1
|
Mancini A, Gentile MT, Pentimalli F, Cortellino S, Grieco M, Giordano A. Multiple aspects of matrix stiffness in cancer progression. Front Oncol 2024; 14:1406644. [PMID: 39015505 PMCID: PMC11249764 DOI: 10.3389/fonc.2024.1406644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
The biophysical and biomechanical properties of the extracellular matrix (ECM) are crucial in the processes of cell differentiation and proliferation. However, it is unclear to what extent tumor cells are influenced by biomechanical and biophysical changes of the surrounding microenvironment and how this response varies between different tumor forms, and over the course of tumor progression. The entire ensemble of genes encoding the ECM associated proteins is called matrisome. In cancer, the ECM evolves to become highly dysregulated, rigid, and fibrotic, serving both pro-tumorigenic and anti-tumorigenic roles. Tumor desmoplasia is characterized by a dramatic increase of α-smooth muscle actin expressing fibroblast and the deposition of hard ECM containing collagen, fibronectin, proteoglycans, and hyaluronic acid and is common in many solid tumors. In this review, we described the role of inflammation and inflammatory cytokines, in desmoplastic matrix remodeling, tumor state transition driven by microenvironment forces and the signaling pathways in mechanotransduction as potential targeted therapies, focusing on the impact of qualitative and quantitative variations of the ECM on the regulation of tumor development, hypothesizing the presence of matrisome drivers, acting alongside the cell-intrinsic oncogenic drivers, in some stages of neoplastic progression and in some tumor contexts, such as pancreatic carcinoma, breast cancer, lung cancer and mesothelioma.
Collapse
Affiliation(s)
- Alessandro Mancini
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- BioUp Sagl, Lugano, Switzerland
| | - Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University “Giuseppe De Gennaro,” Casamassima, Bari, Italy
| | - Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, NA, Italy
- Sbarro Health Research Organization (S.H.R.O.) Italia Foundation ETS, Candiolo, TO, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
2
|
Cooley MB, Wulftange WJ, Wegierak D, Goreke U, Abenojar EC, Gurkan UA, Exner AA. Real-time imaging of nanobubble ultrasound contrast agent flow, extravasation, and diffusion through an extracellular matrix using a microfluidic model. LAB ON A CHIP 2023; 23:3453-3466. [PMID: 37424286 DOI: 10.1039/d3lc00514c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Lipid shell-stabilized nanoparticles with a perfluorocarbon gas-core, or nanobubbles, have recently attracted attention as a new contrast agent for molecular ultrasound imaging and image-guided therapy. Due to their small size (∼275 nm diameter) and flexible shell, nanobubbles have been shown to extravasate through hyperpermeable vasculature (e.g., in tumors). However, little is known about the dynamics and depth of extravasation of intact, acoustically active nanobubbles. Accordingly, in this work, we developed a microfluidic chip with a lumen and extracellular matrix (ECM) and imaging method that allows real-time imaging and characterization of the extravasation process with high-frequency ultrasound. The microfluidic device has a lumen and is surrounded by an extracellular matrix with tunable porosity. The combination of ultrasound imaging and the microfluidic chip advantageously produces real-time images of the entire length and depth of the matrix. This captures the matrix heterogeneity, offering advantages over other imaging techniques with smaller fields of view. Results from this study show that nanobubbles diffuse through a 1.3 μm pore size (2 mg mL-1) collagen I matrix 25× faster with a penetration depth that was 0.19 mm deeper than a 3.7 μm (4 mg mL-1) matrix. In the 3.7 μm pore size matrix, nanobubbles diffused 92× faster than large nanobubbles (∼875 nm diameter). Decorrelation time analysis was successfully used to differentiate flowing and extra-luminally diffusing nanobubbles. In this work, we show for the first time that combination of an ultrasound-capable microfluidic chip and real-time imaging provided valuable insight into spatiotemporal nanoparticle movement through a heterogeneous extracellular matrix. This work could help accurately predict parameters (e.g., injection dosage) that improve translation of nanoparticles from in vitro to in vivo environments.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - William J Wulftange
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Utku Goreke
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Eric C Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Umut A Gurkan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|
3
|
Cameron AP, Gao S, Liu Y, Zhao CX. Impact of hydrogel biophysical properties on tumor spheroid growth and drug response. BIOMATERIALS ADVANCES 2023; 149:213421. [PMID: 37060634 DOI: 10.1016/j.bioadv.2023.213421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
The extracellular matrix (ECM) plays a critical role in regulating cell-matrix interactions during tumor progression. These interactions are due in large part to the biophysical properties responding to cancer cell interactions. Within in vitro models, the ECM is mimicked by hydrogels, which possess adjustable biophysical properties that are integral to tumor development. This work presents a systematic and comparative study on the impact of the biophysical properties of two widely used natural hydrogels, Matrigel and collagen gel, on tumor growth and drug response. The biophysical properties of Matrigel and collagen including complex modulus, loss tangent, diffusive permeability, and pore size, were characterised. Then the spheroid growth rates in these two hydrogels were monitored for spheroids with two different sizes (140 μm and 500 μm in diameters). An increased migratory growth was observed in the lower concentration of both the gels. The effect of spheroid incorporation within the hydrogel had a minimal impact on the hydrogel's complex modulus. Finally, 3D tumor models using different concentrations of hydrogels were applied for drug treatment using paclitaxel. Spheroids cultured in hydrogels with different concentrations showed different drug response, demonstrating the significant effect of the choice of hydrogels and their concentrations on the drug response results despite using the same spheroids. This study provides useful insights into the effect of hydrogel biophysical properties on spheroid growth and drug response and highlights the importance of hydrogel selection and in vitro model design.
Collapse
Affiliation(s)
- Anna P Cameron
- Australian institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Song Gao
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, Australia
| | - Yun Liu
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, Australia
| | - Chun-Xia Zhao
- Australian institute of Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia; School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
4
|
Bahr JC, Li XY, Feinberg TY, Jiang L, Weiss SJ. Divergent regulation of basement membrane trafficking by human macrophages and cancer cells. Nat Commun 2022; 13:6409. [PMID: 36302921 PMCID: PMC9613642 DOI: 10.1038/s41467-022-34087-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/13/2022] [Indexed: 12/25/2022] Open
Abstract
Macrophages and cancer cells populations are posited to navigate basement membrane barriers by either mobilizing proteolytic enzymes or deploying mechanical forces. Nevertheless, the relative roles, or identity, of the proteinase -dependent or -independent mechanisms used by macrophages versus cancer cells to transmigrate basement membrane barriers harboring physiologically-relevant covalent crosslinks remains ill-defined. Herein, both macrophages and cancer cells are shown to mobilize membrane-anchored matrix metalloproteinases to proteolytically remodel native basement membranes isolated from murine tissues while infiltrating the underlying interstitial matrix ex vivo. In the absence of proteolytic activity, however, only macrophages deploy actomyosin-generated forces to transmigrate basement membrane pores, thereby providing the cells with proteinase-independent access to the interstitial matrix while simultaneously exerting global effects on the macrophage transcriptome. By contrast, cancer cell invasive activity is reliant on metalloproteinase activity and neither mechanical force nor changes in nuclear rigidity rescue basement membrane transmigration. These studies identify membrane-anchored matrix metalloproteinases as key proteolytic effectors of basement membrane remodeling by macrophages and cancer cells while also defining the divergent invasive strategies used by normal and neoplastic cells to traverse native tissue barriers.
Collapse
Affiliation(s)
- Julian C Bahr
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiao-Yan Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tamar Y Feinberg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Long Jiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen J Weiss
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
5
|
Gain of function of a metalloproteinase associated with multiple myeloma, bicuspid aortic valve, and Von Hippel-Lindau syndrome. Biochem J 2022; 479:1533-1542. [PMID: 35789254 PMCID: PMC10084813 DOI: 10.1042/bcj20220166] [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: 04/04/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022]
Abstract
A patient diagnosed with multiple myeloma, bicuspid aortic valve, and Von Hippel-Lindau syndrome underwent whole-exome sequencing seeking a unified genetic cause for these three pathologies. The patient possessed a single-point mutation of arginine to cysteine (R24C) in the N-terminal region(pro-domain) of matrix metalloproteinase 9 (MMP-9). The pro-domain interacts with the catalytic site of this enzyme rendering it inactive. MMP-9 has previously been associated with all three pathologies suffered by the patient. We hypothesized that the observed mutation in the pro-domain would influence the activity of this enzyme. We expressed recombinant versions of MMP-9 and an investigation of their biochemical properties revealed that MMP-9 R24C is a constitutively active zymogen. To our knowledge, this is the first example of a mutation that discloses catalytic activity in the pro-form in any of the 24 human MMPs.
Collapse
|
6
|
Khalaf MM, Abo-Youssef AM, Malak MN, Hamzawy MA. Novel therapeutic modalities target cell signaling of Renin-Angiotensin system/NF-κB-induced cell cycle arrest and apoptosis in urethane-induced lung cancer in mice: An in vivo study. J Biochem Mol Toxicol 2022; 36:e23162. [PMID: 35822566 DOI: 10.1002/jbt.23162] [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: 12/12/2021] [Revised: 05/20/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Lung cancer has risen to the top of the list of cancer-related deaths worldwide. Aliskiren is a direct renin inhibitor. AIM This study aims to investigate the impact of cell signaling of Renin-Angiotensin system (RAS)/NF-κB on lung cancer by investigating the potential therapeutic effects of aliskiren for lung cancer treatment in urethane-induced lung cancer in mice. METHODS Male BALB/c mice were randomly assigned to one of five treatment groups for 150 days, including (1) normal control; (2) aliskiren (25 mg/kg/i.p) daily, (3) urethane at a dose of 1.5 g/kg (i.p) at Day 1 and 60 (nonsmall cell lung cancer[NSCLC] group) (4) NSCLC mice received carboplatin (15 mg/kg/i.p) every other day for the last 4 successive weeks and (5) NSCLC mice treated with aliskiren daily. Tumor size was determined based on blood sampling, and lungs were isolated for biochemical analysis, western blot analysis assay, and histopathological examination. RESULTS Urethane demonstrated significant changes in all biochemical and molecular parameters and histological patterns. Aliskiren-treated mice had significantly lower levels of NF-κB p65, Bcl-2, cyclin D1, ICAM-1, MMP-2, and Nrf2, with an increase in the catalytic activity of caspase-3 due to its RAS inhibitory mechanism. The combined urethane administration with aliskiren demonstrated a significant improvement in the histopathological examination. CONCLUSION RAS/NF-B cell signaling is a potential therapeutic target for preventing and treating lung adenocarcinoma, evidenced by the fundamental cytotoxic mechanism and attenuation of metastasis and angiogenesis induced by the treatment of NSCLC mice with aliskiren.
Collapse
Affiliation(s)
- Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Amira M Abo-Youssef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
7
|
Brás MM, Sousa SR, Carneiro F, Radmacher M, Granja PL. Mechanobiology of Colorectal Cancer. Cancers (Basel) 2022; 14:1945. [PMID: 35454852 PMCID: PMC9028036 DOI: 10.3390/cancers14081945] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022] Open
Abstract
In this review, the mechanobiology of colorectal cancer (CRC) are discussed. Mechanotransduction of CRC is addressed considering the relationship of several biophysical cues and biochemical pathways. Mechanobiology is focused on considering how it may influence epithelial cells in terms of motility, morphometric changes, intravasation, circulation, extravasation, and metastization in CRC development. The roles of the tumor microenvironment, ECM, and stroma are also discussed, taking into account the influence of alterations and surface modifications on mechanical properties and their impact on epithelial cells and CRC progression. The role of cancer-associated fibroblasts and the impact of flow shear stress is addressed in terms of how it affects CRC metastization. Finally, some insights concerning how the knowledge of biophysical mechanisms may contribute to the development of new therapeutic strategies and targeting molecules and how mechanical changes of the microenvironment play a role in CRC disease are presented.
