCID: PMC8344977 DOI: 10.3390/cancers13153648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The excessive desmoplasia is the hallmark of human pancreatic cancer that influences the local T-cell-based immune response. In the present work, the stromal collagen organization in normal and malignant pancreatic tissues as well as its relationsship to T-cell distribution in pancreatic cancer were studied. It was found that differences in collagen organization do not change the spatial orientation of T-cell migration and do not influence the availability of tumor cells for T-cells. The results of the study do not support the concept of use of stroma collagen organization for improvement of spatial T-cell distribution in the tumor. Abstract The dominant intrastromal T-cell infiltration in pancreatic cancer is mainly caused by the contact guidance through the excessive desmoplastic reaction and could represent one of the obstacles to an effective immune response in this tumor type. This study analyzed the collagen organization in normal and malignant pancreatic tissues as well as its influence on T-cell distribution in pancreatic cancer. Human pancreatic tissue was analyzed using immunofluorescence staining and multiphoton and SHG microscopy supported by multistep image processing. The influence of collagen alignment on activated T-cells was studied using 3D matrices and time-lapse microscopy. It was found that the stroma of malignant and normal pancreatic tissues was characterized by complex individual organization. T-cells were heterogeneously distributed in pancreatic cancer and there was no relationship between T-cell distribution and collagen organization. There was a difference in the angular orientation of collagen alignment in the peritumoral and tumor-cell-distant stroma regions in the pancreatic ductal adenocarcinoma tissue, but there was no correlation in the T-cell densities between these regions. The grade of collagen alignment did not influence the directionality of T-cell migration in the 3D collagen matrix. It can be concluded that differences in collagen organization do not change the spatial orientation of T-cell migration or influence stromal T-cell distribution in human pancreatic cancer. The results of the present study do not support the rationale of remodeling of stroma collagen organization for improvement of T-cell–tumor cell contact in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Eva-Maria Kamionka
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Baifeng Qian
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Wolfgang Gross
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Frank Bergmann
- Department of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany;
| | - Thilo Hackert
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Carlo A. Beretta
- CellNetworks Math-Clinic, University of Heidelberg, Bioquant BQ001, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany;
- Institute for Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Nicolas Dross
- Nikon Imaging Center, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Eduard Ryschich
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
- Correspondence: ; Tel.: +49-6221-56-6110; Fax: +49-6221-56-5199
| |
Collapse
|
25
|
Ray SK, Mukherjee S. Consequences of Extracellular Matrix Remodeling in Headway and Metastasis of Cancer along with Novel Immunotherapies: A Great Promise for Future Endeavor. Anticancer Agents Med Chem 2021; 22:1257-1271. [PMID: 34254930 DOI: 10.2174/1871520621666210712090017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/23/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022]
Abstract
Tissues are progressively molded by bidirectional correspondence between denizen cells and extracellular matrix (ECM) via cell-matrix connections along with ECM remodeling. The composition and association of ECM are spatiotemporally directed to control cell conduct and differentiation; however, dysregulation of ECM dynamics prompts the development of diseases, for example, cancer. Emerging information demonstrates that hypoxia may have decisive roles in metastasis. In addition, the sprawling nature of neoplastic cells and chaotic angiogenesis are increasingly influencing microcirculation as well as altering the concentration of oxygen. In various regions of the tumor microenvironment, hypoxia, an essential player in the multistep phase of cancer metastasis, is necessary. Hypoxia can be turned into an advantage for selective cancer therapy because it is much more severe in tumors than in normal tissues. Cellular matrix gives signaling cues that control cell behavior and organize cells' elements in tissue development and homeostasis. The interplay between intrinsic factors of cancer cells themselves, including their genotype and signaling networks, and extrinsic factors of tumor stroma, for example, ECM and ECM remodeling, together decide the destiny and behavior of tumor cells. Tumor matrix encourages the development, endurance, and invasion of neoplastic and immune cell activities to drive metastasis and debilitate treatment. Incipient evidence recommends essential parts of tumor ECM segments and their remodeling in controlling each progression of the cancer-immunity cycle. Scientists have discovered that tumor matrix dynamics as well as matrix remodeling in perspective to anti-tumor immune reactions are especially important for matrix-based biomarkers recognition and followed by immunotherapy and targeting specific drugs.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Department of Applied Sciences, Indira Gandhi Technological and Medical Sciences University, India
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences Bhopal, Madhya pradesh-462020, India
| |
Collapse
|
26
|
Dong Y, Tian J, Yan B, Lv K, Li J, Fu D. Liver-Metastasis-Related Genes are Potential Biomarkers for Predicting the Clinical Outcomes of Patients with Pancreatic Adenocarcinoma. Pathol Oncol Res 2021; 27:1609822. [PMID: 34290570 PMCID: PMC8286999 DOI: 10.3389/pore.2021.1609822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
It is widely acknowledged that metastasis determines the prognosis of pancreatic adenocarcinoma (PAAD), and the liver is the most primary distant metastatic location of PAAD. It is worth exploring the value of liver-metastasis-related genetic prognostic signature (LM-PS) in predicting the clinical outcomes of PAAD patients post R0 resection. We collected 65 tumors and 165 normal pancreatic data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression project (GTEx), respectively. Differentially expressed genes (DEGs) between primary tumor and normal pancreatic samples were intersected with DEGs between primary tumor samples with liver metastasis and those without new tumor events. The intersected 45 genes were input into univariate Cox regression analysis to identify the prognostic genes. Thirty-three prognostic liver-metastasis-related genes were identified and included in least absolute shrinkage and selection operator (LASSO) analysis to develop a seven-gene LM-PS, which included six risk genes (ANO1, FAM83A, GPR87, ITGB6, KLK10, and SERPINE1) and one protective gene (SMIM32). The PAAD patients were grouped into low- and high-risk groups based on the median value of risk scores. The LM-PS harbored an independent predictive ability to distinguish patients with a high-risk of death and liver metastasis after R0 resection. Moreover, a robust prognostic nomogram based on LM-PS, the number of positive lymph nodes, and histologic grade were established to predict the overall survival of PAAD patients. Besides, a transcription factor‐microRNA coregulatory network was constructed for the seven LM-PS genes, and the immune infiltration and genomic alterations were systematically explored in the TGCA-PAAD cohort.
