1
|
Sapudom J, Riedl P, Schricker M, Kroy K, Pompe T. Physical network regimes of 3D fibrillar collagen networks trigger invasive phenotypes of breast cancer cells. BIOMATERIALS ADVANCES 2024; 163:213961. [PMID: 39032434 DOI: 10.1016/j.bioadv.2024.213961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
The mechanical characteristics of the extracellular environment are known to significantly influence cancer cell behavior in vivo and in vitro. The structural complexity and viscoelastic dynamics of the extracellular matrix (ECM) pose significant challenges in understanding its impact on cancer cells. Herein, we report distinct regulatory signatures in the invasion of different breast cancer cell lines into three-dimensional (3D) fibrillar collagen networks, caused by systematic modifications of the physical network properties. By reconstituting collagen networks of thin fibrils, we demonstrate that such networks can display network strand flexibility akin to that of synthetic polymer networks, known to exhibit entropic rubber elasticity. This finding contrasts with the predominant description of the mechanics of fibrillar collagen networks by an enthalpic bending elasticity of rod-like fibrils. Mean-squared displacement analysis of free-standing fibrils confirmed a flexible fiber regime in networks of thin fibrils. Furthermore, collagen fibrils in both networks were softened by the adsorption of highly negatively charged sulfonated polymers and colloidal probe force measurements of network elastic modulus again proofed the occurrence of the two different physical network regimes. Our cell assays revealed that the cellular behavior (morphology, clustering, invasiveness, matrix metalloproteinase (MMP) activity) of the 'weakly invasive' MCF-7 and 'highly invasive' MDA-MB-231 breast cancer cell lines is distinctively affected by the physical (enthalpic/entropic) network regime, and cannot be explained by changes of the network elastic modulus, alone. These results highlight an essential pathway, albeit frequently overlooked, how the physical characteristics of fibrillar ECMs affect cellular behavior. Considering the coexistence of diverse physical network regimes of the ECM in vivo, our findings underscore their critical role of ECM's physical network regimes in tumor progression and other cell functions, and moreover emphasize the significance of 3D in vitro collagen network models for quantifying cell responses in both healthy and pathological states.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany; Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Philipp Riedl
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany
| | - Maria Schricker
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany
| | - Klaus Kroy
- Institute for Theoretical Physics, Leipzig University, Leipzig 04009, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany.
| |
Collapse
|
2
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
3
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
4
|
Qiu S, Qiu Y, Deng L, Nie L, Ge L, Zheng X, Jin D, Jin K, Zhou X, Su X, Cai B, Li J, Tu X, Gong L, Liu L, Liu Z, Bao Y, Ai J, Lin T, Yang L, Wei Q. Cell softness reveals tumorigenic potential via ITGB8/AKT/glycolysis signaling in a mice model of orthotopic bladder cancer. Chin Med J (Engl) 2024; 137:209-221. [PMID: 37390491 PMCID: PMC10798691 DOI: 10.1097/cm9.0000000000002710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Bladder cancer, characterized by a high potential of tumor recurrence, has high lifelong monitoring and treatment costs. To date, tumor cells with intrinsic softness have been identified to function as cancer stem cells in several cancer types. Nonetheless, the existence of soft tumor cells in bladder tumors remains elusive. Thus, our study aimed to develop a micro-barrier microfluidic chip to efficiently isolate deformable tumor cells from distinct types of bladder cancer cells. METHODS The stiffness of bladder cancer cells was determined by atomic force microscopy (AFM). The modified microfluidic chip was utilized to separate soft cells, and the 3D Matrigel culture system was to maintain the softness of tumor cells. Expression patterns of integrin β8 (ITGB8), protein kinase B (AKT), and mammalian target of rapamycin (mTOR) were determined by Western blotting. Double immunostaining was conducted to examine the interaction between F-actin and tripartite motif containing 59 (TRIM59). The stem-cell-like characteristics of soft cells were explored by colony formation assay and in vivo studies upon xenografted tumor models. RESULTS Using our newly designed microfluidic approach, we identified a small fraction of soft tumor cells in bladder cancer cells. More importantly, the existence of soft tumor cells was confirmed in clinical human bladder cancer specimens, in which the number of soft tumor cells was associated with tumor relapse. Furthermore, we demonstrated that the biomechanical stimuli arising from 3D Matrigel activated the F-actin/ITGB8/TRIM59/AKT/mTOR/glycolysis pathways to enhance the softness and tumorigenic capacity of tumor cells. Simultaneously, we detected a remarkable up-regulation in ITGB8, TRIM59, and phospho-AKT in clinical bladder recurrent tumors compared with their non-recurrent counterparts. CONCLUSIONS The ITGB8/TRIM59/AKT/mTOR/glycolysis axis plays a crucial role in modulating tumor softness and stemness. Meanwhile, the soft tumor cells become more sensitive to chemotherapy after stiffening, that offers new insights for hampering tumor progression and recurrence.
Collapse
Affiliation(s)
- Shi Qiu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona 6500, Switzerland
| | - Yaqi Qiu
- Department of Science and Drug Technology, University of Turin, Turin, Italy
| | - Linghui Deng
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liming Ge
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610000, China
| | - Xiaonan Zheng
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Di Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Jin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianghong Zhou
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyang Su
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Boyu Cai
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiakun Li
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiang Tu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Lina Gong
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yige Bao
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, National Clinical Research Center for Geriatrics and Center of Biomedical Big Data, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
5
|
Roman J. Fibroblasts-Warriors at the Intersection of Wound Healing and Disrepair. Biomolecules 2023; 13:945. [PMID: 37371525 DOI: 10.3390/biom13060945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/07/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023] Open
Abstract
Wound healing is triggered by inflammation elicited after tissue injury. Mesenchymal cells, specifically fibroblasts, accumulate in the injured tissues, where they engage in tissue repair through the expression and assembly of extracellular matrices that provide a scaffold for cell adhesion, the re-epithelialization of tissues, the production of soluble bioactive mediators that promote cellular recruitment and differentiation, and the regulation of immune responses. If appropriately deployed, these processes promote adaptive repair, resulting in the preservation of the tissue structure and function. Conversely, the dysregulation of these processes leads to maladaptive repair or disrepair, which causes tissue destruction and a loss of organ function. Thus, fibroblasts not only serve as structural cells that maintain tissue integrity, but are key effector cells in the process of wound healing. The review will discuss the general concepts about the origins and heterogeneity of this cell population and highlight the specific fibroblast functions disrupted in human disease. Finally, the review will explore the role of fibroblasts in tissue disrepair, with special attention to the lung, the role of aging, and how alterations in the fibroblast phenotype underpin disorders characterized by pulmonary fibrosis.
Collapse
Affiliation(s)
- Jesse Roman
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care and The Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
6
|
Nakamura R, Bing R, Gartling GJ, Branski RC. Macrophages alter inflammatory and fibrotic gene expression in human vocal fold fibroblasts. Exp Cell Res 2022; 419:113301. [PMID: 35931141 DOI: 10.1016/j.yexcr.2022.113301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/05/2022] [Accepted: 07/26/2022] [Indexed: 11/04/2022]
Abstract
Macrophage phenotypes are simplistically classified as pro-inflammatory (M1) or anti-inflammatory/pro-fibrotic (M2). Phenotypically different macrophages are putatively involved in vocal fold (VF) fibrosis. The current study investigated interactions between macrophages and VF fibroblasts. THP-1 monocyte-derived macrophages were treated with interferon-gamma (IFN-γ), lipopolysaccharide (LPS)/IFN-γ, interleukin-10 (IL10), transforming growth factor-β1 (TGF-β), or interleukin-4 (IL4) for 24 h (M(IFN), M(IFN/LPS), M(IL10), M(TGF), and M(IL4), respectively; M(-) denotes untreated macrophages). Differentially activated macrophages and human VF fibroblasts were co-cultured ± direct contact. Expression of CXCL10, CCN2, ACTA2, FN1, TGM2, and LOX was quantified by real-time polymerase chain reaction. Type I collagen and smooth muscle actin (SMA) were observed by immunofluorescence. CXCL10 and PTGS2 were upregulated in fibroblasts indirectly co-cultured with M(IFN) and M(IFN/LPS). M(TGF) stimulated CCN2, ACTA2, and FN1 in fibroblasts. Enzymes involved in extracellular matrix crosslinking (TGM2, LOX) were increased in monocultured M(IL4) compared to M(-). Direct co-culture with all macrophages increased type I collagen and SMA in fibroblasts. Macrophage phenotypic shift was consistent with stimulation and had downstream differential effects on VF fibroblasts. Direct contact with macrophages, regardless of phenotype, stimulated a pro-fibrotic response in VF fibroblasts. Collectively, these data suggest meaningful interactions between macrophages and fibroblasts mediate fibrosis.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Renjie Bing
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Gary J Gartling
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Ryan C Branski
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA; Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Alsisi A, Eftimie R, Trucu D. Nonlocal multiscale modelling of tumour-oncolytic viruses interactions within a heterogeneous fibrous/non-fibrous extracellular matrix. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:6157-6185. [PMID: 35603396 DOI: 10.3934/mbe.2022288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this study we investigate computationally tumour-oncolytic virus (OV) interactions that take place within a heterogeneous extracellular matrix (ECM). The ECM is viewed as a mixture of two constitutive phases, namely a fibre phase and a non-fibre phase. The multiscale mathematical model presented here focuses on the nonlocal cell-cell and cell-ECM interactions, and how these interactions might be impacted by the infection of cancer cells with the OV. At macroscale we track the kinetics of cancer cells, virus particles and the ECM. At microscale we track (i) the degradation of ECM by matrix degrading enzymes (MDEs) produced by cancer cells, which further influences the movement of tumour boundary; (ii) the re-arrangement of the microfibres that influences the re-arrangement of macrofibres (i.e., fibres at macroscale). With the help of this new multiscale model, we investigate two questions: (i) whether the infected cancer cell fluxes are the result of local or non-local advection in response to ECM density; and (ii) what is the effect of ECM fibres on the the spatial spread of oncolytic viruses and the outcome of oncolytic virotherapy.