Collapse
Affiliation(s)
- Maria Manuela Brás
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal
- Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
| | - Susana R. Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto Superior de Engenharia do Porto (ISEP), Instituto Politécnico do Porto (IPP), 4200-072 Porto, Portugal
| | - Fátima Carneiro
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-465 Porto, Portugal
- Serviço de Patologia, Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - Manfred Radmacher
- Institute for Biophysics, University of Bremen, 28334 Bremen, Germany
| | - Pedro L. Granja
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
8
|
Rezaeeyazdi M, Colombani T, Eggermont LJ, Bencherif SA. Engineering hyaluronic acid-based cryogels for CD44-mediated breast tumor reconstruction. Mater Today Bio 2022; 13:100207. [PMID: 35198956 PMCID: PMC8844817 DOI: 10.1016/j.mtbio.2022.100207] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 01/03/2023] Open
Abstract
Breast cancer is a major health concern worldwide and is the leading cause of cancer-related death among American women. Traditional therapies, such as surgery, chemotherapy, and radiotherapy, are usually ineffective. Furthermore, cancer recurrence following targeted therapy often results from acquired drug resistance. Therefore, more realistic tumor models than monolayer cell culture for drug screening and discovery in an in vitro setting would facilitate the development of new therapeutic strategies. Toward this goal, we first developed a simple, rapid, low-cost, and high-throughput method for generating uniform multi-cellular tumor spheroids (MCTS) with controllable size. Next, biomimetic cryogel scaffolds fabricated from hyaluronic acid (HA) were utilized as a platform to reconstruct breast tumor microtissues with aspects of the complex tumor microenvironment in three dimensions. Finally, we investigated the interactions between the HA-based cryogels and CD44-positive breast tumor cells, individually or as MCTS. We found that incorporating the adhesive RGD peptide in cryogels led to the formation of a monolayer of tumor cells on the polymer walls, whereas MCTS cultured on RGD-free HA cryogels resulted in the growth of large and dense microtumors, more similar to native tumor masses. As a result, the MCTS-laden HA cryogel system induced a highly aggressive and chemotherapy drug-resistant tumor model. RGD-free HA-based cryogels represent an effective starting point for designing tumor models for preclinical research, therapeutic drug screening, and early cancer diagnosis.
Collapse
Affiliation(s)
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Loek J. Eggermont
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
9
|
Daisuke H, Kato H, Fukumura K, Mayeda A, Miyagi Y, Seiki M, Koshikawa N. Novel LAMC2 fusion protein has tumor-promoting properties in ovarian carcinoma. Cancer Sci 2021; 112:4957-4967. [PMID: 34689384 PMCID: PMC8645749 DOI: 10.1111/cas.15149] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/10/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Laminins are heterotrimeric ECM proteins composed of α, β, and γ chains. The γ2 chain (Lm-γ2) is a frequently expressed monomer and its expression is closely associated with cancer progression. Laminin-γ2 contains an epidermal growth factor (EGF)-like domain in its domain III (DIII or LEb). Matrix metalloproteinases can cleave off the DIII region of Lm-γ2 that retains the ligand activity for EGF receptor (EGFR). Herein, we show that a novel short form of Lm-γ2 (Lm-γ2F) containing DIII is generated without requiring MMPs and chromosomal translocation between LAMC2 on chromosome 1 and NR6A1 gene locus on chromosome 9 in human ovarian cancer SKOV3 cells. Laminin-γ2F is expressed as a truncated form lacking domains I and II, which are essential for its association with Lm-α3 and -β3 chains of Lm-332. Secreted Lm-γ2F can act as an EGFR ligand activating the EGFR/AKT pathways more effectively than does the Lm-γ2 chain, which in turn promotes proliferation, survival, and motility of ovarian cancer cells. LAMC2-NR6A1 translocation was detected using in situ hybridization, and fusion transcripts were expressed in ovarian cancer cell tissues. Overexpression and suppression of fusion transcripts significantly increased and decreased the tumorigenic growth of cells in mouse models, respectively. To the best of our knowledge, this is the first report regarding a fusion gene of ECM showing that translocation of LAMC2 plays a crucial role in the malignant growth and progression of ovarian cancer cells and that the consequent product is a promising therapeutic target against ovarian cancers.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cocarcinogenesis/genetics
- Cocarcinogenesis/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Laminin/genetics
- Laminin/metabolism
- Mice, Inbred BALB C
- Mice, Nude
- Nuclear Receptor Subfamily 6, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Protein Subunits/genetics
- Protein Subunits/metabolism
- RNA Interference
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Hoshino Daisuke
- Division of Cancer Cell ResearchKanagawa Cancer Center Research InstituteYokohamaJapan
| | - Hisamori Kato
- Division of GynecologyKanagawa Cancer Center HospitalYokohamaJapan
| | - Kazuhiro Fukumura
- Division of Gene Expression MechanismInstitute for Comprehensive Medical ScienceFujita Health UniversityToyoakeJapan
| | - Akila Mayeda
- Division of Gene Expression MechanismInstitute for Comprehensive Medical ScienceFujita Health UniversityToyoakeJapan
| | - Yohei Miyagi
- Division of Molecular Pathology and GeneticsKanagawa Cancer Center Research InstituteYokohamaJapan
| | - Motoharu Seiki
- Division of Cancer Cell ResearchInstitute of Medical ScienceUniversity of TokyoTokyoJapan
| | - Naohiko Koshikawa
- Division of Cancer Cell ResearchKanagawa Cancer Center Research InstituteYokohamaJapan
- Division of Cancer Cell ResearchInstitute of Medical ScienceUniversity of TokyoTokyoJapan
- Department of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| |
Collapse
|
10
|
Horii T, Tsujimoto H, Hagiwara A, Isogai N, Sueyoshi Y, Oe Y, Kageyama S, Yoshida T, Kobayashi K, Minato H, Ueda J, Ichikawa H, Kawauchi A. Effects of Fiber Diameter and Spacing Size of an Artificial Scaffold on the In Vivo Cellular Response and Tissue Remodeling. ACS APPLIED BIO MATERIALS 2021; 4:6924-6936. [DOI: 10.1021/acsabm.1c00572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tsunehito Horii
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
- Division of Medical Life System, Department of Life and Medical Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Hiroyuki Tsujimoto
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
- Division of Medical Life System, Department of Life and Medical Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Akeo Hagiwara
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
- Division of Medical Life System, Department of Life and Medical Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Noritaka Isogai
- Department of Plastic and Reconstructive Surgery, Kindai University, Osaka 589-0014, Japan
| | - Yu Sueyoshi
- Department of Plastic and Reconstructive Surgery, Kindai University, Osaka 589-0014, Japan
| | - Yasumitsu Oe
- Department of Gastroenterology, Kusatsu General Hospital, Yabase, Kusatsu, Shiga 525-8585, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
| | - Tetsuya Yoshida
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
| | - Kenichi Kobayashi
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
| | - Hiroshi Minato
- Department of Surgery, Yawata Chuo Hospital, Yawatagotanda, Yawata, Kyoto 614-8071, Japan
| | - Joe Ueda
- Department of Gastroenterology, Ueda Clinic, Kitanakaieshita, Takanosu, Akita 018-3331, Japan
| | - Hiroshi Ichikawa
- Division of Medical Life System, Department of Life and Medical Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Seta Tsukinowa, Otsu, Shiga 610-0321, Japan
| |
Collapse
|
11
|
Trac N, Chung EJ. Overcoming physiological barriers by nanoparticles for intravenous drug delivery to the lymph nodes. Exp Biol Med (Maywood) 2021; 246:2358-2371. [PMID: 33957802 DOI: 10.1177/15353702211010762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The lymph nodes are major sites of cancer metastasis and immune activity, and thus represent important clinical targets. Although not as well-studied compared to subcutaneous administration, intravenous drug delivery is advantageous for lymph node delivery as it is commonly practiced in the clinic and has the potential to deliver therapeutics systemically to all lymph nodes. However, rapid clearance by the mononuclear phagocyte system, tight junctions of the blood vascular endothelium, and the collagenous matrix of the interstitium can limit the efficiency of lymph node drug delivery, which has prompted research into the design of nanoparticle-based drug delivery systems. In this mini review, we describe the physiological and biological barriers to lymph node targeting, how they inform nanoparticle design, and discuss the future outlook of lymph node targeting.
Collapse
Affiliation(s)
- Noah Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, Los Angeles, CA 90033, USA.,Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
12
|
AboYoussef AM, Khalaf MM, Malak MN, Hamzawy MA. Repurposing of sildenafil as antitumour; induction of cyclic guanosine monophosphate/protein kinase G pathway, caspase-dependent apoptosis and pivotal reduction of Nuclear factor kappa light chain enhancer of activated B cells in lung cancer. J Pharm Pharmacol 2021; 73:1080-1091. [PMID: 33856030 DOI: 10.1093/jpp/rgab049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Lung cancer is one of the most frequent types of cancers that lead to death. Sildenafil is a potent inhibitor of phosphodiesterase-5 and showed potential anticancer effects, which has not yet been fully evaluated. Thus, this study aims to investigate the potential anticancer effect of sildenafil in urethane-induced lung cancer in BALB/c mice. METHODS Five-week-old male BALB/c mice were treated with either (i) normal saline only, (ii) sildenafil only 50 mg kg-1/ P.O every other day for the last four successive weeks, (iii) urethane 1.5 gm kg-1 i.p (at day 1 and day 60), (iv) carboplatin after urethane induction, or (v) sildenafil after urethane induction. KEY FINDINGS It was shown that sildenafil significantly increased the levels of cGMP and Caspase-3 with a reduction of NF-κB, Bcl-2, Cyclin D1, intercellular adhesion molecule 1, matrix metalloproteinase-2 levels and normalisation of Nrf2 along with pronounced improvement in the histological patterns. CONCLUSIONS These results indicated that sildenafil markedly induces cell cycle arrest, apoptosis and inhibits the metastatic activity through activation of cyclic guanosine monophosphate/protein kinase G pathway and down-regulation of cyclin D1 and nuclear factor kappa light chain enhancer of activated B cells with downstream anti-apoptotic gene Bcl-2, which underscores the critical importance of future using sildenafil in the treatment of lung cancer.