Collapse
Affiliation(s)
- Yinlei Dong
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Junjie Tian
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingqian Yan
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Kun Lv
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Li
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Burn GL, Foti A, Marsman G, Patel DF, Zychlinsky A. The Neutrophil. Immunity 2021; 54:1377-1391. [PMID: 34260886 DOI: 10.1016/j.immuni.2021.06.006] [Citation(s) in RCA: 247] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 12/17/2022]
Abstract
Neutrophils are immune cells with unusual biological features that furnish potent antimicrobial properties. These cells phagocytose and subsequently kill prokaryotic and eukaryotic organisms very efficiently. Importantly, it is not only their ability to attack microbes within a constrained intracellular compartment that endows neutrophils with antimicrobial function. They can unleash their effectors into the extracellular space, where, even post-mortem, their killing machinery can endure and remain functional. The antimicrobial activity of neutrophils must not be misconstrued as being microbe specific and should be viewed more generally as biotoxic. Outside of fighting infections, neutrophils can harness their noxious machinery in other contexts, like cancer. Inappropriate or dysregulated neutrophil activation damages the host and contributes to autoimmune and inflammatory disease. Here we review a number of topics related to neutrophil biology based on contemporary findings.
Collapse
Affiliation(s)
- Garth Lawrence Burn
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Alessandro Foti
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Gerben Marsman
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Dhiren Ferise Patel
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
28
|
Suveges S, Chamseddine I, Rejniak KA, Eftimie R, Trucu D. Collective Cell Migration in a Fibrous Environment: A Hybrid Multiscale Modelling Approach. FRONTIERS IN APPLIED MATHEMATICS AND STATISTICS 2021; 7:680029. [PMID: 34322539 PMCID: PMC8315487 DOI: 10.3389/fams.2021.680029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The specific structure of the extracellular matrix (ECM), and in particular the density and orientation of collagen fibres, plays an important role in the evolution of solid cancers. While many experimental studies discussed the role of ECM in individual and collective cell migration, there are still unanswered questions about the impact of nonlocal cell sensing of other cells on the overall shape of tumour aggregation and its migration type. There are also unanswered questions about the migration and spread of tumour that arises at the boundary between different tissues with different collagen fibre orientations. To address these questions, in this study we develop a hybrid multi-scale model that considers the cells as individual entities and ECM as a continuous field. The numerical simulations obtained through this model match experimental observations, confirming that tumour aggregations are not moving if the ECM fibres are distributed randomly, and they only move when the ECM fibres are highly aligned. Moreover, the stationary tumour aggregations can have circular shapes or irregular shapes (with finger-like protrusions), while the moving tumour aggregations have elongate shapes (resembling to clusters, strands or files). We also show that the cell sensing radius impacts tumour shape only when there is a low ratio of fibre to non-fibre ECM components. Finally, we investigate the impact of different ECM fibre orientations corresponding to different tissues, on the overall tumour invasion of these neighbouring tissues.
Collapse
Affiliation(s)
| | - Ibrahim Chamseddine
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa Florida, USA
| | - Katarzyna A. Rejniak
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa Florida, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa Florida, USA
| | - Raluca Eftimie
- Laboratoire Mathématiques de Besançon, UMR-CNRS 6623, Université de Bourgogne Franche-Comté, 16 Route de Gray, Besançon, France
| | - Dumitru Trucu
- Department of Mathematics, University of Dundee, Dundee, UK
| |
Collapse
|
29
|
Wu Y, Zanotelli MR, Zhang J, Reinhart-King CA. Matrix-driven changes in metabolism support cytoskeletal activity to promote cell migration. Biophys J 2021; 120:1705-1717. [PMID: 33705759 PMCID: PMC8204337 DOI: 10.1016/j.bpj.2021.02.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 01/21/2023] Open
Abstract
The microenvironment provides both active and passive mechanical cues that regulate cell morphology, adhesion, migration, and metabolism. Although the cellular response to those mechanical cues often requires energy-intensive actin cytoskeletal remodeling and actomyosin contractility, it remains unclear how cells dynamically adapt their metabolic activity to altered mechanical cues to support migration. Here, we investigated the changes in cellular metabolic activity in response to different two-dimensional and three-dimensional microenvironmental conditions and how these changes relate to cytoskeletal activity and migration. Utilizing collagen micropatterning on polyacrylamide gels, intracellular energy levels and oxidative phosphorylation were found to be correlated with cell elongation and spreading and necessary for membrane ruffling. To determine whether this relationship holds in more physiological three-dimensional matrices, collagen matrices were used to show that intracellular energy state was also correlated with protrusive activity and increased with matrix density. Pharmacological inhibition of oxidative phosphorylation revealed that cancer cells rely on oxidative phosphorylation to meet the elevated energy requirements for protrusive activity and migration in denser matrices. Together, these findings suggest that mechanical regulation of cytoskeletal activity during spreading and migration by the physical microenvironment is driven by an altered metabolic profile.