Collapse
Affiliation(s)
- Abdulhamed Alsisi
- Division of Mathematics, University of Dundee, Dundee DD1 4HN, United Kingdom
| | - Raluca Eftimie
- Laboratoire Mathematiques de Besançon, UMR-CNRS 6623, Université de Bourgogne Franche-Comté, 16 Route de Gray, Besançon, France
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee DD1 4HN, United Kingdom
| |
Collapse
|
8
|
Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol 2022; 15:34. [PMID: 35331296 PMCID: PMC8943941 DOI: 10.1186/s13045-022-01252-0] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer microenvironment is critical for tumorigenesis and cancer progression. The extracellular matrix (ECM) interacts with tumor and stromal cells to promote cancer cells proliferation, migration, invasion, angiogenesis and immune evasion. Both ECM itself and ECM stiffening-induced mechanical stimuli may activate cell membrane receptors and mechanosensors such as integrin, Piezo1 and TRPV4, thereby modulating the malignant phenotype of tumor and stromal cells. A better understanding of how ECM stiffness regulates tumor progression will contribute to the development of new therapeutics. The rapidly expanding evidence in this research area suggests that the regulators and effectors of ECM stiffness represent potential therapeutic targets for cancer. This review summarizes recent work on the regulation of ECM stiffness in cancer, the effects of ECM stiffness on tumor progression, cancer immunity and drug resistance. We also discuss the potential targets that may be druggable to intervene ECM stiffness and tumor progression. Based on these advances, future efforts can be made to develop more effective and safe drugs to interrupt ECM stiffness-induced oncogenic signaling, cancer progression and drug resistance.
Collapse
|
9
|
Alsisi A, Eftimie R, Trucu D. Non-local multiscale approach for the impact of go or grow hypothesis on tumour-viruses interactions. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:5252-5284. [PMID: 34517487 DOI: 10.3934/mbe.2021267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We propose and study computationally a novel non-local multiscale moving boundary mathematical model for tumour and oncolytic virus (OV) interactions when we consider the go or grow hypothesis for cancer dynamics. This spatio-temporal model focuses on two cancer cell phenotypes that can be infected with the OV or remain uninfected, and which can either move in response to the extracellular-matrix (ECM) density or proliferate. The interactions between cancer cells, those among cancer cells and ECM, and those among cells and OV occur at the macroscale. At the micro-scale, we focus on the interactions between cells and matrix degrading enzymes (MDEs) that impact the movement of tumour boundary. With the help of this multiscale model we explore the impact on tumour invasion patterns of two different assumptions that we consider in regard to cell-cell and cell-matrix interactions. In particular we investigate model dynamics when we assume that cancer cell fluxes are the result of local advection in response to the density of extracellular matrix (ECM), or of non-local advection in response to cell-ECM adhesion. We also investigate the role of the transition rates between mainly-moving and mainly-growing cancer cell sub-populations, as well as the role of virus infection rate and virus replication rate on the overall tumour dynamics.
Collapse
Affiliation(s)
- Abdulhamed Alsisi
- Division of Mathematics, University of Dundee, Dundee DD1 4HN, United Kingdom
| | - Raluca Eftimie
- Laboratoire Mathematiques de Besançon, UMR-CNRS 6623, Université de Bourgogne Franche-Comté, 16 Route de Gray, Besançon, France
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee DD1 4HN, United Kingdom
| |
Collapse
|
10
|
Ji C, McCulloch CA. TRPV4 integrates matrix mechanosensing with Ca 2+ signaling to regulate extracellular matrix remodeling. FEBS J 2020; 288:5867-5887. [PMID: 33300268 DOI: 10.1111/febs.15665] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/23/2020] [Indexed: 12/23/2022]
Abstract
In healthy connective tissues, mechanosensors trigger the generation of Ca2+ signals, which enable cells to maintain the structure of the fibrillar collagen matrix through actomyosin contractile forces. Transient receptor potential vanilloid type 4 (TRPV4) is a mechanosensitive Ca2+ -permeable channel that, when expressed in cell-matrix adhesions of the plasma membrane, regulates extracellular matrix (ECM) remodeling. In high prevalence disorders such as fibrosis and tumor metastasis, dysregulated matrix remodeling is associated with disruptions of Ca2+ homeostasis and TRPV4 function. Here, we consider that ECM polymers transmit cell-activating mechanical signals to TRPV4 in cell adhesions. When activated, TRPV4 regulates fibrillar collagen remodeling, thereby altering the mechanical properties of the ECM. In this review, we integrate functionally connected processes of matrix remodeling to highlight how TRPV4 in cell adhesions and matrix mechanics are reciprocally regulated through Ca2+ signaling.
Collapse
Affiliation(s)
- Chenfan Ji
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, ON, Canada
| | | |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Abstract
Vascular smooth muscle cells (VSMCs) shift from a physiological contractile phenotype to an adverse proliferative or synthetic state, which is a major event leading to aortic disease. VSMCs are exposed to multiple mechanical signals from their microenvironment including vascular extracellular matrix (ECM) stiffness and stretch which regulate VSMC contraction. How ECM stiffness regulates the function and phenotype of VSMCs is not well understood. In this study, we introduce in vitro and in vivo models to evaluate the impact of ECM stiffnesses on VSMC function. Through unbiased transcriptome sequencing analysis, we detected upregulation of synthetic phenotype-related genes including osteopontin, matrix metalloproteinases, and inflammatory cytokines in VSMCs cultured using soft matrix hydrogels in vitro, suggesting VSMC dedifferentiation toward a synthetic phenotype upon ECM softening. For the in vivo model, the lysyl oxidase inhibitor β-aminopropionitrile monofumarate (BAPN) was administrated to disrupt the cross-linking of collagen to induce ECM softening. Consistently, decreased ECM stiffnesses promoted VSMC phenotypic switching to a synthetic phenotype as evidenced by upregulation of synthetic phenotype-related genes in the aortas of mice following BAPN treatment. Finally, BAPN-treated mice showed severe expansion and developed aortic dissection. Our study reveals the pivotal role of ECM softening in regulating the VSMC phenotype switch and provides a potential target for treating VSMC dysfunction and aortic dissection disease.
Collapse
|
14
|
Shuttleworth R, Trucu D. Cell-Scale Degradation of Peritumoural Extracellular Matrix Fibre Network and Its Role Within Tissue-Scale Cancer Invasion. Bull Math Biol 2020; 82:65. [PMID: 32458057 PMCID: PMC7250813 DOI: 10.1007/s11538-020-00732-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
Abstract
Local cancer invasion of tissue is a complex, multiscale process which plays an essential role in tumour progression. During the complex interaction between cancer cell population and the extracellular matrix (ECM), of key importance is the role played by both bulk two-scale dynamics of ECM fibres within collective movement of the tumour cells and the multiscale leading edge dynamics driven by proteolytic activity of the matrix-degrading enzymes (MDEs) that are secreted by the cancer cells. As these two multiscale subsystems share and contribute to the same tumour macro-dynamics, in this work we develop further the model introduced in Shuttleworth and Trucu (Bull Math Biol 81:2176–2219, 2019. 10.1007/s11538-019-00598-w) by exploring a new aspect of their interaction that occurs at the cell scale. Specifically, here we will focus on understanding the cell-scale cross talk between the micro-scale parts of these two multiscale subsystems which get to interact directly in the peritumoural region, with immediate consequences both for MDE micro-dynamics occurring at the leading edge of the tumour and for the cell-scale rearrangement of the naturally oriented ECM fibres in the peritumoural region, ultimately influencing the way tumour progresses in the surrounding tissue. To that end, we will propose a new modelling that captures the ECM fibres degradation not only at macro-scale in the bulk of the tumour but also explicitly in the micro-scale neighbourhood of the tumour interface as a consequence of the interactions with molecular fluxes of MDEs that exercise their spatial dynamics at the invasive edge of the tumour.