Collapse
Affiliation(s)
- Amira M AboYoussef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
13
|
Wang H, Qi C, Wan D. MicroRNA-377-3p targeting MMP-16 inhibits ovarian cancer cell growth, invasion, and interstitial transition. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:124. [PMID: 33569426 PMCID: PMC7867897 DOI: 10.21037/atm-20-8027] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background To evaluate role of microRNA (miRNA)-377-3p on the remission of ovarian cancer (OC) cell proliferation, invasion, and interstitial transition in vivo and vitro. Methods SKOV3 cells were used as the object of in vitro research and four-week-old immunodeficient BABL/c female nude mice were used to form the xenograft model. Cell models were constructed by transfecting NC mimics, miR-377 mimic, plasmid cloning DNA (pcDNA), pc-matrix metalloproteinase (MMP)-16, or co-transfecting miR-377 mimic and pc-MMP-16. TargetScan software was used to predict the targeting relationship between miRNA-377-3p and MMP-16 in OC cells. The combination of miRNA-377-3p and MMP-16 was detected by dual luciferase report experiment. miRNA expression levels of miRNA-377-3p and MMP-16 in each transfection group cells were detected by reverse transcription-polymerase chain reaction (RT-PCR). The proliferation of SKOV3 cells were assessed by 5-ethynyl-2'-deoxyuridine (EdU) staining and microtubule formation, while the invasion ability of SKOV3 cells was detected by Transwell assay. Protein expression levels of MMP-16, survivin, Ki67, vascular endothelial growth factor (VEGF), E-cadherin, and N-cadherin were detected by Western blot (WB), and the positive cells of Ki67 and VEGF were detected by immunohistochemistry (IHC). Results MMP-16 overexpression markedly increased the EDU-positive cell percentage, upregulated survivin and Ki67 levels, increased the number of invasive cells per field, and enhanced VEGF and N-cadherin expression. Importantly, co-transfection of miRNA-377-3p and MMP-16 reversed these abnormal phenomena. Xenotransplantation mouse models were formed by injecting SKOV-3 cells subcutaneously. Tumor size, tumor volume, and tumor weight were all reduced in the miR-377-3p mimic–transfected group. The results of IHC indicated that Ki67 and VEGF expression were decreased in the miR-377-3p mimic–transfected group. Conclusions These findings indicate that miR-377-3p could be a promising therapeutic agent for OC cell growth, invasion, and interstitial transition with MMP-16 being its likely target.
Collapse
Affiliation(s)
- Huabin Wang
- Department of Obstetrics and Gynecology, Eastern Hospital, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Changmin Qi
- Department of Cardiac Surgery Centre, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Wan
- Department of Gynecology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
14
|
Hayn A, Fischer T, Mierke CT. Inhomogeneities in 3D Collagen Matrices Impact Matrix Mechanics and Cancer Cell Migration. Front Cell Dev Biol 2020; 8:593879. [PMID: 33251219 PMCID: PMC7674772 DOI: 10.3389/fcell.2020.593879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cell motility under physiological and pathological conditions including malignant progression of cancer and subsequent metastasis are founded on environmental confinements. During the last two decades, three-dimensional cell migration has been studied mostly by utilizing biomimetic extracellular matrix models. In the majority of these studies, the in vitro collagen scaffolds are usually assumed to be homogenous, as they consist commonly of one specific type of collagen, such as collagen type I, isolated from one species. These collagen matrices should resemble in vivo extracellular matrix scaffolds physiologically, however, mechanical phenotype and functional reliability have been addressed poorly due to certain limitations based on the assumption of homogeneity. How local variations of extracellular matrix structure impact matrix mechanics and cell migration is largely unknown. Here, we hypothesize that local inhomogeneities alter cell movement due to alterations in matrix mechanics, as they frequently occur in in vivo tissue scaffolds and were even changed in diseased tissues. To analyze the effect of structural inhomogeneities on cell migration, we used a mixture of rat tail and bovine dermal collagen type I as well as pure rat and pure bovine collagens at four different concentrations to assess three-dimensional scaffold inhomogeneities. Collagen type I from rat self-assembled to elongated fibrils, whereas bovine collagen tended to build node-shaped inhomogeneous scaffolds. We have shown that the elastic modulus determined with atomic force microscopy in combination with pore size analysis using confocal laser scanning microscopy revealed distinct inhomogeneities within collagen matrices. We hypothesized that elastic modulus and pore size govern cancer cell invasion in three-dimensional collagen matrices. In fact, invasiveness of three breast cancer cell types is altered due to matrix-type and concentration indicating that these two factors are crucial for cellular invasiveness. Our findings revealed that local matrix scaffold inhomogeneity is another crucial parameter to explain differences in cell migration, which not solely depended on pore size and stiffness of the collagen matrices. With these three distinct biophysical parameters, characterizing structure and mechanics of the studied collagen matrices, we were able to explain differences in the invasion behavior of the studied cancer cell lines in dependence of the used collagen model.
Collapse
Affiliation(s)
- Alexander Hayn
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| | - Tony Fischer
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| |
Collapse
|
15
|
Tinospora cordifolia and arabinogalactan exert chemopreventive action during benzo(a)pyrene-induced pulmonary carcinogenesis: studies on ultrastructural, molecular, and biochemical alterations. Eur J Cancer Prev 2020; 30:21-39. [PMID: 33122541 DOI: 10.1097/cej.0000000000000595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to unveil the chemopreventive potentials of aqueous Tinospora cordifolia stem extract and its active component viz. Arabinogalactan against Benzo(a)pyrene-induced pulmonary carcinogenesis. Animals were divided into six groups: (I) Control, (II) aqueous Tinospora cordifolia (200 mg/kg b.wt, p.o.), (III) arabinogalactan (7.5 mg/kg b.wt, p.o.), (IV) benzo(a)pyrene (50 mg/kg b.wt, i.p.) at second and fourth week of study, (V) benzo(a)pyrene + aqueous Tinospora cordifolia, and (VI) benzo(a)pyrene + arabinogalactan. The benzo(a)pyrene treatment resulted in severe alterations in the cellular arrangement and morphology of the alveolar tissue in benzo(a)pyrene group. However, benzo(a)pyrene + aqueous Tinospora cordifolia and benzo(a)pyrene + arabinogalactan groups revealed classical features of apoptosis including chromatin condensation and formation of apoptotic bodies. Furthermore, Fourier transform Infrared spectroscopy analysis showed disturbed phospholipid saturation and protein secondary structures in benzo(a)pyrene treated animals. Depletion in relative glycogen and enhancement in total nucleic acid content was observed in benzo(a)pyrene treated animals, and the same was found to be restored upon arabinogalactan and aqueous Tinospora cordifolia supplementation. Benzo(a)pyrene insult also upregulated the phase I carcinogen metabolizing enzymes and differentially modulated the phase II metabolizing enzymes during pulmonary carcinogenesis. Also, depleted (reduced glutathione) and increased lipid peroxidation levels were observed in benzo(a)pyrene treated animals, which was found to be normalized upon aqueous Tinospora cordifolia and arabinogalactan administration. Clastogenic damage inflicted by benzo(a)pyrene was also reversed in benzo(a)pyrene + aqueous Tinospora cordifolia and benzo(a)pyrene + arabinogalactan group. Thus, the present study infers that aqueous Tinospora cordifolia and arabinogalactan showed promising anticancer activity against lung tumorigenesis in terms of ultrastructural, biochemical, and biomolecular aspects.
Collapse
|
16
|
Mierke CT. Mechanical Cues Affect Migration and Invasion of Cells From Three Different Directions. Front Cell Dev Biol 2020; 8:583226. [PMID: 33043017 PMCID: PMC7527720 DOI: 10.3389/fcell.2020.583226] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Cell migration and invasion is a key driving factor for providing essential cellular functions under physiological conditions or the malignant progression of tumors following downward the metastatic cascade. Although there has been plentiful of molecules identified to support the migration and invasion of cells, the mechanical aspects have not yet been explored in a combined and systematic manner. In addition, the cellular environment has been classically and frequently assumed to be homogeneous for reasons of simplicity. However, motility assays have led to various models for migration covering only some aspects and supporting factors that in some cases also include mechanical factors. Instead of specific models, in this review, a more or less holistic model for cell motility in 3D is envisioned covering all these different aspects with a special emphasis on the mechanical cues from a biophysical perspective. After introducing the mechanical aspects of cell migration and invasion and presenting the heterogeneity of extracellular matrices, the three distinct directions of cell motility focusing on the mechanical aspects are presented. These three different directions are as follows: firstly, the commonly used invasion tests using structural and structure-based mechanical environmental signals; secondly, the mechano-invasion assay, in which cells are studied by mechanical forces to migrate and invade; and thirdly, cell mechanics, including cytoskeletal and nuclear mechanics, to influence cell migration and invasion. Since the interaction between the cell and the microenvironment is bi-directional in these assays, these should be accounted in migration and invasion approaches focusing on the mechanical aspects. Beyond this, there is also the interaction between the cytoskeleton of the cell and its other compartments, such as the cell nucleus. In specific, a three-element approach is presented for addressing the effect of mechanics on cell migration and invasion by including the effect of the mechano-phenotype of the cytoskeleton, nucleus and the cell's microenvironment into the analysis. In precise terms, the combination of these three research approaches including experimental techniques seems to be promising for revealing bi-directional impacts of mechanical alterations of the cellular microenvironment on cells and internal mechanical fluctuations or changes of cells on the surroundings. Finally, different approaches are discussed and thereby a model for the broad impact of mechanics on cell migration and invasion is evolved.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Human Schlafen 5 regulates reversible epithelial and mesenchymal transitions in breast cancer by suppression of ZEB1 transcription. Br J Cancer 2020; 123:633-643. [PMID: 32488136 PMCID: PMC7435190 DOI: 10.1038/s41416-020-0873-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/17/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Human Schlafen 5 (SLFN5) has been reported to inhibit or promote cell invasion in tumours depending on their origin. However, its role in breast cancer (BRCA) is undetermined. METHODS Differential expression analyses using The Cancer Genome Atlas (TCGA) data, clinical samples and cell lines were performed. Lentiviral knockdown and overexpression experiments were performed to detect changes in cell morphology, molecular markers and invasion. Chromatin immunoprecipitation-sequencing (ChIP-Seq) and luciferase reporter assays were performed to detect the SLFN5-binding motif. RESULTS TCGA, clinical samples and cell lines showed that SLFN5 expression was negatively correlated with BRCA metastasis. SLFN5 knockdown induced epithelial-mesenchymal transition (EMT) and enhanced invasion in BRCA cell lines. However, overexpression triggered mesenchymal-epithelial transition (MET). SLFN5 inhibited the expression of ZEB1 but not ZEB2, SNAI1, SNAI2, TWIST1 or TWIST2. Knockdown and overexpression of ZEB1 indicated that it was a mediator of the SLFN5-governed phenotype and invasion changes. Moreover, SLFN5 inhibited ZEB1 transcription by directly binding to the SLFN5-binding motif on the ZEB1 promoter, but a SLFN5 C-terminal deletion mutant did not. CONCLUSION SLFN5 regulates reversible epithelial and mesenchymal transitions, and inhibits BRCA metastasis by suppression of ZEB1 transcription, suggesting that SLFN5 could be a potential target for BRCA therapy.
Collapse
|
18
|
Park S, Lim S, Siriviriyakul P, Jeon JS. Three-dimensional pore network characterization of reconstructed extracellular matrix. Phys Rev E 2020; 101:052414. [PMID: 32575345 DOI: 10.1103/physreve.101.052414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/26/2020] [Indexed: 06/11/2023]
Abstract
The extracellular matrix (ECM) has a fiber network that provides physical scaffolds to cells and plays important roles by regulating cellular functions. Some previous works characterized the mechanical and geometrical properties of the ECM fiber network using reconstituted collagen-I. However, the characterization of the porous structure of reconstituted collagen-I has been limited to the pore diameter measurement, and pore network extraction has not been applied to reconstituted collagen-I despite the importance of pore interconnectivity. Here, we aim to show the importance of characterizing the pore network of reconstituted collagen-I by comparing the pore networks of structures that have different fiber alignments. We show that the fiber alignment significantly changes the pore throat area but not the pore diameter. Also, we demonstrate that larger pore throats are directed in the direction of the fiber alignment, which may help in understanding the enhanced cell migration when fibers are aligned.