Collapse
Affiliation(s)
- Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
30
|
Ratri MC, Brilian AI, Setiawati A, Nguyen HT, Soum V, Shin K. Recent Advances in Regenerative Tissue Fabrication: Tools, Materials, and Microenvironment in Hierarchical Aspects. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Monica Cahyaning Ratri
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
- Department of Chemistry Education Sanata Dharma University Yogyakarta 55281 Indonesia
| | - Albertus Ivan Brilian
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Agustina Setiawati
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
- Department of Life Science Sogang University Seoul 04107 Republic of Korea
- Faculty of Pharmacy Sanata Dharma University Yogyakarta 55281 Indonesia
| | - Huong Thanh Nguyen
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Veasna Soum
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| | - Kwanwoo Shin
- Department of Chemistry and Institute of Biological Interfaces Sogang University Seoul 04107 Republic of Korea
| |
Collapse
|
31
|
Wang WY, Jarman EH, Lin D, Baker BM. Dynamic Endothelial Stalk Cell-Matrix Interactions Regulate Angiogenic Sprout Diameter. Front Bioeng Biotechnol 2021; 9:620128. [PMID: 33869150 PMCID: PMC8044977 DOI: 10.3389/fbioe.2021.620128] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a complex, multicellular process that involves bidirectional interactions between extracellular matrix (ECM) and collectively invading endothelial cell (EC) sprouts that extend the microvasculature during development, wound healing, and disease processes. While many aspects of angiogenesis have been well studied, the relationship between endothelial sprout morphology and subsequent neovessel function remains relatively unknown. Here, we investigated how various soluble and physical matrix cues that regulate endothelial sprouting speed and proliferation correspond to changes in sprout morphology, namely, sprout stalk diameter. We found that sprout stalk cells utilize a combination of cytoskeletal forces and proteolysis to physically compact and degrade the surrounding matrix, thus creating sufficient space in three-dimensional (3D) ECM for lateral expansion. As increasing sprout diameter precedes lumenization to generate perfusable neovessels, this work highlights how dynamic endothelial stalk cell-ECM interactions promote the generation of functional neovessels during sprouting angiogenesis to provide insight into the design of vascularized, implantable biomaterials.
Collapse
Affiliation(s)
| | | | | | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
32
|
Leggett SE, Hruska AM, Guo M, Wong IY. The epithelial-mesenchymal transition and the cytoskeleton in bioengineered systems. Cell Commun Signal 2021; 19:32. [PMID: 33691719 PMCID: PMC7945251 DOI: 10.1186/s12964-021-00713-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/26/2021] [Indexed: 01/04/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is intrinsically linked to alterations of the intracellular cytoskeleton and the extracellular matrix. After EMT, cells acquire an elongated morphology with front/back polarity, which can be attributed to actin-driven protrusion formation as well as the gain of vimentin expression. Consequently, cells can deform and remodel the surrounding matrix in order to facilitate local invasion. In this review, we highlight recent bioengineering approaches to elucidate EMT and functional changes in the cytoskeleton. First, we review transitions between multicellular clusters and dispersed individuals on planar surfaces, which often exhibit coordinated behaviors driven by leader cells and EMT. Second, we consider the functional role of vimentin, which can be probed at subcellular length scales and within confined spaces. Third, we discuss the role of topographical patterning and EMT via a contact guidance like mechanism. Finally, we address how multicellular clusters disorganize and disseminate in 3D matrix. These new technologies enable controlled physical microenvironments and higher-resolution spatiotemporal measurements of EMT at the single cell level. In closing, we consider future directions for the field and outstanding questions regarding EMT and the cytoskeleton for human cancer progression. Video Abstract.
Collapse
Affiliation(s)
- Susan E Leggett
- Department of Chemical and Biological Engineering, Princeton University, William St, Princeton, NJ, 08544, USA
| | - Alex M Hruska
- School of Engineering, Center for Biomedical Engineering, and Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA
| | - Ming Guo
- Department of Mechanical Engineering, MIT, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, and Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA.
| |
Collapse
|
33
|
Aronovich A, Moyal L, Gorovitz B, Amitay-Laish I, Naveh HP, Forer Y, Maron L, Knaneh J, Ad-El D, Yaacobi D, Barel E, Erez N, Hodak E. Cancer-Associated Fibroblasts in Mycosis Fungoides Promote Tumor Cell Migration and Drug Resistance through CXCL12/CXCR4. J Invest Dermatol 2021; 141:619-627.e2. [DOI: 10.1016/j.jid.2020.06.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
|
34
|
Efthymiou G, Radwanska A, Grapa AI, Beghelli-de la Forest Divonne S, Grall D, Schaub S, Hattab M, Pisano S, Poet M, Pisani DF, Counillon L, Descombes X, Blanc-Féraud L, Van Obberghen-Schilling E. Fibronectin Extra Domains tune cellular responses and confer topographically distinct features to fibril networks. J Cell Sci 2021; 134:jcs.252957. [PMID: 33526715 DOI: 10.1242/jcs.252957] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Cellular fibronectin (FN; also known as FN1) variants harboring one or two alternatively spliced so-called extra domains (EDB and EDA) play a central bioregulatory role during development, repair processes and fibrosis. Yet, how the extra domains impact fibrillar assembly and function of the molecule remains unclear. Leveraging a unique biological toolset and image analysis pipeline for direct comparison of the variants, we demonstrate that the presence of one or both extra domains impacts FN assembly, function and physical properties of the matrix. When presented to FN-null fibroblasts, extra domain-containing variants differentially regulate pH homeostasis, survival and TGF-β signaling by tuning the magnitude of cellular responses, rather than triggering independent molecular switches. Numerical analyses of fiber topologies highlight significant differences in variant-specific structural features and provide a first step for the development of a generative model of FN networks to unravel assembly mechanisms and investigate the physical and functional versatility of extracellular matrix landscapes.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Agata Radwanska
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Anca-Ioana Grapa
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France.,Université Côte d'Azur, Inria, CNRS, i3S, Nice 06902, France
| | | | - Dominique Grall
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | | | - Maurice Hattab
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Sabrina Pisano
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | - Mallorie Poet
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
The physical microenvironment of cells plays a fundamental role in regulating cellular behavior and cell fate, especially in the context of cancer metastasis. For example, capillary deformation can destroy arrested circulating tumor cells while the dense extracellular matrix can form a physical barrier for invading cancer cells. Understanding how metastatic cancer cells overcome the challenges brought forth by physical confinement can help in developing better therapeutics that can put a stop to this migratory stage of the metastatic cascade. Numerous in vivo and in vitro assays have been developed to recapitulate the metastatic processes and study cancer cell migration in a confining microenvironment. In this review, we summarize some of the representative techniques and the exciting new findings. We critically review the advantages, as well as challenges associated with these tools and methodologies, and provide a guide on the applications that they are most suited for. We hope future efforts that push forward our current understanding on metastasis under confinement can lead to novel and more effective diagnostic and therapeutic strategies against this dreaded disease.
