151
|
Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses. Adv Drug Deliv Rev 2014; 79-80:193-213. [PMID: 24969478 DOI: 10.1016/j.addr.2014.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Tissue engineering technologies, which have originally been designed to reconstitute damaged tissue structure and function, can mimic not only tissue regeneration processes but also cancer development and progression. Bioengineered approaches allow cell biologists to develop sophisticated experimentally and physiologically relevant cancer models to recapitulate the complexity of the disease seen in patients. Tissue engineering tools enable three-dimensionality based on the design of biomaterials and scaffolds that re-create the geometry, chemistry, function and signalling milieu of the native tumour microenvironment. Three-dimensional (3D) microenvironments, including cell-derived matrices, biomaterial-based cell culture models and integrated co-cultures with engineered stromal components, are powerful tools to study dynamic processes like proteolytic functions associated with cancer progression, metastasis and resistance to therapeutics. In this review, we discuss how biomimetic strategies can reproduce a humanised niche for human cancer cells, such as peritoneal or bone-like microenvironments, addressing specific aspects of ovarian and prostate cancer progression and therapy response.
Collapse
|
152
|
In vitro modeling of the prostate cancer microenvironment. Adv Drug Deliv Rev 2014; 79-80:214-21. [PMID: 24816064 DOI: 10.1016/j.addr.2014.04.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 01/01/2023]
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and advanced disease is incurable. Model systems are a fundamental tool for research and many in vitro models of prostate cancer use cancer cell lines in monoculture. Although these have yielded significant insight they are inherently limited by virtue of their two-dimensional (2D) growth and inability to include the influence of tumour microenvironment. These major limitations can be overcome with the development of newer systems that more faithfully recreate and mimic the complex in vivo multi-cellular, three-dimensional (3D) microenvironment. This article presents the current state of in vitro models for prostate cancer, with particular emphasis on 3D systems and the challenges that remain before their potential to advance our understanding of prostate disease and aid in the development and testing of new therapeutic agents can be realised.
Collapse
|
153
|
3D tissue-engineered model of Ewing's sarcoma. Adv Drug Deliv Rev 2014; 79-80:155-71. [PMID: 25109853 DOI: 10.1016/j.addr.2014.07.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/28/2014] [Accepted: 07/24/2014] [Indexed: 12/30/2022]
Abstract
Despite longstanding reliance upon monolayer culture for studying cancer cells, and numerous advantages from both a practical and experimental standpoint, a growing body of evidence suggests that more complex three-dimensional (3D) models are necessary to properly mimic many of the critical hallmarks associated with the oncogenesis, maintenance and spread of Ewing's sarcoma (ES), the second most common pediatric bone tumor. And as clinicians increasingly turn to biologically-targeted therapies that exert their effects not only on the tumor cells themselves, but also on the surrounding extracellular matrix, it is especially important that preclinical models evolve in parallel to reliably measure antineoplastic effects and possible mechanisms of de novo and acquired drug resistance. Herein, we highlight a number of innovative methods used to fabricate biomimetic ES tumors, encompassing both the surrounding cellular milieu and the extracellular matrix (ECM), and suggest potential applications to advance our understanding of ES biology, preclinical drug testing, and personalized medicine.
Collapse
|
154
|
Chang F, Lemmon CA, Nilaratanakul V, Rotter V, Romer L. Endothelial matrix assembly during capillary morphogenesis: insights from chimeric TagRFP-fibronectin matrix. J Histochem Cytochem 2014; 62:774-90. [PMID: 25063001 PMCID: PMC4209295 DOI: 10.1369/0022155414547419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/13/2014] [Indexed: 11/22/2022] Open
Abstract
Biologically relevant, three-dimensional extracellular matrix is an essential component of in vitro vasculogenesis models. WI-38 fibroblasts assemble a 3D matrix that induces endothelial tubulogenesis, but this model is challenged by fibroblast senescence and the inability to distinguish endothelial cell-derived matrix from matrix made by WI-38 fibroblasts. Matrices produced by hTERT-immortalized WI-38 recapitulated those produced by wild type fibroblasts. ECM fibrils were heavily populated by tenascin-C, fibronectin, and type VI collagen. Nearly half of the total type I collagen, but only a small fraction of the type IV collagen, were incorporated into ECM. Stable hTERT-WI-38 transfectants expressing TagRFP-fibronectin incorporated TagRFP into ~90% of the fibronectin in 3D matrices. TagRFP-fibronectin colocalized with tenascin-C and with type I collagen in a pattern that was similar to that seen in matrices from wild type WI-38. Human Umbilical Vein Endothelial Cells (HUVEC) formed 3D adhesions and tubes on WI38-hTERT-TagRFP-FN-derived matrices, and the TagRFP-fibronectin component of this new 3D human fibroblast matrix model facilitated the demonstration of concentrated membrane type 1 metalloprotease and new HUVEC FN and collagen type IV fibrils during EC tubulogenesis. These findings indicate that WI-38-hTERT- and WI-38-hTERT-TagRFP-FN-derived matrices provide platforms for the definition of new matrix assembly and remodeling events during vasculogenesis.