Collapse
Affiliation(s)
- Seongjin Park
- Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Seongjin Lim
- Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Pan Siriviriyakul
- Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Jessie S Jeon
- Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
19
|
Dominijanni A, Mazzocchi A, Shelkey E, Forsythe S, Devarsetty M, Soker S. Bioengineered Tumor Organoids. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 13:168-173. [PMID: 32432209 DOI: 10.1016/j.cobme.2020.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances in biofabrication technologies, such as cell culture systems, and biomaterials have led to the development of three-dimensional (3D) cell culture platforms, such as tumor organoids. Tumor organoids are more physiologically accurate to the in vivo system, which they are intended to model, compared with traditional 2D cancer cell culture systems. Tumor organoids can mimic pathological and physical characteristics of tumors as well as maintain genetic stability of the cancer cells. Furthermore tumor organoids have advantage over animal models, being made from human cells and easily controlled in the laboratory to attain the desired tissue characteristics. In this section, we describe general tumor organoid technologies, the importance of the tumor microenvironment (TME) in model culture systems, and the use of tumor organoids in drug development and precision medicine. Organoid technologies continue to develop rapidly for applications in academic, clinical, and pharmaceutical settings.
Collapse
Affiliation(s)
- Anthony Dominijanni
- Wake Forest School of Medicine; Wake Forest Institute for Regenerative Medicine; Winston-Salem, NC 27103
| | - Andrea Mazzocchi
- Wake Forest School of Medicine; Wake Forest Institute for Regenerative Medicine; Winston-Salem, NC 27103
| | - Ethan Shelkey
- Wake Forest School of Medicine; Wake Forest Institute for Regenerative Medicine; Winston-Salem, NC 27103
| | - Steven Forsythe
- Wake Forest School of Medicine; Wake Forest Institute for Regenerative Medicine; Winston-Salem, NC 27103
| | - Mahesh Devarsetty
- Wake Forest School of Medicine; Wake Forest Institute for Regenerative Medicine; Winston-Salem, NC 27103
| | - Shay Soker
- Wake Forest School of Medicine; Wake Forest Institute for Regenerative Medicine; Winston-Salem, NC 27103
| |
Collapse
|
20
|
Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, Hynes RO, Jain RK, Janowitz T, Jorgensen C, Kimmelman AC, Kolonin MG, Maki RG, Powers RS, Puré E, Ramirez DC, Scherz-Shouval R, Sherman MH, Stewart S, Tlsty TD, Tuveson DA, Watt FM, Weaver V, Weeraratna AT, Werb Z. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer 2020; 20:174-186. [PMID: 31980749 PMCID: PMC7046529 DOI: 10.1038/s41568-019-0238-1] [Citation(s) in RCA: 2037] [Impact Index Per Article: 509.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are a key component of the tumour microenvironment with diverse functions, including matrix deposition and remodelling, extensive reciprocal signalling interactions with cancer cells and crosstalk with infiltrating leukocytes. As such, they are a potential target for optimizing therapeutic strategies against cancer. However, many challenges are present in ongoing attempts to modulate CAFs for therapeutic benefit. These include limitations in our understanding of the origin of CAFs and heterogeneity in CAF function, with it being desirable to retain some antitumorigenic functions. On the basis of a meeting of experts in the field of CAF biology, we summarize in this Consensus Statement our current knowledge and present a framework for advancing our understanding of this critical cell type within the tumour microenvironment.
Collapse
Affiliation(s)
- Erik Sahai
- The Francis Crick Institute, London, UK.
| | - Igor Astsaturov
- Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Edna Cukierman
- Cancer Biology Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - David G DeNardo
- Division of Oncology, Washington University Medical School, St Louis, MO, USA
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Douglas Fearon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rakesh K Jain
- Edwin L Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Northwell Health Cancer Institute, New Hyde Park, NY, USA
| | - Claus Jorgensen
- Cancer Research UK Manchester Institute, University of Manchester, Nether Alderley, UK
| | - Alec C Kimmelman
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY, USA
| | - Mikhail G Kolonin
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Robert G Maki
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Northwell Health Cancer Institute, New York, NY, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - R Scott Powers
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel C Ramirez
- Zucker School of Medicine at Hofstra/Northwell Health System, New York, NY, USA
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Mara H Sherman
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sheila Stewart
- Department of Cell Biology and Physiology, Department of Medicine, ICCE Institute, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA
| | - Thea D Tlsty
- UCSF Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
- Department of Pathology, UCSF, San Francisco, CA, USA
| | | | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - Valerie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Ashani T Weeraratna
- Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
21
|
Ferreira HH, Toledo DC, Faria AM, Araújo DP, Nardi ABD, Porto RNG, Moura VMBDD. Immunoexpression of metalloproteinases 9 (MMP-9) and 2 (MMP-2) and their inhibitors (TIMP-1 and TIMP-2) in normal and neoplastic canine mammary tissue. CIÊNCIA ANIMAL BRASILEIRA 2020. [DOI: 10.1590/1809-6891v21e-58640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Abstract The aim of this study was to perform the immunostaining of MMP-9 and MMP-2 and its inhibitors, TIMP-1 and TIMP-2, on normal and neoplastic canine mammary tissue in order to evaluate the behavior of these proteins in extracellular matrix (ECM) remodeling in different neoplastic mammary types. Thus, 48 samples of canine mammary tissue were analyzed, 14 of which complex carcinomas, 13 tubulopapillary carcinomas, six single adenomas and 15 normal mammary tissue. There were differences in MMP-9, TIMP-1 and TIMP-2 according to mammary histomorphology, and MMP-9 presented increased immunoexpression in epithelial and stromal cells in tubulopapillary and complex carcinomas. TIMP-1 exhibited reduced immunostaining in the stromal cells of the complex carcinomas and TIMP-2 enhanced immunostaining in the epithelial cells of tubulopapillary carcinomas. There was a positive correlation between MMP-9 and TIMP-1 in epithelial and stromal cells regarding immunostaining intensity and number of labeled cells in the normal breast. There was a positive correlation between MMP-9 and TIMP-2 in the epithelial cells of tubulopapillary carcinomas. It is concluded that balanced activity between MMP-9, MMP-2, TIMP-1 and TIMP-2 maintains normal canine mammary tissue homeostasis while increased immunoexpression of MMP-9 and TIMP-2 and reduced TIMP- 1 in carcinomas suggest a favorable condition for tumor evolution.
Collapse
|
22
|
Yang L, Yang S, Li X, Li B, Li Y, Zhang X, Ma Y, Peng X, Jin H, Fan Q, Wei S, Liu J, Li H. Tumor organoids: From inception to future in cancer research. Cancer Lett 2019; 454:120-133. [PMID: 30981763 DOI: 10.1016/j.canlet.2019.04.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 12/18/2022]
Abstract
Tumor models have created new avenues for personalized medicine and drug development. A new culture model derived from a three-dimensional system, the tumor organoid, is gradually being used in many fields. An organoid can simulate the physiological structure and function of tissue in situ and maintain the characteristics of tumor cells in vivo, overcoming the disadvantages of traditional experimental tumor models. Organoids can mimic pathological features of tumors and maintain genetic stability, making them suitable for both molecular mechanism studies and pharmacological experiments of clinical transformation. In addition, the application of tumor organoids combined with other technologies, such as liquid biopsy technology, microraft array (MRA), and high-content screening (HCS), for the development of personalized diagnosis and cancer treatment has a promising future. In this review, we introduce the evolution of organoids and discuss their specific application and advantages. We also summarize the characteristics of several tumor organoids culture systems.
Collapse
Affiliation(s)
- Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Bowen Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Xiaodong Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Qing Fan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110000, PR China.
| |
Collapse
|
23
|
Rady H, Salem S, Ez El-Arab M. Primmorph extracts and mesohyls of marine sponges inhibit proliferation and migration of hepatocellular carcinoma cells in vitro. J Pharm Anal 2019; 9:284-291. [PMID: 31452967 PMCID: PMC6704043 DOI: 10.1016/j.jpha.2019.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 01/01/2023] Open
Abstract
Cancer recurrence and severe side effects of currently being used chemotherapeutic agents reduce their clinical efficacy. Thus, there is a constant need to develop alternative anticancer drugs. Sustainable supply is an important challenge facing marine-based drug discovery. Primmorph, a 3D cell culture system, could provide a sustainable source to produce metabolites for anticancer drugs from marine sponges. In the present work, the anticancer activity of primmorph extracts and mesohyls of Negombata magnifica, Hemimycle arabica, Crella spinulata, and Stylissa carteri sponges was evaluated. Antiproliferative activity was studied in terms of cytotoxicity, colony formation, cell cycle, and apoptosis. Migration was assessed by migration assay and matrix metalloproteinase activity. The expression of proliferation and migration-related genes was analyzed using real time PCR. Migration and proliferation activities of HepG2 cells were inhibited by treatment with primmorph extracts and mesohyls of N. magnifica, H. arabica, and C. spinulata. The mesohyl of S. carteri did not show any anticancer activity although the primmorph extract led to cell cycle arrest. Among the selected sponge species, the primmorph extract of C. spinulata was the most promising anticancer agent regarding antiproliferative and antimigratory activities. In addition, primmorph extracts have the advantage of working under well-defined and controlled conditions, which allows the easy application as a bioreactor.
Collapse
Affiliation(s)
- Hanaa Rady
- Chemistry of Natural Compound Department, National Research Centre, Cairo, Egypt
| | - Sohair Salem
- Molecular Genetics and Enzymology Department, National Research Centre, Cairo, Egypt
| | - Mohamed Ez El-Arab
- National Institute of Oceanography and Fisheries (NIOF), Hurghada, Egypt
| |
Collapse
|
24
|
MMP-1 Over-expression Promotes Malignancy and Stem-Like Properties of Human Osteosarcoma MG-63 Cells In Vitro. Curr Med Sci 2018; 38:809-817. [PMID: 30594980 DOI: 10.1007/s11596-018-1947-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/10/2018] [Indexed: 12/28/2022]
Abstract
Osteosarcoma is the most common primary malignant bone tumor in childhood, and it maintains a high level of recurrence. Matrix metalloproteinase-1 (MMP-1) was found to contribute to cancer progression. The present study was to investigate the in vitro effects of MMP-1 over-expression on the proliferation, invasion, metastasis and stem-like properties of osteosarcoma MG-63 cells. The MG-63 cells were cultured and had a full length MMP-1 cDNA inserted by the lentiviral vector (MG-63MMP-1+). MG-63 negative control and MG-63 blank control groups were established as well. MMP-1 expression was detected in MG-63MMP-1+, MG-63 negative control and MG-63 blank control cells using qPCR, Western blotting and immunofluorescence after 24 h of culture. The cell proliferation assay was performed with a camera attached to a bioreactor, which was programmed to photograph five regions of each well every 10 min over a period of 48 h. The cell invasion assay was conducted with Matrigel to assess the invasive potential of MG-63 cells over 24 h, the qPCR analysis to measure stem cell markers, including Oct4, Sox-2, Nanog, and Pax-7, and Western blot analysis to detect invasive and metastatic potential markers TIMP-1, VEGF and BMP2/4, after 24 h of culture. Immunofluorescence was used to investigate the presence of the stem cell marker Pax-7 after 24-h culture. The results showed that over-expression of MMP-1 after transfection could significantly increase tumor cell proliferation and invasion (P<0.05, MG-63MMP-1+versus controls). Pax-7 was highly expressed in MG-63MMP-1+ cells, with no significant changes of Oct-4, Sox-2, and Nanog observed (P<0.05). MG-63MMP-1+ cells showed higher expression of VEGF and BMP 2/4 proteins and lower expression of TIMP-1 protein than controls (P<0.05). It was concluded that MMP-1 over-expression in MG-63 cells contributed to the proliferation, invasion, metastasis and stem-like properties of osteosarcoma cells. Future studies should focus on in vivo effects of MMP-1 over-expression and the application of MMP-1 and Pax-7 inhibition in vivo to osteosarcoma therapies.