Collapse
Affiliation(s)
- Kuan Jiang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Lanfeng Liang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
| |
Collapse
|
36
|
Chen H, Li L, He S, Sa G. Podosome formation in the murine palatal mucosae: Its proteolytic role in rete peg formation. Ann Anat 2021; 235:151703. [PMID: 33600951 DOI: 10.1016/j.aanat.2021.151703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Basement membrane remodeling is an indispensable factor for oral mucosal rete peg formation, but how the basement membrane is remodeled remains unclear. Our previous study indicated that keratinocyte growth factor induces the assembly of podosomes, which are dynamic organelles critical for matrix remodeling in human immortalized oral epithelial cells. This study explores podosome formation and its role in basement membrane remodeling during murine oral mucosal rete peg formation. METHODS Perinatal murine palatal tissue slices were obtained from embryonic day 17.5 (E 17.5) to postnatal day 10.5 (P 10.5) BALB/c mice. Rete peg formation was observed by hematoxylin and eosin (HE) staining. Proteolysis of the basement membrane was detected by immunofluorescence staining. The assembly of podosomes and their correlation with basement membrane proteolysis were investigated by laser scanning confocal microscopy. RESULTS The shape of basal layer keratinocytes at the sites of emerging rete pegs changed from typically polygonal to spindle-shaped. Basement membrane proteolysis, indicated by decreased type IV collagen (Col IV) staining, was detected during rete peg formation. Classical markers for podosomes, including cortactin/Tks5, WASP, and matrix metalloproteinase foci, were easily observed at the spindle-shaped cells. Podosomes were visible in regions where there was a significant decrease in Col IV staining. CONCLUSIONS These observations indicated that podosomes form at the front of the emerging rete peg and may play a pivotal role in basement membrane remodeling during rete peg formation.
Collapse
Affiliation(s)
- Heng Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Sangang He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Guoliang Sa
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
37
|
Kim HJ, Kim E, Lee H, Jung JW, Kim JE, Pack CG, Lee JW. SLAC2B-dependent microtubule acetylation regulates extracellular matrix-mediated intracellular TM4SF5 traffic to the plasma membranes. FASEB J 2021; 35:e21369. [PMID: 33554392 DOI: 10.1096/fj.202002138rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 11/11/2022]
Abstract
Transmembrane 4 L six family member 5 (TM4SF5) translocates intracellularly and promotes cell migration, but how subcellular TM4SF5 traffic is regulated to guide cellular migration is unknown. We investigated the influences of the extracellular environment and intracellular signaling on the TM4SF5 traffic with regard to migration directionality. Cell adhesion to fibronectin (FN) but not poly-l-lysine enhanced the traffic velocity and straightness of the TM4SF5WT (but not palmitoylation-deficient mutant TM4SF5 Pal - ) toward the leading edges, depending on tubulin acetylation. Acetylated-microtubules in SLAC2B-positive cells reached mostly the juxtanuclear regions, but reached-out toward the leading edges upon SLAC2B suppression. TM4SF5 expression caused SLAC2B not to be localized at the leading edges. TM4SF5 colocalization with HDAC6 depended on paxillin expression. The trimeric complex consisting of TM4SF5, HDAC6, and SLAC2B might, thus, be enriched at the perinuclear cytosols toward the leading edges. More TM4SF5WT translocation to the leading edges was possible when acetylated-microtubules reached the frontal edges following HDAC6 inhibition by paxillin presumably at new cell-FN adhesions, leading to persistent cell migration. Collectively, this study revealed that cell-FN adhesion and microtubule acetylation could control intracellular traffic of TM4SF5 vesicles to the leading edges via coordinated actions of paxillin, SLAC2B, and HDAC6, leading to TM4SF5-dependent cell migration.
Collapse
Affiliation(s)
- Hye-Jin Kim
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eunmi Kim
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Haesong Lee
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jae Woo Jung
- Interdisciplinary Program in Genetic Engineering, Seoul National University, Seoul, Republic of Korea
| | - Ji Eon Kim
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jung Weon Lee
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea.,Interdisciplinary Program in Genetic Engineering, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
38
|
Belotti Y, McGloin D, Weijer CJ. Effects of spatial confinement on migratory properties of Dictyostelium discoideum cells. Commun Integr Biol 2021; 14:5-14. [PMID: 33552382 PMCID: PMC7849737 DOI: 10.1080/19420889.2021.1872917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Migratory environments of various eukaryotic cells, such as amoeba, leukocytes and cancer cells, typically involve spatial confinement. Numerous studies have recently emerged, aimed to develop experimental platforms that better recapitulate the characteristics of the cellular microenvironment. Using microfluidic technologies, we show that increasing confinement of Dictyostelium discoideum cells into narrower micro-channels resulted in a significant change in the mode of migration and associated arrangement of the actomyosin cytoskeleton. We observed that cells tended to migrate at constant speed, the magnitude of which was dependent on the size of the channels, as was the locomotory strategy adopted by each cell. Two different migration modes were observed, pseudopod-based and bleb-based migration, with bleb based migration being more frequent with increasing confinement and leading to slower migration. Beside the migration mode, we found that the major determinants of cell speed are its protrusion rate, the amount of F-actin at its leading edge and the number of actin foci. Our results highlighted the impact of the microenvironments on cell behavior. Furthermore, we developed a novel quantitative movement analysis platform for mono-dimensional cell migration that allows for standardization and simplification of the experimental conditions and aids investigation of the complex and dynamic processes occurring at the single-cell level.