Collapse
Affiliation(s)
- Robyn Shuttleworth
- Division of Mathematics, University of Dundee, Dundee, DD1 4HN Scotland, UK
| | - Dumitru Trucu
- Division of Mathematics, University of Dundee, Dundee, DD1 4HN Scotland, UK
| |
Collapse
|
15
|
Dean T, Li NT, Cadavid JL, Ailles L, McGuigan AP. A TRACER culture invasion assay to probe the impact of cancer associated fibroblasts on head and neck squamous cell carcinoma cell invasiveness. Biomater Sci 2020; 8:3078-3094. [PMID: 32347842 DOI: 10.1039/c9bm02017a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer associated fibroblasts (CAFs) are a major cellular component of the tumour stroma and have been shown to promote tumour cell invasion and disease progression. CAF-cancer cell interactions are bi-directional and occur via both soluble factor dependent and extracellular matrix (ECM) remodelling mechanisms, which are incompletely understood. Previously we developed the Tissue Roll for Analysis of Cellular Environment and Response (TRACER), a novel stacked paper tumour model in which cells embedded in a hydrogel are infiltrated into a porous cellulose scaffold that is then rolled around an aluminum core to generate a multi-layered 3D tissue. Here, we use the TRACER platform to explore the impact of CAFs derived from three different patients on the invasion of two head and neck squamous cell carcinoma (HNSCC) cell lines (CAL33 and FaDu). We find that co-culture with CAFs enhances HNSCC tumour cell invasion into an acellular collagen layer in TRACER and this enhanced migration occurs independently of proliferation. We show that CAF-enhanced invasion of CAL33 cells is driven by a soluble factor independent mechanism, likely involving CAF mediated ECM remodelling via matrix metalloprotenases (MMPs). Furthermore, we find that CAF-enhanced tumour cell invasion is dependent on the spatial pattern of collagen density within the culture. Our results highlight the utility of the co-culture TRACER platform to explore soluble factor independent interactions between CAFs and tumour cells that drive increased tumour cell invasion.
Collapse
Affiliation(s)
- Teresa Dean
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada.
| | | | | | | | | |
Collapse
|
16
|
Shuttleworth R, Trucu D. Multiscale dynamics of a heterotypic cancer cell population within a fibrous extracellular matrix. J Theor Biol 2019; 486:110040. [PMID: 31604075 DOI: 10.1016/j.jtbi.2019.110040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/27/2019] [Accepted: 10/07/2019] [Indexed: 11/28/2022]
Abstract
Local cancer cell invasion is a complex process involving many cellular and tissue interactions and is an important prerequisite for metastatic spread, the main cause of cancer related deaths. As a tumour increases in malignancy, the cancer cells adopt the ability to mutate into secondary cell subpopulations giving rise to a heterogeneous tumour. This new cell subpopulation often carries higher invasive abilities and permits a quicker spread of the tumour. Building upon the recent multiscale modelling framework for cancer invasion within a fibrous ECM introduced in Shuttleworth and Trucu, (2019), in this paper we consider the process of local invasion by a heterotypic tumour consisting of two cancer cell populations mixed with a two-phase ECM. To that end, we address the double feedback link between the tissue-scale cancer dynamics and the cell-scale molecular processes through the development of a two-part modelling framework that crucially incorporates the multiscale dynamic redistribution of oriented fibres occurring within a two-phase extra-cellular matrix and combines this with the multiscale leading edge dynamics exploring key matrix-degrading enzymes molecular processes along the tumour interface that drive the movement of the cancer boundary. The modelling framework will be accompanied by computational results that explore the effects of the underlying fibre network on the overall pattern of cancer invasion.
Collapse
Affiliation(s)
| | - Dumitru Trucu
- University of Dundee, Dundee, Scotland DD1 4HN, United Kingdom.
| |
Collapse
|
17
|
Taubenberger AV, Girardo S, Träber N, Fischer-Friedrich E, Kräter M, Wagner K, Kurth T, Richter I, Haller B, Binner M, Hahn D, Freudenberg U, Werner C, Guck J. 3D Microenvironment Stiffness Regulates Tumor Spheroid Growth and Mechanics via p21 and ROCK. ACTA ACUST UNITED AC 2019; 3:e1900128. [PMID: 32648654 DOI: 10.1002/adbi.201900128] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 01/01/2023]
Abstract
The mechanical properties of cancer cells and their microenvironment contribute to breast cancer progression. While mechanosensing has been extensively studied using 2D substrates, much less is known about it in a physiologically more relevant 3D context. Here it is demonstrated that breast cancer tumor spheroids, growing in 3D polyethylene glycol-heparin hydrogels, are sensitive to their environment stiffness. During tumor spheroid growth, compressive stresses of up to 2 kPa build up, as quantitated using elastic polymer beads as stress sensors. Atomic force microscopy reveals that tumor spheroid stiffness increases with hydrogel stiffness. Also, constituent cell stiffness increases in a Rho associated kinase (ROCK)- and F-actin-dependent manner. Increased hydrogel stiffness correlated with attenuated tumor spheroid growth, a higher proportion of cells in G0/G1 phase, and elevated levels of the cyclin-dependent kinase inhibitor p21. Drug-mediated ROCK inhibition not only reverses cell stiffening upon culture in stiff hydrogels but also increases tumor spheroid growth. Taken together, a mechanism by which the growth of a tumor spheroid can be regulated via cytoskeleton rearrangements in response to its mechanoenvironment is revealed here. Thus, the findings contribute to a better understanding of how cancer cells react to compressive stress when growing under confinement in stiff environments.
Collapse
Affiliation(s)
- Anna V Taubenberger
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Salvatore Girardo
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Max Planck Institute for the Science of Light, Max-Planck-Zentrum für Physik und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Nicole Träber
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Elisabeth Fischer-Friedrich
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Martin Kräter
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Max Planck Institute for the Science of Light, Max-Planck-Zentrum für Physik und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Katrin Wagner
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Thomas Kurth
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Isabel Richter
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Barbara Haller
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Marcus Binner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Dominik Hahn
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Carsten Werner
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Jochen Guck
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Max Planck Institute for the Science of Light, Max-Planck-Zentrum für Physik und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| |
Collapse
|
18
|
Dlugos CP, Picciotto C, Lepa C, Krakow M, Stöber A, Eddy ML, Weide T, Jeibmann A, P Krahn M, Van Marck V, Klingauf J, Ricker A, Wedlich-Söldner R, Pavenstädt H, Klämbt C, George B. Nephrin Signaling Results in Integrin β1 Activation. J Am Soc Nephrol 2019; 30:1006-1019. [PMID: 31097607 DOI: 10.1681/asn.2018040362] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with certain mutations in the gene encoding the slit diaphragm protein Nephrin fail to develop functional slit diaphragms and display severe proteinuria. Many adult-onset glomerulopathies also feature alterations in Nephrin expression and function. Nephrin signals from the podocyte slit diaphragm to the Actin cytoskeleton by recruiting proteins that can interact with C3G, a guanine nucleotide exchange factor of the small GTPase Rap1. Because Rap activity affects formation of focal adhesions, we hypothesized that Nephrin transmits signals to the Integrin receptor complex, which mediates podocyte adhesion to the extracellular matrix. METHODS To investigate Nephrin's role in transmitting signals to the Integrin receptor complex, we conducted genetic studies in Drosophila nephrocytes and validated findings from Drosophila in a cultured human podocyte model. RESULTS Drosophila nephrocytes form a slit diaphragm-like filtration barrier and express the Nephrin ortholog Sticks and stones (Sns). A genetic screen identified c3g as necessary for nephrocyte function. In vivo, nephrocyte-specific gene silencing of sns or c3g compromised nephrocyte filtration and caused nephrocyte diaphragm defects. Nephrocytes with impaired Sns or C3G expression displayed an altered localization of Integrin and the Integrin-associated protein Talin. Furthermore, gene silencing of c3g partly rescued nephrocyte diaphragm defects of an sns overexpression phenotype, pointing to genetic interaction of sns and c3g in nephrocytes. We also found that activated Nephrin recruited phosphorylated C3G and resulted in activation of Integrin β1 in cultured podocytes. CONCLUSIONS Our findings suggest that Nephrin can mediate a signaling pathway that results in activation of Integrin β1 at focal adhesions, which may affect podocyte attachment to the extracellular matrix.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christian Klämbt
- Neurobiology, Westfälische-Wilhelms University Münster, Münster, Germany
| | | |
Collapse
|
19
|
Zhang X, Xing XX, Cui JF. Invadopodia formation: An important step in matrix stiffness-regulated tumor invasion and metastasis. Shijie Huaren Xiaohua Zazhi 2019; 27:589-597. [DOI: 10.11569/wcjd.v27.i9.589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Highly motile and invasive abilities are symbolic features of metastatic tumor cells. Being a critical molecular event for maintaining the highly migratory and invasive capabilities of tumor cells, invadopodia formation undoubtedly determines the progression of tumor invasion and metastasis. Growing numbers of studies suggest that increased matrix stiffness, as a notable property of physical mechanics in solid tumors, participates in the regulation of tumor invasion and metastasis via different molecular mechanisms. However, to date the relevant mechanisms of matrix stiffness-induced invadopodia formation and activity in tumor cells remain largely unclear. This paper is to make a review on the structure and function of invadopodia, the stages and inductive factors of invadopodia formation, the regulatory mechanisms of matrix stiffness-induced invadopodia formation and so on, with an aim to reveal the important roles of invadopodia in matrix stiffness-regulated tumor invasion and metastasis.