Collapse
|
25
|
Puls TJ, Tan X, Husain M, Whittington CF, Fishel ML, Voytik-Harbin SL. Development of a Novel 3D Tumor-tissue Invasion Model for High-throughput, High-content Phenotypic Drug Screening. Sci Rep 2018; 8:13039. [PMID: 30158688 PMCID: PMC6115445 DOI: 10.1038/s41598-018-31138-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/13/2018] [Indexed: 12/22/2022] Open
Abstract
While much progress has been made in the war on cancer, highly invasive cancers such as pancreatic cancer remain difficult to treat and anti-cancer clinical trial success rates remain low. One shortcoming of the drug development process that underlies these problems is the lack of predictive, pathophysiologically relevant preclinical models of invasive tumor phenotypes. While present-day 3D spheroid invasion models more accurately recreate tumor invasion than traditional 2D models, their shortcomings include poor reproducibility and inability to interface with automated, high-throughput systems. To address this gap, a novel 3D tumor-tissue invasion model which supports rapid, reproducible setup and user-definition of tumor and surrounding tissue compartments was developed. High-cell density tumor compartments were created using a custom-designed fabrication system and standardized oligomeric type I collagen to define and modulate ECM physical properties. Pancreatic cancer cell lines used within this model showed expected differential invasive phenotypes. Low-passage, patient-derived pancreatic cancer cells and cancer-associated fibroblasts were used to increase model pathophysiologic relevance, yielding fibroblast-mediated tumor invasion and matrix alignment. Additionally, a proof-of-concept multiplex drug screening assay was applied to highlight this model's ability to interface with automated imaging systems and showcase its potential as a predictive tool for high-throughput, high-content drug screening.
Collapse
Affiliation(s)
- T J Puls
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiaohong Tan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Mahera Husain
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Catherine F Whittington
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Oncology, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Melissa L Fishel
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN, 46202, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
26
|
Jie Z, Xie Z, Zhao X, Sun X, Yu H, Pan X, Shen S, Qin A, Fang X, Fan S. Glabridin inhibits osteosarcoma migration and invasion via blocking the p38- and JNK-mediated CREB-AP1 complexes formation. J Cell Physiol 2018; 234:4167-4178. [PMID: 30146723 DOI: 10.1002/jcp.27171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/17/2018] [Indexed: 01/11/2023]
Abstract
Osteosarcoma is the most common bone malignancy, and it seriously affects the quality of life of affected children and adolescents. Glabridin (GLA), a major component of licorice root extract, has been reported to exert antitumor effects against a variety of tumor types; however, its effects on osteosarcoma have not been elucidated. In the current study, we investigate the effects and potential antimetastatic mechanisms of GLA on osteosarcoma in vitro and in vivo. Flow cytometry showed that GLA induced G2/M cell cycle phase arrest and promoted cell apoptosis. Transwell and wound-healing assays showed that GLA significantly decreased the migration and invasion of osteosarcoma cells. Further western blotting and quantitative real-time polymerase chain reaction showed that the expression of matrix metalloproteinase (MMP)-2 and MMP-9 in MG63 and HOS cells were reduced after GLA treatment. Moreover, western blotting demonstrated that GLA downregulated the phosphorylation of p38 mitogen-activated protein kinases and c-Jun N-terminal kinase. A coimmunoprecipitation assay illustrated that formation of cAMP response element-binding protein (CREB)-activating protein 1 (AP1) complexes and the DNA binding activities of CREB and AP1 in MG63 and HOS cells were impaired following treatment with GLA. Finally, GLA inhibited tumor growth and suppressed osteosarcoma cell metastasis in vivo. Overall, our findings highlight the potential of GLA as a therapeutic agent for the prevention and treatment of tumor metastasis.
Collapse
Affiliation(s)
- Zhiwei Jie
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Ziang Xie
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Xuewu Sun
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Hejun Yu
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Pan
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Shuying Shen
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai, China
| | - Xiangqian Fang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shunwu Fan
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Erickson AE, Levengood SKL, Sun J, Chang FC, Zhang M. Fabrication and Characterization of Chitosan-Hyaluronic Acid Scaffolds with Varying Stiffness for Glioblastoma Cell Culture. Adv Healthc Mater 2018; 7:e1800295. [PMID: 29893067 PMCID: PMC6116517 DOI: 10.1002/adhm.201800295] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/05/2018] [Indexed: 12/19/2022]
Abstract
The invasive and recurrent nature of glioblastoma multiforme (GBM) is linked to a small subpopulation of cancer cells, which are self-renewing, resistant to standard treatment regimens, and induce formation of new tumors. Matrix stiffness is implicated in the regulation of cell proliferation, drug resistance, and reversion to a more invasive phenotype. Therefore, understanding the relationship between matrix stiffness and tumor cell behavior is vital to develop appropriate in vitro tumor models. Here, chitosan-hyaluronic acid (CHA) polyelectrolyte complex scaffolds are fabricated with statistically significant stiffness variances to characterize the effect of scaffold stiffness on morphology, proliferation, drug resistance, and gene expression in human glioblastoma cells (U-87 MG). All scaffolds support GBM proliferation over a 12-day culture period, yet larger spheroids are observed in scaffolds with higher stiffness. Additionally, GBM cells cultured in stiffer CHA scaffolds prove significantly more resistant to the common chemotherapeutic temozolomide. Moreover, the stiffer 8% CHA scaffolds exhibit an increase in expression of drug resistance and invasion related genes compared to 2D culture. CHA scaffolds present a tunable microenvironment for enhanced tumor cell malignancy and may provide a valuable in vitro microenvironment for studying tumor progression and screening anticancer therapies.
Collapse
Affiliation(s)
- Ariane E. Erickson
- Department of Materials Science & Engineering, University of Washington, Seattle, Washington 98195, USA,
| | - Sheeny K. Lan Levengood
- Department of Materials Science & Engineering, University of Washington, Seattle, Washington 98195, USA,
| | - Jialu Sun
- Department of Materials Science & Engineering, University of Washington, Seattle, Washington 98195, USA,
| | - Fei-Chien Chang
- Department of Materials Science & Engineering, University of Washington, Seattle, Washington 98195, USA,
| | - Miqin Zhang
- Department of Materials Science & Engineering, University of Washington, Seattle, Washington 98195, USA,
| |
Collapse
|
28
|
Actin-Based Cell Protrusion in a 3D Matrix. Trends Cell Biol 2018; 28:823-834. [PMID: 29970282 PMCID: PMC6158345 DOI: 10.1016/j.tcb.2018.06.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Cell migration controls developmental processes (gastrulation and tissue patterning), tissue homeostasis (wound repair and inflammatory responses), and the pathobiology of diseases (cancer metastasis and inflammation). Understanding how cells move in physiologically relevant environments is of major importance, and the molecular machinery behind cell movement has been well studied on 2D substrates, beginning over half a century ago. Studies over the past decade have begun to reveal the mechanisms that control cell motility within 3D microenvironments – some similar to, and some highly divergent from those found in 2D. In this review we focus on migration and invasion of cells powered by actin, including formation of actin-rich protrusions at the leading edge, and the mechanisms that control nuclear movement in cells moving in a 3D matrix. Cell migration has been well studied in 2D, but how this relates to movement in physiological 3D tissues and matrix is not clear, particularly in vertebrate interstitial matrix. In 3D matrix cells actin polymerisation directly contributes to the formation of lamellipodia to facilitate migration and invasion (mesenchymal movement), analogous to 2D migration; actomyosin contractility promotes bleb formation to indirectly promote protrusion (amoeboid movement). Mesenchymal migration can be characterised by polymerisation of actin to form filopodial protrusions, in the absence of lamellipodia. Translocation of the nucleus is emerging as a critical step due to the constrictive environment of 3D matrices, and the mechanisms that transmit force to the nucleus and allow movement are beginning to be uncovered.
Collapse
|
29
|
Mohammadi H, Sahai E. Mechanisms and impact of altered tumour mechanics. Nat Cell Biol 2018; 20:766-774. [PMID: 29950570 DOI: 10.1038/s41556-018-0131-2] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
Abstract
The physical characteristics of tumours are intricately linked to the tumour phenotype and difficulties during treatment. Many factors contribute to the increased stiffness of tumours; from increased matrix deposition, matrix remodelling by forces from cancer cells and stromal fibroblasts, matrix crosslinking, increased cellularity, and the build-up of both solid and interstitial pressure. Increased stiffness then feeds back to increase tumour invasiveness and reduce therapy efficacy. Increased understanding of this interplay is offering new therapeutic avenues.
Collapse
|
30
|
Alkasalias T, Moyano-Galceran L, Arsenian-Henriksson M, Lehti K. Fibroblasts in the Tumor Microenvironment: Shield or Spear? Int J Mol Sci 2018; 19:ijms19051532. [PMID: 29883428 PMCID: PMC5983719 DOI: 10.3390/ijms19051532] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/11/2022] Open
Abstract
Tumorigenesis is a complex process involving dynamic interactions between malignant cells and their surrounding stroma, including both the cellular and acellular components. Within the stroma, fibroblasts represent not only a predominant cell type, but also a major source of the acellular tissue microenvironment comprising the extracellular matrix (ECM) and soluble factors. Normal fibroblasts can exert diverse suppressive functions against cancer initiating and metastatic cells via direct cell-cell contact, paracrine signaling by soluble factors, and ECM integrity. The loss of such suppressive functions is an inherent step in tumor progression. A tumor cell-induced switch of normal fibroblasts into cancer-associated fibroblasts (CAFs), in turn, triggers a range of pro-tumorigenic signals accompanied by distraction of the normal tissue architecture, thus creating an optimal niche for cancer cells to grow extensively. To further support tumor progression and metastasis, CAFs secrete factors such as ECM remodeling enzymes that further modify the tumor microenvironment in combination with the altered adhesive forces and cell-cell interactions. These paradoxical tumor suppressive and promoting actions of fibroblasts are the focus of this review, highlighting the heterogenic molecular properties of both normal and cancer-associated fibroblasts, as well as their main mechanisms of action, including the emerging impact on immunomodulation and different therapy responses.
Collapse
Affiliation(s)
- Twana Alkasalias
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
- Department of Biology, College of Science, Salahaddin University, Irbil 44002, Kurdistan-Iraq.
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
| | - Marie Arsenian-Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden.
- Research Programs Unit, Genome-Scale Biology and Medicum, University of Helsinki, and Helsinki University Hospital, P.O. Box 63, FI-00014 Helsinki, Finland.
| |
Collapse
|
31
|
Itoh Y. Discoidin domain receptors: Microenvironment sensors that promote cellular migration and invasion. Cell Adh Migr 2018; 12:378-385. [PMID: 29671358 PMCID: PMC6363040 DOI: 10.1080/19336918.2018.1460011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular matrix (ECM) provides cells scaffolding for cell migration and microenvironment for various cellular functions. Collagens are major ECM components in tissue and discoidin domain receptors (DDRs) are receptor tyrosine kinases (RTK) that recognise fibrillar collagens. Unlike other RTK, their ligands are solid ECM the that are abundantly present in the pericellular environment in various tissue, and thus its activation and regulations are unique amongst RTK family. It is emerging that DDRs may be the sensors that monitor and detects changes in ECM microenvironment and determines the cellular fates upon tissue injuries. In this mini-review, recent findings on the role of DDRs as microenvironment sensor and their roles in cell migration and invasion are discussed.