Collapse
Affiliation(s)
- Fumin Chang
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Christopher A Lemmon
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Voraphoj Nilaratanakul
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Varda Rotter
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Lewis Romer
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| |
Collapse
|
155
|
Ivers LP, Cummings B, Owolabi F, Welzel K, Klinger R, Saitoh S, O'Connor D, Fujita Y, Scholz D, Itasaki N. Dynamic and influential interaction of cancer cells with normal epithelial cells in 3D culture. Cancer Cell Int 2014; 14:108. [PMID: 25379014 PMCID: PMC4221723 DOI: 10.1186/s12935-014-0108-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/15/2014] [Indexed: 02/04/2023] Open
Abstract
Background The cancer microenvironment has a strong impact on the growth and dynamics of cancer cells. Conventional 2D culture systems, however, do not reflect in vivo conditions, impeding detailed studies of cancer cell dynamics. This work aims to establish a method to reveal the interaction of cancer and normal epithelial cells using 3D time-lapse. Methods GFP-labelled breast cancer cells, MDA-MB-231, were co-cultured with mCherry-labelled non-cancerous epithelial cells, MDCK, in a gel matrix. In the 3D culture, the epithelial cells establish a spherical morphology (epithelial sphere) thus providing cancer cells with accessibility to the basal surface of epithelia, similar to the in vivo condition. Cell movement was monitored using time-lapse analyses. Ultrastructural, immunocytochemical and protein expression analyses were also performed following the time-lapse study. Results In contrast to the 2D culture system, whereby most MDA-MB-231 cells exhibit spindle-shaped morphology as single cells, in the 3D culture the MDA-MB-231 cells were found to be single cells or else formed aggregates, both of which were motile. The single MDA-MB-231 cells exhibited both round and spindle shapes, with dynamic changes from one shape to the other, visible within a matter of hours. When co-cultured with epithelial cells, the MDA-MB-231 cells displayed a strong attraction to the epithelial spheres, and proceeded to surround and engulf the epithelial cell mass. The surrounded epithelial cells were eventually destroyed, becoming debris, and were taken into the MDA-MB-231 cells. However, when there was a relatively large population of normal epithelial cells, the MDA-MB-231 cells did not engulf the epithelial spheres effectively, despite repeated contacts. MDA-MB-231 cells co-cultured with a large number of normal epithelial cells showed reduced expression of monocarboxylate transporter-1, suggesting a change in the cell metabolism. A decreased level of gelatin-digesting ability as well as reduced production of matrix metaroproteinase-2 was also observed. Conclusions This culture method is a powerful technique to investigate cancer cell dynamics and cellular changes in response to the microenvironment. The method can be useful for various aspects such as; different combinations of cancer and non-cancer cell types, addressing the organ-specific affinity of cancer cells to host cells, and monitoring the cellular response to anti-cancer drugs. Electronic supplementary material The online version of this article (doi:10.1186/s12935-014-0108-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura P Ivers
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| | - Brendan Cummings
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| | - Funke Owolabi
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| | | | - Rut Klinger
- Conway Institute, University College Dublin, Dublin, 4 Ireland ; School of Biomolecular and Biomedical Science, University College Dublin, Dublin, 4 Ireland
| | - Sayaka Saitoh
- Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815 Japan
| | - Darran O'Connor
- Conway Institute, University College Dublin, Dublin, 4 Ireland ; School of Biomolecular and Biomedical Science, University College Dublin, Dublin, 4 Ireland
| | - Yasuyuki Fujita
- Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815 Japan
| | - Dimitri Scholz
- Conway Institute, University College Dublin, Dublin, 4 Ireland
| | - Nobue Itasaki
- School of Medicine and Medical Science, University College Dublin, Dublin, 4 Ireland
| |
Collapse
|
156
|
Paye A, Truong A, Yip C, Cimino J, Blacher S, Munaut C, Cataldo D, Foidart JM, Maquoi E, Collignon J, Delvenne P, Jerusalem G, Noel A, Sounni NE. EGFR Activation and Signaling in Cancer Cells Are Enhanced by the Membrane-Bound Metalloprotease MT4-MMP. Cancer Res 2014; 74:6758-70. [DOI: 10.1158/0008-5472.can-13-2994] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
157
|
Pietraszek K, Chatron-Colliet A, Brézillon S, Perreau C, Jakubiak-Augustyn A, Krotkiewski H, Maquart FX, Wegrowski Y. Lumican: A new inhibitor of matrix metalloproteinase-14 activity. FEBS Lett 2014; 588:4319-24. [DOI: 10.1016/j.febslet.2014.09.040] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/24/2014] [Accepted: 09/24/2014] [Indexed: 11/27/2022]