Collapse
Affiliation(s)
- Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Xia Xing
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jie-Feng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Multiscale Modelling of Fibres Dynamics and Cell Adhesion within Moving Boundary Cancer Invasion. Bull Math Biol 2019; 81:2176-2219. [PMID: 30980344 PMCID: PMC6612324 DOI: 10.1007/s11538-019-00598-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022]
Abstract
Recognised as one of the hallmarks of cancer, local cancer cell invasion is a complex multiscale process that combines the secretion of matrix-degrading enzymes with a series of altered key cell processes (such as abnormal cell proliferation and changes in cell–cell and cell–matrix adhesion leading to enhanced migration) to degrade important components of the surrounding extracellular matrix (ECM) and this way spread further in the human tissue. In order to gain a deeper understanding of the invasion process, we pay special attention to the interacting dynamics between the cancer cell population and various constituents of the surrounding tumour microenvironment. To that end, we consider the key role that ECM plays within the human body tissue, and in particular we focus on the special contribution of its fibrous proteins components, such as collagen and fibronectin, which play an important part in cell proliferation and migration. In this work, we consider the two-scale dynamic cross-talk between cancer cells and a two-component ECM (consisting of both a fibre and a non-fibre phase). To that end, we incorporate the interlinked two-scale dynamics of cell–ECM interactions within the tumour support that contributes simultaneously both to cell adhesion and to the dynamic rearrangement and restructuring of the ECM fibres. Furthermore, this is embedded within a multiscale moving boundary approach for the invading cancer cell population, in the presence of cell adhesion at the tissue scale and cell-scale fibre redistribution activity and leading edge matrix-degrading enzyme molecular proteolytic processes. The overall modelling framework will be accompanied by computational results that will explore the impact on cancer invasion patterns of different levels of cell adhesion in conjunction with the continuous ECM fibres rearrangement.
Collapse
|
21
|
Wong SW, Lenzini S, Shin JW. Perspective: Biophysical regulation of cancerous and normal blood cell lineages in hematopoietic malignancies. APL Bioeng 2018; 2:031802. [PMID: 31069313 PMCID: PMC6324213 DOI: 10.1063/1.5025689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/04/2018] [Indexed: 01/15/2023] Open
Abstract
It is increasingly appreciated that physical forces play important roles in cancer biology, in terms of progression, invasiveness, and drug resistance. Clinical progress in treating hematological malignancy and in developing cancer immunotherapy highlights the role of the hematopoietic system as a key model in devising new therapeutic strategies against cancer. Understanding mechanobiology of the hematopoietic system in the context of cancer will thus yield valuable fundamental insights that can information about novel cancer therapeutics. In this perspective, biophysical insights related to blood cancer are defined and detailed. The interactions with immune cells relevant to immunotherapy against cancer are considered and expounded, followed by speculation of potential regulatory roles of mesenchymal stromal cells (MSCs) in this complex network. Finally, a perspective is presented as to how insights from these complex interactions between matrices, blood cancer cells, immune cells, and MSCs can be leveraged to influence and engineer the treatment of blood cancers in the clinic.
Collapse
Affiliation(s)
- Sing Wan Wong
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA and Department of Bioengineering, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Stephen Lenzini
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA and Department of Bioengineering, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Jae-Won Shin
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA and Department of Bioengineering, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| |
Collapse
|
22
|
Rose JC, De Laporte L. Hierarchical Design of Tissue Regenerative Constructs. Adv Healthc Mater 2018; 7:e1701067. [PMID: 29369541 DOI: 10.1002/adhm.201701067] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/01/2017] [Indexed: 02/05/2023]
Abstract
The worldwide shortage of organs fosters significant advancements in regenerative therapies. Tissue engineering and regeneration aim to supply or repair organs or tissues by combining material scaffolds, biochemical signals, and cells. The greatest challenge entails the creation of a suitable implantable or injectable 3D macroenvironment and microenvironment to allow for ex vivo or in vivo cell-induced tissue formation. This review gives an overview of the essential components of tissue regenerating scaffolds, ranging from the molecular to the macroscopic scale in a hierarchical manner. Further, this review elaborates about recent pivotal technologies, such as photopatterning, electrospinning, 3D bioprinting, or the assembly of micrometer-scale building blocks, which enable the incorporation of local heterogeneities, similar to most native extracellular matrices. These methods are applied to mimic a vast number of different tissues, including cartilage, bone, nerves, muscle, heart, and blood vessels. Despite the tremendous progress that has been made in the last decade, it remains a hurdle to build biomaterial constructs in vitro or in vivo with a native-like structure and architecture, including spatiotemporal control of biofunctional domains and mechanical properties. New chemistries and assembly methods in water will be crucial to develop therapies that are clinically translatable and can evolve into organized and functional tissues.
Collapse
Affiliation(s)
- Jonas C. Rose
- DWI—Leibniz Institute for Interactive Materials Forckenbeckstr. 50 Aachen D‐52074 Germany
| | - Laura De Laporte
- DWI—Leibniz Institute for Interactive Materials Forckenbeckstr. 50 Aachen D‐52074 Germany
| |
Collapse
|
23
|
Maturation State and Matrix Microstructure Regulate Interstitial Cell Migration in Dense Connective Tissues. Sci Rep 2018; 8:3295. [PMID: 29459687 PMCID: PMC5818574 DOI: 10.1038/s41598-018-21212-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/31/2018] [Indexed: 12/23/2022] Open
Abstract
Few regenerative approaches exist for the treatment of injuries to adult dense connective tissues. Compared to fetal tissues, adult connective tissues are hypocellular and show limited healing after injury. We hypothesized that robust repair can occur in fetal tissues with an immature extracellular matrix (ECM) that is conducive to cell migration, and that this process fails in adults due to the biophysical barriers imposed by the mature ECM. Using the knee meniscus as a platform, we evaluated the evolving micromechanics and microstructure of fetal and adult tissues, and interrogated the interstitial migratory capacity of adult meniscal cells through fetal and adult tissue microenvironments with or without partial enzymatic digestion. To integrate our findings, a computational model was implemented to determine how changing biophysical parameters impact cell migration through these dense networks. Our results show that the micromechanics and microstructure of the adult meniscus ECM sterically hinder cell mobility, and that modulation of these ECM attributes via an exogenous matrix-degrading enzyme permits migration through this otherwise impenetrable network. By addressing the inherent limitations to repair imposed by the mature ECM, these studies may define new clinical strategies to promote repair of damaged dense connective tissues in adults.
Collapse
|
24
|
Haak AJ, Tan Q, Tschumperlin DJ. Matrix biomechanics and dynamics in pulmonary fibrosis. Matrix Biol 2017; 73:64-76. [PMID: 29274939 DOI: 10.1016/j.matbio.2017.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/09/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
The composition and mechanical properties of the extracellular matrix are dramatically altered during the development and progression of pulmonary fibrosis. Recent evidence indicates that these changes in matrix composition and mechanics are not only end-results of fibrotic remodeling, but active participants in driving disease progression. These insights have stimulated interest in identifying the components and physical aspects of the matrix that contribute to cell activation and disease initiation and progression. This review summarizes current knowledge regarding the biomechanics and dynamics of the ECM in mouse models and human IPF, and discusses how matrix mechanical and compositional changes might be non-invasively assessed, therapeutically targeted, and biologically restored to resolve fibrosis.
Collapse
Affiliation(s)
- Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States
| | - Qi Tan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States.
| |
Collapse
|
25
|
Chou CK, Fan CC, Lin PS, Liao PY, Tung JC, Hsieh CH, Hung MC, Chen CH, Chang WC. Sciellin mediates mesenchymal-to-epithelial transition in colorectal cancer hepatic metastasis. Oncotarget 2017; 7:25742-54. [PMID: 27013588 PMCID: PMC5041940 DOI: 10.18632/oncotarget.8264] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/11/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatic metastasis is the major cause of mortality in colorectal cancer (CRC) patients. Using proteomic analysis, we found sciellin (SCEL) to be specifically expressed in hepatic metastatic CRC cell lines. SCEL knockdown increased CRC cell migration and invasion, while overexpression had the opposite effect. SCEL knockdown also caused cancer cells to form more invasive structures within 3D cultures, increased the mesenchymal marker vimentin, and attenuated the epithelial marker E-cadherin. SCEL increased WNT signaling by activating β-catenin and its downstream target c-myc, and activated mesenchymal-to-epithelial transition (MET) through a SCEL-β-catenin-E-cadherin axis. SCEL showed higher expression in late stage primary CRC than in its hepatic metastatic counterpart. SCEL expression is dynamically modulated by TGF-β1 and hypoxia, revealing a plastic MET mechanism for tumor colonization. Intrahepatic injection in immunodeficient mice revealed that SCEL is necessary for metastatic CRC tumor growth in the liver. These results demonstrate that SCEL is a MET inducer dynamically regulated through the metastasis process. They suggest SCEL may be a useful therapeutic target for preventing or eliminating CRC hepatic metastasis.
Collapse
Affiliation(s)
- Chuan-Kai Chou
- National Applied Research Laboratories, National Laboratory Animal Center, Taipei, Taiwan
| | - Chi-Chen Fan
- Superintendent Office, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan
| | - Pei-Shan Lin
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Yu Liao
- National Applied Research Laboratories, National Laboratory Animal Center, Taipei, Taiwan
| | - Jia-Chen Tung
- Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| | - Chang-Hsun Hsieh
- Department of Orthopaedic Surgery, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Mien-Chie Hung
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chung-Hsuan Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Biochemistry & Molecular Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Chemistry, National Taiwan University, Taipei, Taiwan.,Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| |
Collapse
|
26
|
Ngan E, Kiepas A, Brown CM, Siegel PM. Emerging roles for LPP in metastatic cancer progression. J Cell Commun Signal 2017; 12:143-156. [PMID: 29027626 DOI: 10.1007/s12079-017-0415-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/03/2017] [Indexed: 01/21/2023] Open
Abstract
LIM domain containing proteins are important regulators of diverse cellular processes, and play pivotal roles in regulating the actin cytoskeleton. Lipoma Preferred Partner (LPP) is a member of the zyxin family of LIM proteins that has long been characterized as a promoter of mesenchymal/fibroblast cell migration. More recently, LPP has emerged as a critical inducer of tumor cell migration, invasion and metastasis. LPP is thought to contribute to these malignant phenotypes by virtue of its ability to shuttle into the nucleus, localize to adhesions and, most recently, to promote invadopodia formation. In this review, we will examine the mechanisms through which LPP regulates the functions of adhesions and invadopodia, and discuss potential roles of LPP in mediating cellular responses to mechanical cues within these mechanosensory structures.