Collapse
Affiliation(s)
- Yuri Belotti
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| | - David McGloin
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| | | |
Collapse
|
39
|
Gu H, Hu Z, Shi L, Liu X. Assay of extracellular matrix degradation and transmigration of chicken peripheral blood mononuclear cells after infection with genotype VII Newcastle disease virus in vitro. J Virol Methods 2021; 290:114076. [PMID: 33515662 DOI: 10.1016/j.jviromet.2021.114076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 11/04/2020] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
Previous studies showed that, compared to genotype IV Newcastle disease virus (NDV), genotype VII NDV induced extensive extracellular matrix (ECM) degradation by up-regulating the protein expression of matrix metalloproteinase (MMP)-14 in chicken spleens. To investigate potential relationship between MMP-14 function and the ECM degradation, an in vitro peripheral blood mononuclear cells (PBMCs) infection model was established to study the effect of genotype VII NDV (JS5/05) infection on MMP-14 expression, ECM degradation and cell transmigration. The gene and protein expression levels of MMP-14 in NDV-infected chicken PBMCs were measured by quantitative real-time PCR (qRT-PCR) and Western blot, and the subcellular location of MMP-14 was analyzed using immunofluorescence microscopy. A fluorescence-based collagen degradation assay was optimized to measure ECM degradation in PBMCs. Additionally, parameters of a transwell-based transmigration assay were also optimized to determine chemotaxis and transmigration of virus-infected PBMCs. The results showed that JS5/05 up-regulated significantly the expression of MMP-14 in PBMCs at the mRNA and protein levels compared to genotype IV NDV (Herts/33). MMP-14 was transported towards the membrane and accumulated on the cell surface of the JS5/05-infected cells, whereas it remained mainly in the cytoplasm of the Herts/33-infected cells. Collagen degradation assay showed that JS5/05-infected cells exhibited significant collagen degradation compared to the Herts/33-infected cells, and the areas of collagen degradation co-localized with cell surface MMP-14 in the JS5/05-infected cells. The transwell-based transmigration system showed that the transmigration of the JS5/05-infected PBMCs was enhanced significantly compared to the Herts/33-infected cells. These results demonstrated that genotype VII NDV induced up-regulation and surface accumulation of MMP-14 in PBMCs, leading to enhanced ECM degradation and cell migration, and the assays optimized for this study were useful for investigating the regulation of cell behaviour by NDV.
Collapse
Affiliation(s)
- Han Gu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China
| | - Zenglei Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Liwei Shi
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
40
|
Niland S, Eble JA. Hold on or Cut? Integrin- and MMP-Mediated Cell-Matrix Interactions in the Tumor Microenvironment. Int J Mol Sci 2020; 22:ijms22010238. [PMID: 33379400 PMCID: PMC7794804 DOI: 10.3390/ijms22010238] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) has become the focus of interest in cancer research and treatment. It includes the extracellular matrix (ECM) and ECM-modifying enzymes that are secreted by cancer and neighboring cells. The ECM serves both to anchor the tumor cells embedded in it and as a means of communication between the various cellular and non-cellular components of the TME. The cells of the TME modify their surrounding cancer-characteristic ECM. This in turn provides feedback to them via cellular receptors, thereby regulating, together with cytokines and exosomes, differentiation processes as well as tumor progression and spread. Matrix remodeling is accomplished by altering the repertoire of ECM components and by biophysical changes in stiffness and tension caused by ECM-crosslinking and ECM-degrading enzymes, in particular matrix metalloproteinases (MMPs). These can degrade ECM barriers or, by partial proteolysis, release soluble ECM fragments called matrikines, which influence cells inside and outside the TME. This review examines the changes in the ECM of the TME and the interaction between cells and the ECM, with a particular focus on MMPs.
Collapse
|
41
|
Directionality of Macrophages Movement in Tumour Invasion: A Multiscale Moving-Boundary Approach. Bull Math Biol 2020; 82:148. [PMID: 33211193 PMCID: PMC7677171 DOI: 10.1007/s11538-020-00819-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Invasion of the surrounding tissue is one of the recognised hallmarks of cancer (Hanahan and Weinberg in Cell 100: 57–70, 2000. 10.1016/S0092-8674(00)81683-9), which is accomplished through a complex heterotypic multiscale dynamics involving tissue-scale random and directed movement of the population of both cancer cells and other accompanying cells (including here, the family of tumour-associated macrophages) as well as the emerging cell-scale activity of both the matrix-degrading enzymes and the rearrangement of the cell-scale constituents of the extracellular matrix (ECM) fibres. The involved processes include not only the presence of cell proliferation and cell adhesion (to other cells and to the extracellular matrix), but also the secretion of matrix-degrading enzymes. This is as a result of cancer cells as well as macrophages, which are one of the most abundant types of immune cells in the tumour micro-environment. In large tumours, these tumour-associated macrophages (TAMs) have a tumour-promoting phenotype, contributing to tumour proliferation and spread. In this paper, we extend a previous multiscale moving-boundary mathematical model for cancer invasion, by considering also the multiscale effects of TAMs, with special focus on the influence that their directional movement exerts on the overall tumour progression. Numerical investigation of this new model shows the importance of the interactions between pro-tumour TAMs and the fibrous ECM, highlighting the impact of the fibres on the spatial structure of solid tumour.