|
158
|
Frittoli E, Palamidessi A, Marighetti P, Confalonieri S, Bianchi F, Malinverno C, Mazzarol G, Viale G, Martin-Padura I, Garré M, Parazzoli D, Mattei V, Cortellino S, Bertalot G, Di Fiore PP, Scita G. A RAB5/RAB4 recycling circuitry induces a proteolytic invasive program and promotes tumor dissemination. ACTA ACUST UNITED AC 2014; 206:307-28. [PMID: 25049275 PMCID: PMC4107781 DOI: 10.1083/jcb.201403127] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The mechanisms by which tumor cells metastasize and the role of endocytic proteins in this process are not well understood. We report that overexpression of the GTPase RAB5A, a master regulator of endocytosis, is predictive of aggressive behavior and metastatic ability in human breast cancers. RAB5A is necessary and sufficient to promote local invasion and distant dissemination of various mammary and nonmammary tumor cell lines, and this prometastatic behavior is associated with increased intratumoral cell motility. Specifically, RAB5A is necessary for the formation of invadosomes, membrane protrusions specialized in extracellular matrix (ECM) degradation. RAB5A promotes RAB4- and RABENOSYN-5-dependent endo/exocytic cycles (EECs) of critical cargos (membrane-type 1 matrix metalloprotease [MT1-MMP] and β3 integrin) required for invadosome formation in response to motogenic stimuli. This trafficking circuitry is necessary for spatially localized hepatocyte growth factor (HGF)/MET signaling that drives invasive, proteolysis-dependent chemotaxis in vitro and for conversion of ductal carcinoma in situ to invasive ductal carcinoma in vivo. Thus, RAB5A/RAB4 EECs promote tumor dissemination by controlling a proteolytic, mesenchymal invasive program.
Collapse
Affiliation(s)
- Emanuela Frittoli
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy
| | - Andrea Palamidessi
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy
| | - Paola Marighetti
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy
| | - Stefano Confalonieri
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | - Fabrizio Bianchi
- Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | - Chiara Malinverno
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy
| | - Giovanni Mazzarol
- Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | - Giuseppe Viale
- Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | - Ines Martin-Padura
- Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | | | - Dario Parazzoli
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy
| | - Valentina Mattei
- Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | | | - Giovanni Bertalot
- Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy
| | - Pier Paolo Di Fiore
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy Dipartimento di Oncologia Sperimentale, Istituto Europeo di Oncologia, 20141 Milan, Italy Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20122 Milan, Italy
| | - Giorgio Scita
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), 20139 Milan, Italy Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
159
|
A 3D matrix platform for the rapid generation of therapeutic anti-human carcinoma monoclonal antibodies. Proc Natl Acad Sci U S A 2014; 111:14882-7. [PMID: 25267635 DOI: 10.1073/pnas.1410996111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Efforts to develop unbiased screens for identifying novel function-blocking monoclonal antibodies (mAbs) in human carcinomatous states have been hampered by the limited ability to design in vitro models that recapitulate tumor cell behavior in vivo. Given that only invasive carcinoma cells gain permanent access to type I collagen-rich interstitial tissues, an experimental platform was established in which human breast cancer cells were embedded in 3D aldimine cross-linked collagen matrices and used as an immunogen to generate mAb libraries. In turn, cancer-cell-reactive antibodies were screened for their ability to block carcinoma cell proliferation within collagen hydrogels that mimic the in vivo environment. As a proof of principle, a single function-blocking mAb out of 15 identified was selected for further analysis and found to be capable of halting carcinoma cell proliferation, inducing apoptosis, and exerting global changes in gene expression in vitro. The ability of this mAb to block carcinoma cell proliferation and metastatic activity was confirmed in vivo, and the target antigen was identified by mass spectroscopy as the α2 subunit of the α2β1 integrin, one of the major type I collagen-binding receptors in mammalian cells. Validating the ability of the in vitro model to predict patterns of antigen expression in the disease setting, immunohistochemical analyses of tissues from patients with breast cancer verified markedly increased expression of the α2 subunit in vivo. These results not only highlight the utility of this discovery platform for rapidly selecting and characterizing function-blocking, anticancer mAbs in an unbiased fashion, but also identify α2β1 as a potential target in human carcinomatous states.