Collapse
Affiliation(s)
- Elaine Ngan
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 508, Montréal, Québec, H3A 1A3, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Alex Kiepas
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Claire M Brown
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 508, Montréal, Québec, H3A 1A3, Canada. .,Department of Medicine, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
27
|
Urbano RL, Furia C, Basehore S, Clyne AM. Stiff Substrates Increase Inflammation-Induced Endothelial Monolayer Tension and Permeability. Biophys J 2017; 113:645-655. [PMID: 28793219 PMCID: PMC5550298 DOI: 10.1016/j.bpj.2017.06.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/15/2017] [Accepted: 06/13/2017] [Indexed: 01/22/2023] Open
Abstract
Arterial stiffness and inflammation are associated with atherosclerosis, and each have individually been shown to increase endothelial monolayer tension and permeability. The objective of this study was to determine if substrate stiffness enhanced endothelial monolayer tension and permeability in response to inflammatory cytokines. Porcine aortic endothelial cells were cultured at confluence on polyacrylamide gels of varying stiffness and treated with either tumor necrosis factor-α (TNFα) or thrombin. Monolayer tension was measured through vinculin localization at the cell membrane, traction force microscopy, and phosphorylated myosin light chain quantity and actin fiber colocalization. Cell permeability was measured by cell-cell junction confocal microscopy and a dextran permeability assay. When treated with TNFα or thrombin, endothelial monolayers on stiffer substrates showed increased traction forces, vinculin at the cell membrane, and vinculin phosphorylation, suggesting elevated monolayer tension. Interestingly, VE-cadherin shifted toward a smaller molecular weight in endothelial monolayers on softer substrates, which may relate to increased VE-cadherin endocytosis and degradation. Phosphorylated myosin light chain colocalization with actin stress fibers increased in endothelial monolayers treated with TNFα or thrombin on stiffer substrates, indicating elevated cell monolayer contractility. Endothelial monolayers also developed focal adherens intercellular junctions and became more permeable when cultured on stiffer substrates in the presence of the inflammatory cytokines. Whereas each of these effects was likely mitigated by Rho/ROCK, Rho/ROCK pathway inhibition via Y27632 disrupted cell-cell junction morphology, showing that cell contractility is required to maintain adherens junction integrity. These data suggest that stiff substrates change intercellular junction protein localization and degradation, which may counteract the inflammation-induced increase in endothelial monolayer tension and thereby moderate inflammation-induced junction loss and associated endothelial monolayer permeability on stiffer substrates.
Collapse
|
28
|
Saitakis M, Dogniaux S, Goudot C, Bufi N, Asnacios S, Maurin M, Randriamampita C, Asnacios A, Hivroz C. Different TCR-induced T lymphocyte responses are potentiated by stiffness with variable sensitivity. eLife 2017; 6. [PMID: 28594327 PMCID: PMC5464771 DOI: 10.7554/elife.23190] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/07/2017] [Indexed: 12/26/2022] Open
Abstract
T cells are mechanosensitive but the effect of stiffness on their functions is still debated. We characterize herein how human primary CD4+ T cell functions are affected by stiffness within the physiological Young’s modulus range of 0.5 kPa to 100 kPa. Stiffness modulates T lymphocyte migration and morphological changes induced by TCR/CD3 triggering. Stiffness also increases TCR-induced immune system, metabolism and cell-cycle-related genes. Yet, upon TCR/CD3 stimulation, while cytokine production increases within a wide range of stiffness, from hundreds of Pa to hundreds of kPa, T cell metabolic properties and cell cycle progression are only increased by the highest stiffness tested (100 kPa). Finally, mechanical properties of adherent antigen-presenting cells modulate cytokine production by T cells. Together, these results reveal that T cells discriminate between the wide range of stiffness values found in the body and adapt their responses accordingly. DOI:http://dx.doi.org/10.7554/eLife.23190.001 Our immune system contains many cells that play various roles in defending the body against infection, cancer and other threats. For example, T cells constantly patrol the body ready to detect and respond to dangers. They do so by gathering cues from their surroundings, which can be specific chemical signals or physical properties such as the stiffness of tissues. Once the T cells are active they respond in several different ways including releasing hormones and dividing to produce more T cells. Tissue stiffness varies considerably between different organs. Furthermore, disease can lead to changes in tissue stiffness. For example, tissues become more rigid when they are inflamed. The stiffness and other physical properties of the surfaces that T cells interact with affect how the cells respond when they detect a threat, but few details are known about exactly how these cues tune T cell responses. Saitakis et al. studied how human T cells respond to artificial surfaces of varying stiffness that mimic the range found in the body. The experiments show that T cells that interact with stiff surfaces become more active than T cells that interact with softer surfaces. However, some responses are more sensitive to the stiffness of the surface than others. For example, the ability of the T cells to release hormones was affected by the whole range of stiffnesses tested in the experiments, whereas only very stiff surfaces stimulated the T cells to divide. These findings show that T cells can detect the stiffness of surfaces in the body and use this to adapt how they respond to threats. Future challenges will be to find out how T cells sense the physical properties of their surroundings and investigate whether cell and tissue stiffness affects immune responses in the body. This will help us to understand how T cells fight infections and other threats, and could be used to develop new ways of boosting these cells to fight cancer and other diseases. DOI:http://dx.doi.org/10.7554/eLife.23190.002
Collapse
Affiliation(s)
- Michael Saitakis
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Stéphanie Dogniaux
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Christel Goudot
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Nathalie Bufi
- Laboratoire Matières et systèmes complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France
| | - Sophie Asnacios
- Laboratoire Matières et systèmes complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France.,Department of Physics, Sorbonne Universités, UPMC Université Paris, Paris, France
| | - Mathieu Maurin
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| | - Clotilde Randriamampita
- INSERM, U1016, Institut Cochin & UMR8104, CNRS & Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Atef Asnacios
- Laboratoire Matières et systèmes complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France
| | - Claire Hivroz
- Institut Curie Section Recherche, INSERM U932 & PSL Research University, Paris, France
| |
Collapse
|
29
|
LPP is a Src substrate required for invadopodia formation and efficient breast cancer lung metastasis. Nat Commun 2017; 8:15059. [PMID: 28436416 PMCID: PMC5413977 DOI: 10.1038/ncomms15059] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/24/2017] [Indexed: 01/17/2023] Open
Abstract
We have previously shown that lipoma preferred partner (LPP) mediates TGFβ-induced breast cancer cell migration and invasion. Herein, we demonstrate that diminished LPP expression reduces circulating tumour cell numbers, impairs cancer cell extravasation and diminishes lung metastasis. LPP localizes to invadopodia, along with Tks5/actin, at sites of matrix degradation and at the tips of extravasating breast cancer cells as revealed by intravital imaging of the chick chorioallantoic membrane (CAM). Invadopodia formation, breast cancer cell extravasation and metastasis require an intact LPP LIM domain and the ability of LPP to interact with α-actinin. Finally, we show that Src-mediated LPP phosphorylation at specific tyrosine residues (Y245/301/302) is critical for invadopodia formation, breast cancer cell invasion and metastasis. Together, these data define a previously unknown function for LPP in the formation of invadopodia and reveal a requirement for LPP in mediating the metastatic ability of breast cancer cells.
Collapse
|
30
|
Pinto M, Rios E, Silva A, Neves S, Caires H, Pinto A, Durães C, Carvalho F, Cardoso A, Santos N, Barrias C, Nascimento D, Pinto-do-Ó P, Barbosa M, Carneiro F, Oliveira M. Decellularized human colorectal cancer matrices polarize macrophages towards an anti-inflammatory phenotype promoting cancer cell invasion via CCL18. Biomaterials 2017; 124:211-224. [DOI: 10.1016/j.biomaterials.2017.02.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/31/2017] [Accepted: 02/03/2017] [Indexed: 02/06/2023]
|
31
|
Chi Q, Shan J, Ding X, Yin T, Wang Y, Jia D, Wang G. Smart mechanosensing machineries enable migration of vascular smooth muscle cells in atherosclerosis-relevant 3D matrices. Cell Biol Int 2017; 41:586-598. [PMID: 28328100 DOI: 10.1002/cbin.10764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/19/2017] [Indexed: 11/05/2022]
Abstract
At the early stage of atherosclerosis, neointima is formed due to the migration of vascular smooth muscle cells (VSMCs) from the media to the intima. VSMCs are surrounded by highly adhesive 3D matrices. They take specific strategies to cross various 3D matrices in the media, including heterogeneous collagen and mechanically strong basement membrane. Migration of VSMCs is potentially caused by biomechanical mechanism. Most in vitro studies focus on cell migration on 2D substrates in response to biochemical factors. How the cells move through 3D matrices under the action of mechanosensing machineries remains unexplored. In this review, we propose that several interesting tension-dependent machineries act as "tractor"-posterior myosin II accumulation, and "wrecker"-anterior podosome maintaining, to power VSMCs ahead. VSMCs embedded in 3D matrices may accumulate a minor myosin II isoform, myosin IIB, at the cell rear. Anisotropic myosin IIB distribution creates cell rear, polarizes cell body, pushes the nucleus and reshapes the cell body, and cooperates with a uniformly distributed myosin IIA to propel the cell forward. On the other hand, matrix digestion by podosome further promote the migration when the matrix becomes denser. Actomyosin tension activates Src to induce podosome in soft 3D matrices and retain the podosome integrity to steadily digest the matrix.