Collapse
|
42
|
ShcD Binds DOCK4, Promotes Ameboid Motility and Metastasis Dissemination, Predicting Poor Prognosis in Melanoma. Cancers (Basel) 2020; 12:cancers12113366. [PMID: 33202906 PMCID: PMC7696252 DOI: 10.3390/cancers12113366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Metastasis formation and dissemination is a complex process that relies on several steps. Even though highly inefficient, metastasis spreading is the primary cause of cancer morbidity and mortality in patients. The aim of our study was to investigate the molecular pathways leading to metastases making use of human-in-mouse melanoma models of patient-derived xenografts. We demonstrate that the modulation of the expression of an adaptor protein of the Shc family, ShcD, can change the phenotype and the invasive properties of melanoma cells when highly expressed. We also show that ShcD binds DOCK4 and confines it into the cytoplasm, blocking the Rac1 signaling pathways, thus leading to metastasis development. Moreover, our results indicate that melanoma cells are more sensitive to therapeutic treatments when the ShcD molecular pathway is inactivated, suggesting that new therapeutic strategies can be designed in melanomas. Abstract Metastases are the primary cause of cancer-related deaths. The underlying molecular and biological mechanisms remain, however, elusive, thus preventing the design of specific therapies. In melanomas, the metastatic process is influenced by the acquisition of metastasis-associated mutational and epigenetic traits and the activation of metastatic-specific signaling pathways in the primary melanoma. In the current study, we investigated the role of an adaptor protein of the Shc family (ShcD) in the acquisition of metastatic properties by melanoma cells, exploiting our cohort of patient-derived xenografts (PDXs). We provide evidence that the depletion of ShcD expression increases a spread cell shape and the capability of melanoma cells to attach to the extracellular matrix while its overexpression switches their morphology from elongated to rounded on 3D matrices, enhances cells’ invasive phenotype, as observed on collagen gel, and favors metastasis formation in vivo. ShcD overexpression sustains amoeboid movement in melanoma cells, by suppressing the Rac1 signaling pathway through the confinement of DOCK4 in the cytoplasm. Inactivation of the ShcD signaling pathway makes melanoma cells more sensitive to therapeutic treatments. Consistently, ShcD expression predicts poor outcome in a cohort of 183 primary melanoma patients.
Collapse
|
43
|
Hayn A, Fischer T, Mierke CT. Inhomogeneities in 3D Collagen Matrices Impact Matrix Mechanics and Cancer Cell Migration. Front Cell Dev Biol 2020; 8:593879. [PMID: 33251219 PMCID: PMC7674772 DOI: 10.3389/fcell.2020.593879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cell motility under physiological and pathological conditions including malignant progression of cancer and subsequent metastasis are founded on environmental confinements. During the last two decades, three-dimensional cell migration has been studied mostly by utilizing biomimetic extracellular matrix models. In the majority of these studies, the in vitro collagen scaffolds are usually assumed to be homogenous, as they consist commonly of one specific type of collagen, such as collagen type I, isolated from one species. These collagen matrices should resemble in vivo extracellular matrix scaffolds physiologically, however, mechanical phenotype and functional reliability have been addressed poorly due to certain limitations based on the assumption of homogeneity. How local variations of extracellular matrix structure impact matrix mechanics and cell migration is largely unknown. Here, we hypothesize that local inhomogeneities alter cell movement due to alterations in matrix mechanics, as they frequently occur in in vivo tissue scaffolds and were even changed in diseased tissues. To analyze the effect of structural inhomogeneities on cell migration, we used a mixture of rat tail and bovine dermal collagen type I as well as pure rat and pure bovine collagens at four different concentrations to assess three-dimensional scaffold inhomogeneities. Collagen type I from rat self-assembled to elongated fibrils, whereas bovine collagen tended to build node-shaped inhomogeneous scaffolds. We have shown that the elastic modulus determined with atomic force microscopy in combination with pore size analysis using confocal laser scanning microscopy revealed distinct inhomogeneities within collagen matrices. We hypothesized that elastic modulus and pore size govern cancer cell invasion in three-dimensional collagen matrices. In fact, invasiveness of three breast cancer cell types is altered due to matrix-type and concentration indicating that these two factors are crucial for cellular invasiveness. Our findings revealed that local matrix scaffold inhomogeneity is another crucial parameter to explain differences in cell migration, which not solely depended on pore size and stiffness of the collagen matrices. With these three distinct biophysical parameters, characterizing structure and mechanics of the studied collagen matrices, we were able to explain differences in the invasion behavior of the studied cancer cell lines in dependence of the used collagen model.
Collapse
Affiliation(s)
- Alexander Hayn
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| | - Tony Fischer
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| |
Collapse
|
44
|
Suter N, Stebel S, Rianna C, Radmacher M, Brüggemann D. Spatial patterning of nanofibrous collagen scaffolds modulates fibroblast morphology. Biofabrication 2020; 13:015007. [DOI: 10.1088/1758-5090/abb744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
45
|
Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun 2020; 11:5120. [PMID: 33037194 PMCID: PMC7547708 DOI: 10.1038/s41467-020-18794-x] [Citation(s) in RCA: 1016] [Impact Index Per Article: 254.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Tissues are dynamically shaped by bidirectional communication between resident cells and the extracellular matrix (ECM) through cell-matrix interactions and ECM remodelling. Tumours leverage ECM remodelling to create a microenvironment that promotes tumourigenesis and metastasis. In this review, we focus on how tumour and tumour-associated stromal cells deposit, biochemically and biophysically modify, and degrade tumour-associated ECM. These tumour-driven changes support tumour growth, increase migration of tumour cells, and remodel the ECM in distant organs to allow for metastatic progression. A better understanding of the underlying mechanisms of tumourigenic ECM remodelling is crucial for developing therapeutic treatments for patients. Tumors are more than cancer cells — the extracellular matrix is a protein structure that organizes all tissues and is altered in cancer. Here, the authors review recent progress in understanding how the cancer cells and tumor-associated stroma cells remodel the extracellular matrix to drive tumor growth and metastasis.