Collapse
Affiliation(s)
- Yoshifumi Itoh
- a Kennedy Institute of Rheumatology, University of Oxford , Roosevelt Drive, Headington , Oxford , UK
| |
Collapse
|
32
|
Malandrino A, Mak M, Kamm RD, Moeendarbary E. Complex mechanics of the heterogeneous extracellular matrix in cancer. EXTREME MECHANICS LETTERS 2018; 21:25-34. [PMID: 30135864 PMCID: PMC6097546 DOI: 10.1016/j.eml.2018.02.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/07/2018] [Accepted: 02/20/2018] [Indexed: 05/14/2023]
Abstract
The extracellular matrix (ECM) performs many critical functions, one of which is to provide structural and mechanical integrity, and many of the constituent proteins have clear mechanical roles. The composition and structural characteristics of the ECM are widely variable among different tissues, suiting diverse functional needs. In diseased tissues, particularly solid tumors, the ECM is complex and influences disease progression. Cancer and stromal cells can significantly influence the matrix composition and structure and thus the mechanical properties of the tumor microenvironment (TME). In this review, we describe the interactions that give rise to the structural heterogeneity of the ECM and present the techniques that are widely employed to measure ECM properties and remodeling dynamics. Furthermore, we review the tools for measuring the distinct nature of cell-ECM interactions within the TME.
Collapse
Affiliation(s)
- Andrea Malandrino
- Institute for Bioengineering of Catalonia, Barcelona, Spain
- European Molecular Biology Laboratory, Barcelona, Spain
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Roger D. Kamm
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emad Moeendarbary
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, University College London, London, UK
| |
Collapse
|
33
|
Cheng HL, Hsieh MJ, Yang JS, Lin CW, Lue KH, Lu KH, Yang SF. Nobiletin inhibits human osteosarcoma cells metastasis by blocking ERK and JNK-mediated MMPs expression. Oncotarget 2018; 7:35208-23. [PMID: 27144433 PMCID: PMC5085222 DOI: 10.18632/oncotarget.9106] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/11/2016] [Indexed: 12/12/2022] Open
Abstract
Nobiletin, a polymethoxyflavone, has a few pharmacological activities, including anti-inflammation and anti-cancer effects. However, its effect on human osteosarcoma progression remains uninvestigated. Therefore, we examined the effectiveness of nobiletin against cellular metastasis of human osteosarcoma and the underlying mechanisms. Nobiletin, up to 100 μM without cytotoxicity, significantly decreased motility, migration and invasion as well as enzymatic activities, protein levels and mRNA expressions of matrix metalloproteinase (MMP)-2 and MMP-9 in U2OS and HOS cells. In addition to inhibition of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), the inhibitory effect of nobiletin on the DNA-binding activity of the transcription factor nuclear factor-kappa B (NF-κB), cAMP response element-binding protein (CREB), and specificity protein 1 (SP-1) in U2OS and HOS cells. Co-treatment with ERK and JNK inhibitors and nobiletin further reduced U2OS cells migration and invasion. These results indicated that nobiletin inhibits human osteosarcoma U2OS and HOS cells motility, migration and invasion by down-regulating MMP-2 and MMP-9 expressions via ERK and JNK pathways and through the inactivation of downstream NF-κB, CREB, and SP-1. Nobiletin has the potential to serve as an anti-metastatic agent for treating osteosarcoma.
Collapse
Affiliation(s)
- Hsin-Lin Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Ming-Ju Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Jia-Sin Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Ko-Haung Lue
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ko-Hsiu Lu
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Orthopedics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
34
|
Dong X, Han Y, Sun Z, Xu J. Actin Gamma 1, a new skin cancer pathogenic gene, identified by the biological feature‐based classification. J Cell Biochem 2017; 119:1406-1419. [DOI: 10.1002/jcb.26301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/19/2017] [Indexed: 01/27/2023]
Affiliation(s)
- Xinqian Dong
- Pathology DepartmentLiaocheng People's HospitalLiaochengChina
| | - Yingsheng Han
- Dermatology DepartmentLiaocheng People's HospitalLiaochengChina
| | - Zhen Sun
- Gastroenterology DepartmentLiaocheng People's HospitalLiaochengChina
| | - Junlong Xu
- Pathology DepartmentLiaocheng People's HospitalLiaochengChina
| |
Collapse
|
35
|
Yuda A, McCulloch CA. A Screening System for Evaluating Cell Extension Formation, Collagen Compaction, and Degradation in Drug Discovery. SLAS DISCOVERY 2017; 23:132-143. [PMID: 28957641 DOI: 10.1177/2472555217733421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The generation of cell extensions is critical for matrix remodeling in tissue invasion by cancer cells, but current methods for identifying molecules that regulate cell extension formation and matrix remodeling are not well adapted for screening purposes. We applied a grid-supported, floating collagen gel system (~100 Pa stiffness) to examine cell extension formation, collagen compaction, and collagen degradation in a single assay. With the use of cultured diploid fibroblasts, a fibroblast cell line, and two cancer cell lines, we found that compared with attached collagen gels (~2800 Pa), the mean number and length of cell extensions were respectively greater in the floating gels. In assessing specific processes in cell extension formation, compared with controls, the number of cell extensions was reduced by latrunculin B, β1 integrin blockade, and a formin FH2 domain inhibitor. Screening of a kinase inhibitor library (480 compounds) with the floating gel assay showed that compared with vehicle-treated cells, there were large reductions of collagen compaction, pericellular collagen degradation, and number of cell extensions after treatment with SB431542, SIS3, Fasudil, GSK650394, and PKC-412. These data indicate that the grid-supported floating collagen gel model can be used to screen for inhibitors of cell extension formation and critical matrix remodeling events associated with cancer cell invasion.
Collapse
Affiliation(s)
- Asuka Yuda
- 1 Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
36
|
Smuczek B, Santos EDS, Siqueira AS, Pinheiro JJ, Freitas VM, Jaeger RG. The laminin-derived peptide C16 regulates GPNMB expression and function in breast cancer. Exp Cell Res 2017; 358:323-334. [DOI: 10.1016/j.yexcr.2017.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 11/27/2022]
|
37
|
Dilmac S, Erin N, Demir N, Tanriover G. Nephronectin is Decreased in Metastatic Breast Carcinoma and Related to Metastatic Organs. Pathol Oncol Res 2017; 24:679-688. [PMID: 28842827 DOI: 10.1007/s12253-017-0289-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/09/2017] [Indexed: 01/07/2023]
Abstract
Breast cancer causes death mostly due to distant metastasis. During metastasis, cancer cells create new conditions in which normal tissue structure can be disturbed. Nephronectin, which is the primary ligand for α8β1 integrin, plays an important role in kidney development. There are conflicting findings regarding its role in cancer progression and metastasis, especially in breast carcinoma. The aim of this study was to determine changes in nephronectin expression in primary tumor tissues and metastatic visceral organs, using metastatic and non-metastatic cell lines in a mouse model of breast cancer. In our study, 4T1-Liver Metastatic and 4T1-Heart Metastatic cells, originally derived from 4T1-murine breast carcinoma, and non-metastatic 67NR carcinoma cells were used. Cancer cells were injected orthotopically into the mammary gland of 8-10 week-old Balb-c mice. Primary tumors, lung, liver tissues were collected on 12th and 25th days after the tumor injection. Immunohistochemistry was used to determine expression of nephronectin in tissues. We also investigated the expression levels of the protein by using western blot technique. We found that lung and liver tissue of control animals (not-injected with tumor cells) expressed nephronectin which was lost in animals bearing metastatic tumor for 25 days. In accordance, nephronectin staining of lung and liver was preserved in animals injected with non-metastatic 67NR tumors. These results demonstrate that loss of nephronectin may play an important role in formation metastatic milieu for cancer cells. This is the first study demonstrating that tumor-induced loss of nephronectin expression in visceral organs in which metastatic growth takes place.
Collapse
Affiliation(s)
- Sayra Dilmac
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Nuray Erin
- Department of Medical Pharmacology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Necdet Demir
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Gamze Tanriover
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
38
|
Kramer B, Schultz JD, Hock C, Sauter A, Stuck BA, Hörmann K, Birk R, Aderhold C. Effect of selective small molecule inhibitors on MMP-9 and VEGFR-1 expression in p16-positive and -negative squamous cell carcinoma. Oncol Lett 2017; 13:3269-3276. [PMID: 28521433 DOI: 10.3892/ol.2017.5844] [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: 10/06/2016] [Accepted: 01/12/2017] [Indexed: 11/06/2022] Open
Abstract
The identification of molecular targets in the therapy of human papilloma virus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) is a primary aim of cancer research. Matrix metalloproteinase 9 (MMP-9) and vascular endothelial growth factor receptor (VEGFR) have important roles in the development of HNSCC. The tyrosine kinase inhibitors, nilotinib, dasatinib, erlotinib and gefitinib are well established in the targeted therapy of tumors other than HNSCC. The present study aimed to investigate the alteration of MMP-9 and VEGFR-1 expression patterns following treatment with these tyrosine kinase inhibitors in p16-positive and -negative squamous carcinoma cells. MMP-9 and VEGFR-1 expression was evaluated using an ELISA in HNSCC 11A, HNSCC 14C and p16-positive CERV196 tumor cell lines, following treatment with nilotinib, dasatinib, erlotinib and gefitinib. A statistically significant reduction in MMP-9 and VEGFR-1 expression was observed in the p16-negative HNSCC 11A cells following treatment with all inhibitors (P<0.05). VEGFR-1 expression was significantly increased in p16-positive SCC cells following treatment with nilotinib, dasatinib, erlotinib and gefitinib (P<0.05). The expression of MMP-9 and VEGFR-1 was significantly altered by treatment with nilotinib, dasatinib, erlotinib and gefitinib in vitro. The results of the present study are attributed to the efficacy of the tested drugs and present potential compensatory strategies of cancer cells to avoid the antiangiogenic properties of the tested tyrosine kinase inhibitors in vitro.
Collapse
Affiliation(s)
- Benedikt Kramer
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Johannes David Schultz
- Department of Otorhinolaryngology Head and Neck Surgery, Facial Plastic Surgery, Karlsruhe Clinical Center, Karlsruhe, Germany
| | - Clemens Hock
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alexander Sauter
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Boris A Stuck
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karl Hörmann
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Richard Birk
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christoph Aderhold
- Department of Otorhinolaryngology Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
39
|
Wang X, Cui H, Lou Z, Huang S, Ren Y, Wang P, Weng G. Cyclic AMP responsive element-binding protein induces metastatic renal cell carcinoma by mediating the expression of matrix metallopeptidase-2/9 and proteins associated with epithelial-mesenchymal transition. Mol Med Rep 2017; 15:4191-4198. [PMID: 28487942 DOI: 10.3892/mmr.2017.6519] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/10/2017] [Indexed: 11/06/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most frequently occurring malignancy of the kidney worldwide. Anti-angiogenic targeted therapies inhibit the progression of RCC, however, limited effects on the invasion or metastasis of tumor cells have been observed. Cyclic AMP responsive element‑binding protein (CREB) is a serine/threonine kinase that has been implicated in the regulation of cell proliferation, apoptosis, cycle progression and metastasis, amongst others. Our previous research demonstrated that phosphorylated CREB (pCREB) was upregulated in human renal cancer cell lines and tissues, and decreased pCREB at the Ser133 site inhibited the growth and metastatic activity of OS‑RC‑2 cells. However, the role of CREB in RCC metastasis requires further investigation. Thus, the present study further investigated the role of CREB in RCC metastasis. The present study demonstrated that knockdown of CREB using small interfering RNA (siRNA) that targeted CREB (siCREB) significantly inhibited the migration and invasion of 786‑O and OS‑RC‑2 cells, however, the opposite effect was observed in ACHN cells. In addition, knockdown of CREB suppressed the expression of matrix metallopeptidase (MMP)‑2/9 and proteins associated with epithelial‑mesenchymal transition (EMT) in 786‑O and OS‑RC‑2 cells, and promoted expression in ACHN cells. Furthermore, the chromatin immunoprecipitation assay indicated that pCREB (Ser133) had a direct interaction with the fibronectin promoter, however, pCREB (Ser133) did not target the vimentin promoter in RCC. Therefore, the results of the present study indicate that CREB regulated metastatic RCC by mediating the expression of MMP‑2/9 and EMT‑associated proteins, however, CREB‑mediated MMP‑2/9 and EMT‑associated protein expression may be induced by different pathways in different RCC cells.