Collapse
Affiliation(s)
- Qingjia Chi
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jieling Shan
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiaorong Ding
- Department of Electronic Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Yazhou Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Dongyu Jia
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
32
|
Siva Sankar P, Che Mat MF, Muniandy K, Xiang BLS, Ling PS, Hoe SLL, Khoo ASB, Mohana-Kumaran N. Modeling nasopharyngeal carcinoma in three dimensions. Oncol Lett 2017; 13:2034-2044. [PMID: 28454359 DOI: 10.3892/ol.2017.5697] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 08/19/2016] [Indexed: 12/23/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a type of cancer endemic in Asia, including Malaysia, Southern China, Hong Kong and Taiwan. Treatment resistance, particularly in recurring cases, remains a challenge. Thus, studies to develop novel therapeutic agents are important. Potential therapeutic compounds may be effectively examined using two-dimensional (2D) cell culture models, three-dimensional (3D) spheroid models or in vivo animal models. The majority of drug assessments for cancers, including for NPC, are currently performed with 2D cell culture models. This model offers economical and high-throughput screening advantages. However, 2D cell culture models cannot recapitulate the architecture and the microenvironment of a tumor. In vivo models may recapitulate certain architectural and microenvironmental conditions of a tumor, however, these are not feasible for the screening of large numbers of compounds. By contrast, 3D spheroid models may be able to recapitulate a physiological microenvironment not observed in 2D cell culture models, in addition to avoiding the impediments of in vivo animal models. Thus, the 3D spheroid model offers a more representative model for the study of NPC growth, invasion and drug response, which may be cost-effective without forgoing quality.
Collapse
Affiliation(s)
- Prabu Siva Sankar
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Gelugor, Malaysia.,Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Malaysia
| | - Mohd Firdaus Che Mat
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588 Kuala Lumpur, Malaysia
| | - Kalaivani Muniandy
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Gelugor, Malaysia
| | | | - Phang Su Ling
- School of Biological Sciences, Universiti Sains Malaysia, 11800 Gelugor, Malaysia
| | - Susan Ling Ling Hoe
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588 Kuala Lumpur, Malaysia
| | - Alan Soo-Beng Khoo
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588 Kuala Lumpur, Malaysia
| | | |
Collapse
|
33
|
Cepeda MA, Pelling JJ, Evered CL, Leong HS, Damjanovski S. The cytoplasmic domain of MT1-MMP is dispensable for migration augmentation but necessary to mediate viability of MCF-7 breast cancer cells. Exp Cell Res 2016; 350:169-183. [PMID: 27889376 DOI: 10.1016/j.yexcr.2016.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/28/2023]
Abstract
Membrane-type-1 Matrix Metalloproteinase (MT1-MMP) is a multifunctional protease that regulates ECM degradation, proMMP-2 activation, and varied cellular processes including migration and viability. MT1-MMP is believed to be a central mediator of tumourigenesis whose role is dictated by its functionally distinct protein domains. Both the localization and signal transduction capabilities of MT1-MMP are dependent on its cytoplasmic domain, exemplifying diverse regulatory functions. To further our understanding of the multifunctional contributions of MT1-MMP to cellular processes, we overexpressed cytoplasmic domain altered constructs in MCF-7 breast cancer cells and analyzed migration and viability in 2D culture conditions, morphology in 3D Matrigel culture, and tumorigenic ability in vivo. We found that the cytoplasmic domain was not needed for MT1-MMP mediated migration promotion, but was necessary to maintain viability during serum depravation in 2D culture. Similarly, during 3D Matrigel culture the cytoplasmic domain of MT1-MMP was not needed to initiate a protrusive phenotype, but was necessary to prevent colony blebbing when cells were serum deprived. We also tested in vivo tumorigenic potential to show that cells expressing cytoplasmic domain altered constructs demonstrated a reduced ability to vascularize tumours. These results suggest that the cytoplasmic domain regulates MT1-MMP function in a manner required for cell survival, but is dispensable for cell migration.
Collapse
Affiliation(s)
- Mario A Cepeda
- Department of Biology, Faculty of Science, University of Western Ontario, London, ON, Canada
| | - Jacob Jh Pelling
- Department of Biology, Faculty of Science, University of Western Ontario, London, ON, Canada
| | - Caitlin L Evered
- Department of Biology, Faculty of Science, University of Western Ontario, London, ON, Canada
| | - Hon S Leong
- Department of Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Translational Prostate Cancer Research Laboratory, Lawson Health Research Institute, London, ON, Canada
| | - Sashko Damjanovski
- Department of Biology, Faculty of Science, University of Western Ontario, London, ON, Canada; Associate Scientist, Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
34
|
Chan CK, Pan Y, Nyberg K, Marra MA, Lim EL, Jones SJM, Maar D, Gibb EA, Gunaratne PH, Robertson AG, Rowat AC. Tumour-suppressor microRNAs regulate ovarian cancer cell physical properties and invasive behaviour. Open Biol 2016; 6:160275. [PMID: 27906134 PMCID: PMC5133448 DOI: 10.1098/rsob.160275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
The activities of pathways that regulate malignant transformation can be influenced by microRNAs (miRs). Recently, we showed that increased expression of five tumour-suppressor miRs, miR-508-3p, miR-508-5p, miR-509-3p, miR-509-5p and miR-130b-3p, correlate with improved clinical outcomes in human ovarian cancer patients, and that miR-509-3p attenuates invasion of ovarian cancer cell lines. Here, we investigate the mechanism underlying this reduced invasive potential by assessing the impact of these five miRs on the physical properties of cells. Human ovarian cancer cells (HEYA8, OVCAR8) that are transfected with miR mimics representing these five miRs exhibit decreased invasion through collagen matrices, increased cell size and reduced deformability as measured by microfiltration and microfluidic assays. To understand the molecular basis of altered invasion and deformability induced by these miRs, we use predicted and validated mRNA targets that encode structural and signalling proteins that regulate cell mechanical properties. Combined with analysis of gene transcripts by real-time PCR and image analysis of F-actin in single cells, our results suggest that these tumour-suppressor miRs may alter cell physical properties by regulating the actin cytoskeleton. Our findings provide biophysical insights into how tumour-suppressor miRs can regulate the invasive behaviour of ovarian cancer cells, and identify potential therapeutic targets that may be implicated in ovarian cancer progression.
Collapse
Affiliation(s)
- Clara K Chan
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Yinghong Pan
- Department of Biochemistry and Biology, University of Houston, Houston, TX, USA
| | - Kendra Nyberg
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Marco A Marra
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emilia L Lim
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Dianna Maar
- Bio-Rad Laboratories, The Digital Biology Center, Pleasanton, CA, USA
| | - Ewan A Gibb
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Preethi H Gunaratne
- Department of Biochemistry and Biology, University of Houston, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - A Gordon Robertson
- British Columbia Cancer Agency, Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
35
|
Arya AD, Hallur PM, Karkisaval AG, Gudipati A, Rajendiran S, Dhavale V, Ramachandran B, Jayaprakash A, Gundiah N, Chaubey A. Gelatin Methacrylate Hydrogels as Biomimetic Three-Dimensional Matrixes for Modeling Breast Cancer Invasion and Chemoresponse in Vitro. ACS APPLIED MATERIALS & INTERFACES 2016; 8:22005-17. [PMID: 27494432 DOI: 10.1021/acsami.6b06309] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Recent studies have shown that three-dimensional (3D) culture environments allow the study of cellular responses in a setting that more closely resembles the in vivo milieu. In this context, hydrogels have become popular scaffold options for the 3D cell culture. Because the mechanical and biochemical properties of culture matrixes influence crucial cell behavior, selecting a suitable matrix for replicating in vivo cellular phenotype in vitro is essential for understanding disease progression. Gelatin methacrylate (GelMA) hydrogels have been the focus of much attention because of their inherent bioactivity, favorable hydration and diffusion properties, and ease-of-tailoring of their physicochemical characteristics. Therefore, in this study we examined the efficacy of GelMA hydrogels as a suitable platform to model specific attributes of breast cancer. We observed increased invasiveness in vitro and increased tumorigenic ability in vivo in breast cancer cells cultured on GelMA hydrogels. Further, cells cultured on GelMA matrixes were more resistant to paclitaxel treatment, as shown by the results of cell-cycle analysis and gene expression. This study, therefore, validates GelMA hydrogels as inexpensive, cell-responsive 3D platforms for modeling key characteristics associated with breast cancer metastasis, in vitro.