Collapse
Affiliation(s)
- Juliane Winkler
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA.
| | - Abisola Abisoye-Ogunniyan
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Kevin J Metcalf
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Zena Werb
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
46
|
Calabrese L, Bizzoca ME, Grigolato R, Maffini FA, Tagliabue M, Negro R, Leuci S, Mignogna MD, Lo Muzio L. From Bench to Bedside in Tongue Muscle Cancer Invasion and Back again: Gross Anatomy, Microanatomy, Surgical Treatments and Basic Research. Life (Basel) 2020; 10:life10090197. [PMID: 32932638 PMCID: PMC7554763 DOI: 10.3390/life10090197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Tongue squamous cell carcinoma is the most common malignancy in the oral cavity. Despite advances in diagnosis and treatment, the prognosis of advanced states has not significantly improved. Depth of invasion, pattern of invasion such as tumor budding grade, lingual lymph node metastasis in early stages, collective cell migration and circulating tumor cells in peripheral blood are some examples of the mechanisms that are currently receiving increasing attention in the evaluation of the prognosis of tongue cancers. Anatomic-based surgery showed that it is possible to improve loco-regional control of tongue cancer. In patients with a "T-N tract involvement", there is significantly more distant recurrence (40%) in patients undergoing a compartmental tongue surgery. In general, the neoplastic infiltration of the lingual muscles is traced back to the finding of neoplastic tissue along the course of a muscle; however, the muscle fibers, due to their spatial conformation and the organization of the extracellular matrix, could influence the movement of tumor cells through the muscle, leaving its three-dimensional structure unchanged. We need to exclude the possibility that tongue muscle fibers represent a mechanism for the diffusion of cancer cells without muscle invasion.
Collapse
Affiliation(s)
- Luca Calabrese
- Division of Otorhinolaryngology, San Maurizio Hospital, 39100 Bolzano, Italy;
| | - Maria Eleonora Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Roberto Grigolato
- Division of Prevention, San Maurizio Hospital, 13060 Bolzano, Italy;
| | | | - Marta Tagliabue
- Division of Otolaryngology Head & Neck Surgery, European Institute of Oncology IRCCS, 20132 Milan, Italy;
| | - Rosa Negro
- Division of Pathology, San Maurizio Hospital, 39100 Bolzano, Italy;
| | - Stefania Leuci
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (M.D.M.)
| | - Michele Davide Mignogna
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Oral Medicine Unit, Federico II University of Naples, 80138 Naples, Italy; (S.L.); (M.D.M.)
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
- C.I.N.B.O. (Consorzio Interuniversitario Nazionale per la Bio-Oncologia), 66100 Chieti, Italy
- Correspondence: ; Tel.: +39-0881-588090
| |
Collapse
|
47
|
Feng L, Hu YL, Ma P, Feng Y, Guo YB, Huang H, Li P, Mao QS, Xue WJ. Decellularized gastric matrix as a mesh for gastric perforation repair. J Biomed Mater Res B Appl Biomater 2020; 109:451-462. [PMID: 32841467 DOI: 10.1002/jbm.b.34713] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/16/2020] [Accepted: 08/12/2020] [Indexed: 11/10/2022]
Abstract
The development of novel materials with effective defect-repairing properties will help avoid subtotal gastrectomy in patients with large gastric perforations. We prepared perfused decellularized gastric matrix (PDGM) and analyzed its components, spatial structure, biomechanics, cytotoxicity, and histocompatibility to validate its efficacy in the repair of gastric perforation. PDGM retained large amounts of gastric extracellular matrix, while residual glandular cells and muscle fibers were not found. The spatial structure of the tissue was well preserved, while the DNA and glycosaminoglycan contents were significantly decreased compared with normal gastric tissue (p < .01). There was no obvious deformation of the spatial structure and tissue elasticity of PDGM after sterilization by Cobalt-60 irradiation. The PDGM had good histocompatibility. PDGM was then used to repair a rat gastric perforation model. Radiography of the upper gastrointestinal tract at 24 hr postoperatively revealed no contrast agent leakage. There was evidence of early fibroblast proliferation, which was complicated by capillary regeneration. The hyperplastic gastric gland was slightly disarranged after repair. Defects of the muscular layer also healed a little with the regeneration process. PDGM is a nontoxic biocompatible biological mesh that may be useful for repairing relatively large gastric defects.
Collapse
Affiliation(s)
- Liang Feng
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China.,Department of Burns and Plastic Surgery, Nantong Third People's Hospital, Nantong University, Nantong, Jiangsu, China
| | - Yi-Lin Hu
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China.,Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Peng Ma
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Ying Feng
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Yi-Bing Guo
- Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Hua Huang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Peng Li
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Qin-Sheng Mao
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Wan-Jiang Xue
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China.,Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| |
Collapse
|
48
|
Campbell EJ, Bagchi P. A computational study of amoeboid motility in 3D: the role of extracellular matrix geometry, cell deformability, and cell-matrix adhesion. Biomech Model Mechanobiol 2020; 20:167-191. [PMID: 32772275 DOI: 10.1007/s10237-020-01376-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/01/2020] [Indexed: 12/24/2022]
Abstract
Amoeboid cells often migrate using pseudopods, which are membrane protrusions that grow, bifurcate, and retract dynamically, resulting in a net cell displacement. Many cells within the human body, such as immune cells, epithelial cells, and even metastatic cancer cells, can migrate using the amoeboid phenotype. Amoeboid motility is a complex and multiscale process, where cell deformation, biochemistry, and cytosolic and extracellular fluid motions are coupled. Furthermore, the extracellular matrix (ECM) provides a confined, complex, and heterogeneous environment for the cells to navigate through. Amoeboid cells can migrate without significantly remodeling the ECM using weak or no adhesion, instead utilizing their deformability and the microstructure of the ECM to gain enough traction. While a large volume of work exists on cell motility on 2D substrates, amoeboid motility is 3D in nature. Despite recent progress in modeling cellular motility in 3D, there is a lack of systematic evaluations of the role of ECM microstructure, cell deformability, and adhesion on 3D motility. To fill this knowledge gap, here we present a multiscale, multiphysics modeling study of amoeboid motility through 3D-idealized ECM. The model is a coupled fluid‒structure and coarse-grain biochemistry interaction model that accounts for large deformation of cells, pseudopod dynamics, cytoplasmic and extracellular fluid motion, stochastic dynamics of cell-ECM adhesion, and microstructural (pore-scale) geometric details of the ECM. The key finding of the study is that cell deformation and matrix porosity strongly influence amoeboid motility, while weak adhesion and microscale structural details of the ECM have secondary but subtle effects.