Collapse
Affiliation(s)
- Xue Wang
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Hui Cui
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Zhongguan Lou
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Shuaishuai Huang
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Yu Ren
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Ping Wang
- Laboratory of Kidney Carcinoma, Urology and Nephrology Institute of Ningbo University, Ningbo, Zhejiang 315000, P.R. China
| | - Guobin Weng
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
40
|
Xia S, Ji R, Zhan W. Long noncoding RNA papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) inhibits proliferation and invasion of glioma cells by suppressing the Wnt/β-catenin signaling pathway. BMC Neurol 2017; 17:30. [PMID: 28187755 PMCID: PMC5303216 DOI: 10.1186/s12883-017-0813-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/03/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The dysregulation of long noncoding RNAs (lncRNAs) has been identified in a variety of cancers. An increasing number of studies have found the critical role of lncRNAs in the regulation of cellular processes, such as proliferation, invasion and differentiation. Long noncoding RNA papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) is a novel lncRNA that was primarily detected in papillary thyroid carcinoma. However, the biological function and molecular mechanism of lncRNA PTCSC3 in glioma are still unknown. METHODS The expression level of lncRNA PTCSC3 in human microglia and glioma cell lines was examined using quantitative real-time polymerase chain reaction (qRT-PCR). The influence of lncRNA PTCSC3 on cell proliferation were studied using the cell counting kit-8, and cell cycle and apoptosis were analyzed by flow cytometry assays. The migration and invasion abilities were investigated by transwell and wound healing assays. The target genes of lncRNA PTCSC3 were explored by qRT-PCR, immunofluorescence and western blot. RESULTS LncRNA PTCSC3 was significantly downregulated in glioma cell lines. The overexpression of lncRNA PTCSC3 suppressed proliferation and induced apoptosis in U87 and U251 cells. Additionally, the overexpression of lncRNA PTCSC3 inhibited the migration and invasion of U87 and U251 cells. Moreover, lncRNA PTCSC3 inhibited the epithelial-mesenchymal transition of U87 cells. The study also demonstrated that LRP6, as a receptor of the Wnt/β-catenin pathway, was a target of lncRNA PTCSC3. By evaluating the expression levels of Axin1, active β-catenin, c-myc, and cyclin D1, the study indicated that lncRNA PTCSC3 inhibited the activation of the Wnt/β-cateninpathway through targeting LRP6. CONCLUSIONS LncRNA PTCSC3 inhibits the proliferation and migration of glioma cells and suppresses Wnt/β-catenin signaling pathway by targeting LRP6. LncRNA PTCSC3 is a potential therapeutic target for treatment of glioma.
Collapse
Affiliation(s)
- Shujun Xia
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, Zip code: 200025, People's Republic of China
| | - Ri Ji
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, Zip code: 200025, People's Republic of China
| | - Weiwei Zhan
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, Zip code: 200025, People's Republic of China.
| |
Collapse
|
41
|
Pan S, Brentnall TA, Chen R. Glycoproteins and glycoproteomics in pancreatic cancer. World J Gastroenterol 2016; 22:9288-9299. [PMID: 27895417 PMCID: PMC5107693 DOI: 10.3748/wjg.v22.i42.9288] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/23/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
Aberrations in protein glycosylation and polysaccharides play a pivotal role in pancreatic tumorigenesis, influencing cancer progression, metastasis, immuno-response and chemoresistance. Abnormal expression in sugar moieties can impact the function of various glycoproteins, including mucins, surface receptors, adhesive proteins, proteoglycans, as well as their effectors and binding ligands, resulting in an increase in pancreatic cancer invasiveness and a cancer-favored microenvironment. Recent advance in glycoproteomics, glycomics and other chemical biology techniques have been employed to better understand the complex mechanism of glycosylation events and how they orchestrate molecular activities in genomics, proteomics and metabolomics implicated in pancreatic adenocarcinoma. A variety of strategies have been demonstrated targeting protein glycosylation and polysaccharides for diagnostic and therapeutic development.
Collapse
|
42
|
Yu X, Wang Q, Zhou X, Fu C, Cheng M, Guo R, Liu H, Zhang B, Dai M. Celastrol negatively regulates cell invasion and migration ability of human osteosarcoma via downregulation of the PI3K/Akt/NF-κB signaling pathway in vitro. Oncol Lett 2016; 12:3423-3428. [PMID: 27900015 DOI: 10.3892/ol.2016.5049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/27/2016] [Indexed: 12/26/2022] Open
Abstract
Osteosarcoma (OS) is a primary malignant tumor of the bone, with a tendency to metastasize early. Despite the advances in treatment options, more than 30% of patients develop distant metastases, and the prognosis of these patients with metastases is extremely poor. Celastrol has been demonstrated to manifest multiple pharmacological activities, including induction of apoptosis in numerous types of cancer cell lines. Our previous studies have also suggested that Celastrol is capable of inducing apoptosis of human osteosarcoma cells via the mitochondrial-dependent pathway. The purpose of this study was to investigate the effects of Celastrol on the migration and invasion of human osteosarcoma U-2OS cells in vitro. Cell migration and invasion were investigated using wound healing and Boyden chamber Transwell assays. We observed that Celastrol suppressed cell invasion and migration in human osteosarcoma U-2OS cells. Furthermore, protein expression levels of phosphorylated phosphatidylinositol 3-kinase (PI3K), Akt, inhibitor of κB kinase α/β, inhibitor of κB α, nuclear factor-κB (NF-κB subunit p65) and matrix metalloproteinase (MMP)-2 and -9 were measured by western blot analysis. We observed that the PI3K/Akt/NF-κB signaling pathway was inhibited following Celastrol treatment. In addition, the expression levels of MMP-2 and -9 proteins were also reduced significantly following Celastrol treatment. Therefore, we confirmed that Celastrol suppressed osteosarcoma U-2OS cell metastasis via downregulation of the PI3K/Akt/NF-κB signaling pathway in vitro.
Collapse
Affiliation(s)
- Xiaolong Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Qiang Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Xin Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Changlin Fu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Ming Cheng
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Runsheng Guo
- Multidisciplinary Therapy Center of Musculoskeletal Tumor, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hucheng Liu
- Multidisciplinary Therapy Center of Musculoskeletal Tumor, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
43
|
Gucciardo E, Mobashir M, Lehti K. Proactive for invasion: Reuse of matrix metalloproteinase for structural memory. J Cell Biol 2016; 213:11-3. [PMID: 27069020 PMCID: PMC4828696 DOI: 10.1083/jcb.201603066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 01/28/2023] Open
Abstract
Migratory cells translocate membrane type-1 matrix metalloproteinase (MT1-MMP) to podosomes or invadosomes to break extracellular matrix barriers. In this issue, El Azzouzi et al. (2016. J. Cell. Biol.http://dx.doi.org/10.1083/jcb.201510043) describe an unexpected function for the MT1-MMP cytoplasmic domain in imprinting spatial memory for podosome reformation via assembly in membrane islets.
Collapse
Affiliation(s)
- Erika Gucciardo
- Research Programs Unit, Genome-Scale Biology and Haartman Institute, University of Helsinki, Helsinki University Hospital and Finnish Cancer Institute, Helsinki FI-00014, Finland
| | - Mohammad Mobashir
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Stockholm SE-17165, Sweden
| | - Kaisa Lehti
- Research Programs Unit, Genome-Scale Biology and Haartman Institute, University of Helsinki, Helsinki University Hospital and Finnish Cancer Institute, Helsinki FI-00014, Finland Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Stockholm SE-17165, Sweden
| |
Collapse
|
44
|
Castro-Castro A, Marchesin V, Monteiro P, Lodillinsky C, Rossé C, Chavrier P. Cellular and Molecular Mechanisms of MT1-MMP-Dependent Cancer Cell Invasion. Annu Rev Cell Dev Biol 2016; 32:555-576. [PMID: 27501444 DOI: 10.1146/annurev-cellbio-111315-125227] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metastasis is responsible for most cancer-associated deaths. Accumulating evidence based on 3D migration models has revealed a diversity of invasive migratory schemes reflecting the plasticity of tumor cells to switch between proteolytic and nonproteolytic modes of invasion. Yet, initial stages of localized regional tumor dissemination require proteolytic remodeling of the extracellular matrix to overcome tissue barriers. Recent data indicate that surface-exposed membrane type 1-matrix metalloproteinase (MT1-MMP), belonging to a group of membrane-anchored MMPs, plays a central role in pericellular matrix degradation during basement membrane and interstitial tissue transmigration programs. In addition, a large body of work indicates that MT1-MMP is targeted to specialized actin-rich cell protrusions termed invadopodia, which are responsible for matrix degradation. This review describes the multistep assembly of actin-based invadopodia in molecular details. Mechanisms underlying MT1-MMP traffic to invadopodia through endocytosis/recycling cycles, which are key to the invasive program of carcinoma cells, are discussed.
Collapse
Affiliation(s)
| | | | - Pedro Monteiro
- Barts Cancer Institute, University of London John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Catalina Lodillinsky
- Instituto de Oncologia Ángel H. Roffo, Research Area, Buenos Aires, C1417DTB, Argentina
| | - Carine Rossé
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| | - Philippe Chavrier
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| |
Collapse
|
45
|
Carey SP, Goldblatt ZE, Martin KE, Romero B, Williams RM, Reinhart-King CA. Local extracellular matrix alignment directs cellular protrusion dynamics and migration through Rac1 and FAK. Integr Biol (Camb) 2016; 8:821-35. [PMID: 27384462 PMCID: PMC4980151 DOI: 10.1039/c6ib00030d] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell migration within 3D interstitial microenvironments is sensitive to extracellular matrix (ECM) properties, but the mechanisms that regulate migration guidance by 3D matrix features remain unclear. To examine the mechanisms underlying the cell migration response to aligned ECM, which is prevalent at the tumor-stroma interface, we utilized time-lapse microscopy to compare the behavior of MDA-MB-231 breast adenocarcinoma cells within randomly organized and well-aligned 3D collagen ECM. We developed a novel experimental system in which cellular morphodynamics during initial 3D cell spreading served as a reductionist model for the complex process of matrix-directed 3D cell migration. Using this approach, we found that ECM alignment induced spatial anisotropy of cells' matrix probing by promoting protrusion frequency, persistence, and lengthening along the alignment axis and suppressing protrusion dynamics orthogonal to alignment. Preference for on-axis behaviors was dependent upon FAK and Rac1 signaling and translated across length and time scales such that cells within aligned ECM exhibited accelerated elongation, front-rear polarization, and migration relative to cells in random ECM. Together, these findings indicate that adhesive and protrusive signaling allow cells to respond to coordinated physical cues in the ECM, promoting migration efficiency and cell migration guidance by 3D matrix structure.