Collapse
Affiliation(s)
- Anuradha D Arya
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research , Narayana Hrudayalaya Health City, Hosur Road, Bangalore 560 099, India
| | - Pavan M Hallur
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research , Narayana Hrudayalaya Health City, Hosur Road, Bangalore 560 099, India
| | - Abhijith G Karkisaval
- Department of Mechanical Engineering, Indian Institute of Science , Bangalore 560 012, India
| | - Aditi Gudipati
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research , Narayana Hrudayalaya Health City, Hosur Road, Bangalore 560 099, India
| | - Satheesh Rajendiran
- In Vivo Pharmacology-Oncology, Syngene International Ltd. , Plot Nos. 2 & 3, Bommasandra IV Phase, Jigani Link Road, Bangalore 560 099, India
| | - Vaibhav Dhavale
- In Vivo Pharmacology-Oncology, Syngene International Ltd. , Plot Nos. 2 & 3, Bommasandra IV Phase, Jigani Link Road, Bangalore 560 099, India
| | - Balaji Ramachandran
- In Vivo Pharmacology-Oncology, Syngene International Ltd. , Plot Nos. 2 & 3, Bommasandra IV Phase, Jigani Link Road, Bangalore 560 099, India
| | - Aravindakshan Jayaprakash
- In Vivo Pharmacology-Oncology, Syngene International Ltd. , Plot Nos. 2 & 3, Bommasandra IV Phase, Jigani Link Road, Bangalore 560 099, India
| | - Namrata Gundiah
- Department of Mechanical Engineering, Indian Institute of Science , Bangalore 560 012, India
| | - Aditya Chaubey
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research , Narayana Hrudayalaya Health City, Hosur Road, Bangalore 560 099, India
| |
Collapse
|
36
|
Ko P, Kim D, You E, Jung J, Oh S, Kim J, Lee KH, Rhee S. Extracellular Matrix Rigidity-dependent Sphingosine-1-phosphate Secretion Regulates Metastatic Cancer Cell Invasion and Adhesion. Sci Rep 2016; 6:21564. [PMID: 26877098 PMCID: PMC4753492 DOI: 10.1038/srep21564] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022] Open
Abstract
Dynamic interaction between cancer cells and the surrounding microenvironment is critical for cancer progression via changes in cellular behavior including alteration of secreted molecules. However, the molecular mechanisms underlying the influence exerted by the cancer microenvironment on secretion of molecules during cancer progression remain largely unknown. In this study, we report that secretion of spingsine-1-phosphate (S1P) and its regulator, SphK1 expression is dependent of the substrate rigidity, which is critical for the balance between cancer cell invasion and adhesion. Conditioned media (CM) of MDA-MB-231, an aggressive breast cancer cell obtained from soft substrate (~0.5 kPa) induced chemo-attractive invasion, while CM obtained from stiff substrate (~2.5 kPa) increased cell adhesion instead. We found that the expression of SphK1 is upregulated in the stiff substrate, resulting in an increase in S1P levels in the CM. We also found that upregulation of SphK1 expression in the stiff substrate is dominant in metastatic cancer cells but not in primary cancer cells. These results suggest that alterations in the mechanical environment of the ECM surrounding the tumor cells actively regulate cellular properties such as secretion, which in turn, may contribute to cancer progression.
Collapse
Affiliation(s)
- Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Daehwan Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Somi Oh
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jaehyun Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kwang-Ho Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
37
|
Osma-Garcia IC, Punzón C, Fresno M, Díaz-Muñoz MD. Dose-dependent effects of prostaglandin E2 in macrophage adhesion and migration. Eur J Immunol 2015; 46:677-88. [PMID: 26631603 DOI: 10.1002/eji.201545629] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 10/08/2015] [Accepted: 11/26/2015] [Indexed: 12/15/2022]
Abstract
Macrophage migration to the focus of infection is a hallmark of the innate immune response. Macrophage spreading, adhesion, and migration through the extracellular matrix require dynamic remodeling of the actin cytoskeleton associated to integrin clustering in podosomes and focal adhesions. Here, we show that prostaglandin E2 (PGE2 ), the main prostaglandin produced by macrophages during inflammation, promote the distinctive dose-dependent formation of podosomes or focal adhesions in macrophages. Low concentrations of PGE2 increased p110γ PI3K expression, phosphorylation of actin-related protein 2, and formation of podosomes, which enhanced macrophage migration in response to chemokines. However, high doses of PGE2 increased phosphorylation of paxillin and focal adhesion kinase, the expression of serine/threonine protein kinase 1, and promoted focal adhesion formation and macrophage adhesion, reducing macrophage chemotaxis. In summary, we describe the dual role of PGE2 as a promoter of macrophage chemotaxis and adhesion, proposing a new model of macrophage migration to the inflammatory focus in the presence of a gradient of PGE2 .
Collapse
Affiliation(s)
- Inés C Osma-Garcia
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Punzón
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel D Díaz-Muñoz
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
38
|
Abstract
The extracellular matrix (ECM) of the lung serves as both a scaffold for resident cells and a mechanical support for respiratory function. The ECM is deposited during development and undergoes continuous turnover and maintenance during organ growth and homeostasis. Cells of the mesenchyme, including the tissue resident fibroblast, take a leading role in depositing and organizing the matrix and do so in an anatomically distinct fashion, with differing composition, organization, and mechanical properties within the airways, vessels, and alveoli of the lung. Recent technological advancements have allowed the lung's ECM biochemical composition and mechanical properties to be studied with improved resolution, thereby identifying novel disease-related changes in ECM characteristics. In parallel, efforts to study cells seeded on normal and disease-derived matrices have illustrated the powerful role the ECM can play in altering key functions of lung resident cells. The mechanical properties of the matrix have been identified as an important modifier of cell-matrix adhesions, with matrices of pathologic stiffness promoting profibrotic signaling and cell function. Ongoing work is identifying both mechanically activated pathways in mesenchymal cells and disease-related ECM molecules that biochemically regulate cell function. Uncovering the control systems by which cells respond to and regulate the matrix, and the failures in these systems that underlie aberrant repair, remains a major challenge. Progress in this area will be an essential element in efforts to engineer functional lung tissue for regenerative approaches and will be key to identifying new therapeutic strategies for lung diseases characterized by disturbed matrix architecture.
Collapse
|
39
|
Parekh A, Weaver AM. Regulation of invadopodia by mechanical signaling. Exp Cell Res 2015; 343:89-95. [PMID: 26546985 DOI: 10.1016/j.yexcr.2015.10.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/31/2015] [Indexed: 12/15/2022]
Abstract
Mechanical rigidity in the tumor microenvironment is associated with a high risk of tumor formation and aggressiveness. Adhesion-based signaling driven by a rigid microenvironment is thought to facilitate invasion and migration of cancer cells away from primary tumors. Proteolytic degradation of extracellular matrix (ECM) is a key component of this process and is mediated by subcellular actin-rich structures known as invadopodia. Both ECM rigidity and cellular traction stresses promote invadopodia formation and activity, suggesting a role for these structures in mechanosensing. The presence and activity of mechanosensitive adhesive and signaling components at invadopodia further indicates the potential for these structures to utilize myosin-dependent forces to probe and remodel their ECM environments. Here, we provide a brief review of the role of adhesion-based mechanical signaling in controlling invadopodia and invasive cancer behavior.
Collapse
Affiliation(s)
- Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| | - Alissa M Weaver
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| |
Collapse
|
40
|
Brown GT, Murray GI. Current mechanistic insights into the roles of matrix metalloproteinases in tumour invasion and metastasis. J Pathol 2015; 237:273-81. [PMID: 26174849 DOI: 10.1002/path.4586] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/03/2015] [Accepted: 07/08/2015] [Indexed: 12/12/2022]
Abstract
The purpose of this review is to highlight the recent mechanistic developments elucidating the role of matrix metalloproteinases (MMPs) in tumour invasion and metastasis. The ability of tumour cells to invade, migrate, and subsequently metastasize is a fundamental characteristic of cancer. Tumour invasion and metastasis are increasingly being characterized by the dynamic relationship between cancer cells and their microenvironment and developing a greater understanding of these basic pathological mechanisms is crucial. While MMPs have been strongly implicated in these processes as a result of extensive circumstantial evidence--for example, increased expression of individual MMPs in tumours and association of specific MMPs with prognosis--the underpinning mechanisms are only now being elucidated. Recent studies are now providing a mechanistic basis, highlighting and reinforcing the catalytic and non-catalytic roles of specific MMPs as key players in tumour invasion and metastasis.
Collapse
Affiliation(s)
- Gordon T Brown
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Graeme I Murray
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
41
|
Isabella AJ, Horne-Badovinac S. Building from the Ground up: Basement Membranes in Drosophila Development. CURRENT TOPICS IN MEMBRANES 2015; 76:305-36. [PMID: 26610918 DOI: 10.1016/bs.ctm.2015.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Basement membranes (BMs) are sheetlike extracellular matrices found at the basal surfaces of epithelial tissues. The structural and functional diversity of these matrices within the body endows them with the ability to affect multiple aspects of cell behavior and communication; for this reason, BMs are integral to many developmental processes. The power of Drosophila genetics, as applied to the BM, has yielded substantial insight into how these matrices influence development. Here, we explore three facets of BM biology to which Drosophila research has made particularly important contributions. First, we discuss how newly synthesized BM proteins are secreted to and assembled exclusively on basal epithelial surfaces. Next, we examine how regulation of the structural properties of the BM mechanically supports and guides tissue morphogenesis. Finally, we explore how BMs influence development through the modulation of several major signaling pathways.