Collapse
Affiliation(s)
- Eric J Campbell
- Mechanical and Aerospace Engineering Department, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Prosenjit Bagchi
- Mechanical and Aerospace Engineering Department, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
49
|
Poisson L, Lopez-Charcas O, Chadet S, Bon E, Lemoine R, Brisson L, Ouaissi M, Baron C, Besson P, Roger S, Moussata D. Rock inhibition promotes Na V1.5 sodium channel-dependent SW620 colon cancer cell invasiveness. Sci Rep 2020; 10:13350. [PMID: 32770034 PMCID: PMC7414216 DOI: 10.1038/s41598-020-70378-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022] Open
Abstract
The acquisition of invasive capacities by carcinoma cells, i.e. their ability to migrate through and to remodel extracellular matrices, is a determinant process leading to their dissemination and to the development of metastases. these cancer cell properties have often been associated with an increased Rho-ROCK signalling, and ROCK inhibitors have been proposed for anticancer therapies. In this study we used the selective ROCK inhibitor, Y-27632, to address the participation of the Rho-ROCK signalling pathway in the invasive properties of SW620 human colon cancer cells. Contrarily to initial assumptions, Y-27632 induced the acquisition of a pro-migratory cell phenotype and increased cancer cell invasiveness in both 3- and 2-dimensions assays. This effect was also obtained using the other ROCK inhibitor Fasudil as well as with knocking down the expression of ROCK-1 or ROCK-2, but was prevented by the inhibition of NaV1.5 voltage-gated sodium channel activity. Indeed, ROCK inhibition enhanced the activity of the pro-invasive NaV1.5 channel through a pathway that was independent of gene expression regulation. In conclusions, our evidence identifies voltage-gated sodium channels as new targets of the ROCK signalling pathway, as well as responsible for possible deleterious effects of the use of ROCK inhibitors in the treatment of cancers.
Collapse
Affiliation(s)
- Lucile Poisson
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France.,Inserm UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Osbaldo Lopez-Charcas
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Stéphanie Chadet
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Emeline Bon
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Roxane Lemoine
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Lucie Brisson
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Mehdi Ouaissi
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France.,CHRU de Tours, Tours, France
| | - Christophe Baron
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France.,CHRU de Tours, Tours, France
| | - Pierre Besson
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Sébastien Roger
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France. .,Institut Universitaire de France, Paris, France.
| | - Driffa Moussata
- EA4245 Transplantation, Immunologie, Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France.,CHRU de Tours, Tours, France
| |
Collapse
|
50
|
Ruud KF, Hiscox WC, Yu I, Chen RK, Li W. Distinct phenotypes of cancer cells on tissue matrix gel. Breast Cancer Res 2020; 22:82. [PMID: 32736579 PMCID: PMC7395363 DOI: 10.1186/s13058-020-01321-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/23/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Breast cancer cells invading the connective tissues outside the mammary lobule or duct immerse in a reservoir of extracellular matrix (ECM) that is structurally and biochemically distinct from that of their site of origin. The ECM is a spatial network of matrix proteins, which not only provide physical support but also serve as bioactive ligands to the cells. It becomes evident that the dimensional, mechanical, structural, and biochemical properties of ECM are all essential mediators of many cellular functions. To better understand breast cancer development and cancer cell biology in native tissue environment, various tissue-mimicking culture models such as hydrogel have been developed. Collagen I (Col I) and Matrigel are the most common hydrogels used in cancer research and have opened opportunities for addressing biological questions beyond the two-dimensional (2D) cell cultures. Yet, it remains unclear whether these broadly used hydrogels can recapitulate the environmental properties of tissue ECM, and whether breast cancer cells grown on CoI I or Matrigel display similar phenotypes as they would on their native ECM. METHODS We investigated mammary epithelial cell phenotypes and metabolic profiles on animal breast ECM-derived tissue matrix gel (TMG), Col I, and Matrigel. Atomic force microscopy (AFM), fluorescence microscopy, acini formation assay, differentiation experiments, spatial migration/invasion assays, proliferation assay, and nuclear magnetic resonance (NMR) spectroscopy were used to examine biological phenotypes and metabolic changes. Student's t test was applied for statistical analyses. RESULTS Our data showed that under a similar physiological stiffness, the three types of hydrogels exhibited distinct microstructures. Breast cancer cells grown on TMG displayed quite different morphologies, surface receptor expression, differentiation status, migration and invasion, and metabolic profiles compared to those cultured on Col I and Matrigel. Depleting lactate produced by glycolytic metabolism of cancer cells abolished the cell proliferation promoted by the non-tissue-specific hydrogel. CONCLUSION The full ECM protein-based hydrogel system may serve as a biologically relevant model system to study tissue- and disease-specific pathological questions. This work provides insights into tissue matrix regulation of cancer cell biomarker expression and identification of novel therapeutic targets for the treatment of human cancers based on tissue-specific disease modeling.
Collapse
Affiliation(s)
- Kelsey F Ruud
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - William C Hiscox
- Center for NMR Spectroscopy, Washington State University, Pullman, WA, 99164, USA
| | - Ilhan Yu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Roland K Chen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|