Collapse
Affiliation(s)
- Shawn P Carey
- Department of Biomedical Engineering, Cornell University, 302 Weill Hall, 526 Campus Rd, Ithaca, New York 14853, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Mao W, Sun Y, Zhang H, Cao L, Wang J, He P. A combined modality of carboplatin and photodynamic therapy suppresses epithelial-mesenchymal transition and matrix metalloproteinase-2 (MMP-2)/MMP-9 expression in HEp-2 human laryngeal cancer cells via ROS-mediated inhibition of MEK/ERK signalling pathway. Lasers Med Sci 2016; 31:1697-1705. [PMID: 27481258 DOI: 10.1007/s10103-016-2040-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/25/2016] [Indexed: 01/23/2023]
Abstract
Photodynamic therapy (PDT) has been developed as a promising treatment modality for laryngeal cancer. 9-Hydroxypheophorbide α (9-HPbD), a novel chlorophyll-derived photosensitizer, has a longer absorption wavelength, which increases the penetration of light to malignant tissues. Carboplatin (CBDCA), a second-generation platinum derivative, also has gained more popularity for the treatment of laryngeal cancer. Our previous studies have elucidated that 9-HPbD-PDT could inhibit the migration and invasion of HEp-2 cells. The objective of this study is to investigate the change of migration and invasion of HEp-2 cells induced by a combined modality of CBDCA and 9-HPbD-PDT in vitro. A wound healing assay, cell migration assay and Matrigel invasion assay were used to evaluate the cellular migration and invasion. Reactive oxygen species (ROS) and Western blots for epithelial-mesenchymal transition (EMT) markers (E-cadherin, N-cadherin and vimentin), MMPs (MMP-2 and MMP-9) and MEK/ERK signalling pathway were performed to investigate the possible mechanisms that may be involved. We observed that CBDCA and 9-HPbD-PDT administration synergistically inhibited the migration and invasion of HEp-2 cells. Moreover, the combined modality cooperatively repressed the EMT process and down-regulated expressions of MMP-2 and MMP-9 via ROS-mediated inhibition of phosphorylation in the MEK/ERK signalling pathway. Our results suggested that the combination of CBDCA and 9-HPbD-PDT might be a promising therapeutic strategy for laryngeal cancer metastasis.
Collapse
Affiliation(s)
- Wenjing Mao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, No. 83, Fenyang Rd., Shanghai, 200031, China
| | - Yan Sun
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Huankang Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, No. 83, Fenyang Rd., Shanghai, 200031, China
| | - Luhong Cao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, No. 83, Fenyang Rd., Shanghai, 200031, China
| | - Jiajia Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, No. 83, Fenyang Rd., Shanghai, 200031, China
| | - Peijie He
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, No. 83, Fenyang Rd., Shanghai, 200031, China.
| |
Collapse
|
47
|
Expression of MMP and TIMP mRNA in Peripheral Blood Leukocytes of Patients with Invasive Ductal Carcinoma of the Breast. Int J Biol Markers 2016; 31:e309-16. [DOI: 10.5301/jbm.5000203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2016] [Indexed: 11/20/2022]
Abstract
Purpose An imbalance between matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) appears critical for tumor progression and metastasis. This study aimed to determine whether gene expression of MMP1, MMP2, MMP9, TIMP1 and TIMP3 and the MMP/TIMP expression ratio in peripheral blood leukocytes (PBLs) and the MMP1 and TIMP1 contents or MMP1/TIMP1 ratio in plasma were associated with clinicopathological characteristics in invasive ductal carcinoma (IDC) of the breast. Materials and methods Blood samples were collected from women newly diagnosed with IDC who had not received prior treatment (n = 102). Gene expression in PBLs was analyzed by quantitative real-time polymerase chain reaction. Concentrations of MMP1 and TIMP1 in plasma were measured using ELISA. Results In univariate analysis the expression levels of MMP2 and TIMP1 mRNA were significantly higher in premenopausal compared to postmenopausal patients (p<0.001 and p = 0.014, respectively). MMP2 mRNA expression negatively correlated with age (p<0.001, r = -0.43). We found that the MMP2/TIMP3 expression ratio was significantly higher in women after menopause (p = 0.007). The MMP2/TIMP1 expression ratio was higher in human epidermal growth factor receptor 2 (HER2)-positive patients (p = 0.022). Low-grade tumors had significantly lower MMP1/TIMP1 and MMP2/TIMP1 expression ratios (p = 0.047 and p = 0.048, respectively). TIMP1 plasma concentration was significantly higher in small tumors compared with T2-T3 tumors (p = 0.013). Conclusions These findings reveal an important association between tumor characteristics and expression ratios of MMP1/TIMP1 and MMP2/TIMP1 in PBLs and TIMP1 concentration in plasma. Menopausal status may influence the mRNA expression levels of MMP2 and TIMP1 as well as the MMP2/TIMP3 expression ratio in IDC of the breast.
Collapse
|
48
|
Suppression of A549 cell proliferation and metastasis by calycosin via inhibition of the PKC‑α/ERK1/2 pathway: An in vitro investigation. Mol Med Rep 2015; 12:7992-8002. [PMID: 26498639 PMCID: PMC4758323 DOI: 10.3892/mmr.2015.4449] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 08/24/2015] [Indexed: 12/28/2022] Open
Abstract
The migration and invasion of lung cancer cells into the extracellular matrix contributes to the high mortality rates of lung cancer. The protein kinase C (PKC) and downstream signaling pathways are important in the invasion and migration of lung cancer cells. Calycosin (Cal), an effector chemical from Astragalus has been reported to affect the recurrence and metastasis of cancer cells via the regulation of the protein expression of matrix metalloproteinases (MMPs). The inhibition of Cal on the migration and invasion of A549 cells was investigated in the present study. Cell viability and apoptosis assays were performed using MTT and flow cytometric analyses. A wound healing assay and Transwell invasion assay were performed to evaluate the effect of Cal on A549 cell migration and invasion. Invasion-associated proteins, including MMP-2, MMP-9, E-cadherin (E-cad), integrin β1, PKC-α and extracellular signal-regulated kinase 1/2 (ERK1/2) were detected using western blotting. In addition, PKC-α inhibitor, AEB071, and ERK1/2 inhibitor, PD98059, were used to determine the association between the suppression of PKC-α/ERK1/2 and invasion, MMP-2, MMP-9, E-cad and integrin β1. Cal was observed to suppress cell proliferation and induce apoptosis. There were significant differences between the phorbol-12-myristate-13-acetate (TPA)-induced A549 cells treated with Cal and the untreated cells in the rates of migration and invasion. The levels of MMP-2, MMP-9, E-cad and integrin β1 in the TPA-induced A549 cells changed markedly, compared with the untreated cells. In addition, the suppression of Cal was affected by the PKC inhibitor, AEB071, an ERK1/2 inhibitor, PD98059. The results of the present study indicated that Cal inhibited the proliferation, adhesion, migration and invasion of the TPA-induced A549 cells. The Cal-induced repression of PKC-α/ERK1/2, increased the expression of E-Cad and inhibited the expression levels of MMP-2, MMP-9 and integrin β1, which possibly demonstrates the mechanism underlying the biological anticancer effects of Cal.
Collapse
|
49
|
Generating and characterizing the mechanical properties of cell-derived matrices using atomic force microscopy. Methods 2015; 94:85-100. [PMID: 26439175 DOI: 10.1016/j.ymeth.2015.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/26/2015] [Accepted: 09/14/2015] [Indexed: 12/22/2022] Open
Abstract
Mechanical interaction between cells and their surrounding extracellular matrix (ECM) controls key processes such as proliferation, differentiation and motility. For many years, two-dimensional (2D) models were used to better understand the interactions between cells and their surrounding ECM. More recently, variation of the mechanical properties of tissues has been reported to play a major role in physiological and pathological scenarios such as cancer progression. The 3D architecture of the ECM finely tunes cellular behavior to perform physiologically relevant tasks. Technical limitations prevented scientists from obtaining accurate assessment of the mechanical properties of physiologically realistic matrices. There is therefore a need for combining the production of high-quality cell-derived 3D matrices (CDMs) and the characterization of their topographical and mechanical properties. Here, we describe methods that allow to accurately measure the young modulus of matrices produced by various cellular types. In the first part, we will describe and review several protocols for generating CDMs matrices from endothelial, epithelial, fibroblastic, muscle and mesenchymal stem cells. We will discuss tools allowing the characterization of the topographical details as well as of the protein content of such CDMs. In a second part, we will report the methodologies that can be used, based on atomic force microscopy, to accurately evaluate the stiffness properties of the CDMs through the quantification of their young modulus. Altogether, such methodologies allow characterizing the stiffness and topography of matrices deposited by the cells, which is key for the understanding of cellular behavior in physiological conditions.
Collapse
|
50
|
Herrera-Perez M, Voytik-Harbin SL, Rickus JL. Extracellular Matrix Properties Regulate the Migratory Response of Glioblastoma Stem Cells in Three-Dimensional Culture. Tissue Eng Part A 2015; 21:2572-82. [PMID: 26161688 PMCID: PMC4605380 DOI: 10.1089/ten.tea.2014.0504] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 07/09/2015] [Indexed: 12/12/2022] Open
Abstract
Diffuse infiltration across brain tissue is a hallmark of glioblastoma and the main cause of unsuccessful total resection that leads to tumor reappearance. A subpopulation termed glioblastoma stem cells (GSCs) has been directly related to aggressive invasion; nonetheless, their migratory characteristics and regulation by the microenvironment are still unknown. In this study, we developed a composite matrix of hyaluronan (HA) structurally supported by a collagen-oligomer fibril network to simulate the brain tumor extracellular matrix (ECM) composition. Matrigel-coated microfibers were embedded within the matrix to create a tunable dual niche microenvironment that resembles the vascular network of the brain. This model was compared with the most commonly used in vitro three-dimensional (3D) culture formats, Matrigel and collagen type-I monomer matrices, to study how the mechanical and compositional properties of the ECM alter the migration characteristics of GSC neurospheres. The migration mode, distance, velocity, and morphology of the GSCs were monitored over a 72-h period. The cells altered their migration mode depending on the matrix composition, showing migration by expansive growth in Matrigel matrices, multicellular extension along rigid interfaces (as Matrigel glass and coated microfibers), and mesenchymal single-cell migration in collagen matrices. Velocity and distance of migration within each composition varied according to matrix mechanical properties. In the dual niche system, the presence of HA reduced velocity and number of migratory cells; however, cells that came in contact with the pseudovessels exhibited collective migration by an extensive strand and reached higher velocities than cells migrating individually across the 3D matrix. Our results show that GSCs adopt varied migration mechanisms to invade multiple ECM microenvironments, and the migration characteristics exhibited are highly influenced by the matrix physical properties. Moreover, GSC neurospheres exhibit concomitant single and collective migration as a function of the microenvironment topography to reach the most productive migration strategy.
Collapse
Affiliation(s)
- Marisol Herrera-Perez
- Department of Agricultural and Biological Engineering, College of Engineering, Purdue University, West Lafayette, Indiana
- Physiological Sensing Facility at the Bindley Bioscience Center and the Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana
| | - Sherry L. Voytik-Harbin
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, Indiana
- Department of Basic Medical Sciences, College of Veterinary Medicine. Purdue University, West Lafayette, Indiana
| | - Jenna L. Rickus
- Department of Agricultural and Biological Engineering, College of Engineering, Purdue University, West Lafayette, Indiana
- Physiological Sensing Facility at the Bindley Bioscience Center and the Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, Indiana
| |
Collapse
|