Collapse
Affiliation(s)
- Adam J Isabella
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL, USA
| | - Sally Horne-Badovinac
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL, USA; Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
42
|
da Rocha-Azevedo B, Ho CH, Grinnell F. PDGF‑stimulated dispersal of cell clusters and disruption of fibronectin matrix on three-dimensional collagen matrices requires matrix metalloproteinase-2. Mol Biol Cell 2015; 26:1098-105. [PMID: 25589674 PMCID: PMC4357509 DOI: 10.1091/mbc.e14-09-1396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Previous studies showed that morphogenic cell clustering depends on fibronectin fibrillar matrix assembly under procontractile conditions. The present study shows that disruption of fibronectin matrix necessary for dispersal of cell clusters under promigratory conditions requires matrix metalloproteinases, especially MMP-2. Formation of cell clusters is a common morphogenic cell behavior observed during tissue and organ development and homeostasis, as well as during pathological disorders. Dynamic regulation of cell clustering depends on the balance between contraction of cells into clusters and migration of cells as dispersed individuals. Previously we reported that under procontractile culture conditions, fibronectin fibrillar matrix assembly by human fibroblasts functioned as a nucleation center for cell clustering on three-dimensional collagen matrices. Here we report that switching preformed cell clusters from procontractile to promigratory culture conditions results in cell dispersal out of clusters and disruption of FN matrix. Experiments using small interfering RNA silencing and pharmacological inhibition demonstrated that matrix metalloproteinase activity involving MMP-2 was necessary for fibronectin matrix disruption and dispersal of cell clusters.
Collapse
Affiliation(s)
| | - Chin-Han Ho
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390-9039
| | - Frederick Grinnell
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390-9039
| |
Collapse
|
43
|
Fonseca KB, Granja PL, Barrias CC. Engineering proteolytically-degradable artificial extracellular matrices. Prog Polym Sci 2014. [DOI: 10.1016/j.progpolymsci.2014.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
44
|
Martinelli R, Zeiger AS, Whitfield M, Sciuto TE, Dvorak A, Van Vliet KJ, Greenwood J, Carman CV. Probing the biomechanical contribution of the endothelium to lymphocyte migration: diapedesis by the path of least resistance. J Cell Sci 2014; 127:3720-34. [PMID: 25002404 DOI: 10.1242/jcs.148619] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Immune cell trafficking requires the frequent breaching of the endothelial barrier either directly through individual cells ('transcellular' route) or through the inter-endothelial junctions ('paracellular' route). What determines the loci or route of breaching events is an open question with important implications for overall barrier regulation. We hypothesized that basic biomechanical properties of the endothelium might serve as crucial determinants of this process. By altering junctional integrity, cytoskeletal morphology and, consequently, local endothelial cell stiffness of different vascular beds, we could modify the preferred route of diapedesis. In particular, high barrier function was associated with predominantly transcellular migration, whereas negative modulation of junctional integrity resulted in a switch to paracellular diapedesis. Furthermore, we showed that lymphocytes dynamically probe the underlying endothelium by extending invadosome-like protrusions (ILPs) into its surface that deform the nuclear lamina, distort actin filaments and ultimately breach the barrier. Fluorescence imaging and pharmacologic depletion of F-actin demonstrated that lymphocyte barrier breaching efficiency was inversely correlated with local endothelial F-actin density and stiffness. Taken together, these data support the hypothesis that lymphocytes are guided by the mechanical 'path of least resistance' as they transverse the endothelium, a process we term 'tenertaxis'.
Collapse
Affiliation(s)
- Roberta Martinelli
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA Department of Cell Biology, Institute of Ophthalmology, UCL, 11-43 Bath Street, London EC1V 9EL, UK
| | - Adam S Zeiger
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew Whitfield
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tracey E Sciuto
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Ann Dvorak
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Krystyn J Van Vliet
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John Greenwood
- Department of Cell Biology, Institute of Ophthalmology, UCL, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christopher V Carman
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
45
|
Haage A, Nam DH, Ge X, Schneider IC. Matrix metalloproteinase-14 is a mechanically regulated activator of secreted MMPs and invasion. Biochem Biophys Res Commun 2014; 450:213-8. [PMID: 24878529 DOI: 10.1016/j.bbrc.2014.05.086] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 12/21/2022]
Abstract
Matrix metalloproteinases (MMPs) are extracellular matrix (ECM) degrading enzymes and have complex and specific regulation networks. This includes activation interactions, where one MMP family member activates another. ECM degradation and MMP activation can be initiated by several different stimuli including changes in ECM mechanical properties or intracellular contractility. These mechanical stimuli are known enhancers of metastatic potential. MMP-14 facilitates local ECM degradation and is well known as a major mediator of cell migration, angiogenesis and invasion. Recently, function blocking antibodies have been developed to specifically block MMP-14, providing a useful tool for research as well as therapeutic applications. Here we utilize a selective MMP-14 function blocking antibody to delineate the role of MMP-14 as an activator of other MMPs in response to changes in cellular contractility and ECM stiffness. Inhibition using function blocking antibodies reveals that MMP-14 activates soluble MMPs like MMP-2 and -9 under various mechanical stimuli in the pancreatic cancer cell line, Panc-1. In addition, inhibition of MMP-14 abates Panc-1 cell extension into 3D gels to levels seen with non-specific pan-MMP inhibitors at higher concentrations. This strengthens the case for MMP function blocking antibodies as more potent and specific MMP inhibition therapeutics.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Genetics, Development and Cell Biology, Iowa State University, 1210 Molecular Biology Building, Ames, IA 50011-3260, United States; Molecular, Cellular and Developmental Biology Interdepartmental Graduate Program, Iowa State University, 2018 Molecular Biology Building, Ames, IA 50011-3260, United States
| | - Dong Hyun Nam
- Department of Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92512, United States
| | - Xin Ge
- Department of Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92512, United States
| | - Ian C Schneider
- Department of Chemical and Biological Engineering, Iowa State University, 2114 Sweeney Hall, Ames, IA 50011-2230, United States; Department of Genetics, Development and Cell Biology, Iowa State University, 1210 Molecular Biology Building, Ames, IA 50011-3260, United States.
| |
Collapse
|
46
|
Gu Z. 0.1 kilopascal difference for mechanophenotyping: soft matrix precisely regulates cellular architecture for invasion. BIOARCHITECTURE 2014; 4:116-8. [PMID: 25029598 DOI: 10.4161/bioa.29175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Current knowledge understands the mesenchymal cell invasion in a 3D matrix as a combined process of cell-to-matrix adhesion based cell migration and matrix remodeling. Excluding cell invasion stimulated by cytokines and chemokines, the basal cell invasion itself is a complicated process that can be regulated by matrix ligand type, density, geometry, and stiffness, etc. Understanding such a complicated biological process requires delicate dissections into simplified model studies by altering only one or two elements at a time. Past cell motility studies focusing on matrix stiffness have revealed that a stiffer matrix promotes 2D X-Y axis lateral cell motility. Here, we comment on two recent studies that report, instead of stiffer matrix, a softer matrix promotes matrix proteolysis and the formation of invadosome-like protrusions (ILPs) along the 3D Z axis. These studies also reveal that soft matrix precisely regulates such ILPs formation in the stiffness scale range of 0.1 kilopascal in normal cells. In contrast, malignant cells such as cancer cells can form ILPs in response to a much wider range of matrix stiffness. Further, different cancer cells respond to their own favorable range of matrix stiffness to spontaneously form ILPs. Thus, we hereby propose the idea of utilizing the matrix stiffness to precisely regulate ILP formation as a mechanophenotyping tool for cancer metastasis prediction and pathological diagnosis.
Collapse
Affiliation(s)
- Zhizhan Gu
- Division of Rheumatology, Immunology, and Allergy; Department of Medicine; Brigham and Women's Hospital; Harvard Medical School; Boston, MA USA
| |
Collapse
|
47
|
Haage A, Schneider IC. Cellular contractility and extracellular matrix stiffness regulate matrix metalloproteinase activity in pancreatic cancer cells. FASEB J 2014; 28:3589-99. [PMID: 24784579 DOI: 10.1096/fj.13-245613] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The pathogenesis of cancer is often driven by local invasion and metastasis. Recently, mechanical properties of the tumor microenvironment have been identified as potent regulators of invasion and metastasis, while matrix metalloproteinases (MMPs) are classically known as significant enhancers of cancer cell migration and invasion. Here we have been able to sensitively measure MMP activity changes in response to specific extracellular matrix (ECM) environments and cell contractility states. Cells of a pancreatic cancer cell line, Panc-1, up-regulate MMP activities between 3- and 10-fold with increased cell contractility. Conversely, they down-regulate MMP activities when contractility is blocked to levels seen with pan-MMP activity inhibitors. Similar, albeit attenuated, responses are seen in other pancreatic cancer cell lines, BxPC-3 and AsPC-1. In addition, MMP activity was modulated by substrate stiffness, collagen gel concentration, and the degree of collagen cross-linking, when cells were plated on collagen gels ranging from 0.5 to 5 mg/ml that span the physiological range of substrate stiffness (50-2000 Pa). Panc-1 cells showed enhanced MMP activity on stiffer substrates, whereas BxPC-3 and AsPC-1 cells showed diminished MMP activity. In addition, eliminating heparan sulfate proteoglycans using heparinase completely abrogated the mechanical induction of MMP activity. These results demonstrate the first functional link between MMP activity, contractility, and ECM stiffness and provide an explanation as to why stiffer environments result in enhanced cell migration and invasion.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Genetics, Development, and Cell Biology, Molecular, Cellular and Developmental Biology Interdepartmental Graduate Program, and
| | - Ian C Schneider
- Department of Genetics, Development, and Cell Biology, Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, USA
| |
Collapse
|