Collapse
|
160
|
Lee JT, Pamir N, Liu NC, Kirk EA, Averill MM, Becker L, Larson I, Hagman DK, Foster-Schubert KE, van Yserloo B, Bornfeldt KE, LeBoeuf RC, Kratz M, Heinecke JW. Macrophage metalloelastase (MMP12) regulates adipose tissue expansion, insulin sensitivity, and expression of inducible nitric oxide synthase. Endocrinology 2014; 155:3409-20. [PMID: 24914938 PMCID: PMC4138576 DOI: 10.1210/en.2014-1037] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Macrophage metalloelastase, a matrix metallopeptidase (MMP12) predominantly expressed by mature tissue macrophages, is implicated in pathological processes. However, physiological functions for MMP12 have not been described. Because mRNA levels for the enzyme increase markedly in adipose tissue of obese mice, we investigated the role of MMP12 in adipose tissue expansion and insulin resistance. In humans, MMP12 expression correlated positively and significantly with insulin resistance, TNF-α expression, and the number of CD14(+)CD206(+) macrophages in adipose tissue. MMP12 was the most abundant matrix metallopeptidase detected by proteomic analysis of conditioned medium of M2 macrophages and dendritic cells. In contrast, it was detected only at low levels in bone marrow derived macrophages and M1 macrophages. When mice received a high-fat diet, adipose tissue mass increased and CD11b(+)F4/80(+)CD11c(-) macrophages accumulated to a greater extent in MMP12-deficient (Mmp12(-/-)) mice than in wild-type mice (Mmp12(+/+)). Despite being markedly more obese, fat-fed Mmp12(-/-) mice were more insulin sensitive than fat-fed Mmp12(+/+) mice. Expression of inducible nitric oxide synthase (Nos2) by Mmp12(-/-) macrophages was significantly impaired both in vivo and in vitro, suggesting that MMP12 might mediate nitric oxide production during inflammation. We propose that MMP12 acts as a double-edged sword by promoting insulin resistance while combatting adipose tissue expansion.
Collapse
Affiliation(s)
- Jung-Ting Lee
- Departments of Medicine (J.-T.L., N.P., N.-C.L., M.M.A., L.B., K.E.F.-S., B.V.Y., K.E.B., R.C.L., M.K., J.W.H.), Pathology (K.E.B.), and Epidemiology (E.A.K., M.K.), University of Washington, Seattle, Washington 98105; and Fred Hutchinson Cancer Research Center (D.K.H., M.K.), Public Health Sciences, Seattle, Washington 98103
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Gai X, Tu K, Lu Z, Zheng X. MRC2 expression correlates with TGFβ1 and survival in hepatocellular carcinoma. Int J Mol Sci 2014; 15:15011-25. [PMID: 25162823 PMCID: PMC4200867 DOI: 10.3390/ijms150915011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/10/2014] [Accepted: 08/20/2014] [Indexed: 02/04/2023] Open
Abstract
MRC2 (Mannose Receptor C Type 2) is a constitutively recycling endocytic receptor belonging to the mannose receptor family, which has been found to be closely involved with cancer metastasis. This study attempted to determine MRC2 expression on hepatocellular carcinoma (HCC) and its significance on postsurgical prognosis of HCCs. The expression of both MRC2 and transforming growth factor (TGFβ1) was detected in tumor tissues and adjacent liver tissues from 96 HCCs by immunohistochemistry staining, and it was found that MRC2 expression in HCC tissues was significantly higher than in adjacent liver tissues. HCCs with higher MRC2 expression had worse prognosis after liver resection. Univariate analysis showed that advanced TNM staging of HCC, higher Edmonson-Steiner classification, intrahepatic metastases, portal vein invasion, higher MRC2 and higher TGFβ1 were the poor prognostic factors. Furthermore, multivariate analysis revealed that intrahepatic metastases, higher MRC2 and higher TGFβ1 were the independent prognostic factors. TGFβ1 treatment up-regulated MRC2 expression, cell migration and invasion of Huh7 cells notably. In addition, knockdown of MRC2 repressed the effect of TGFβ1 on cell migration and invasion. These data suggest that MRC2 overexpression predicts poor prognosis of HCCs after liver resection and MRC2 potentially contributed to TGFβ1-driven up-regulation of cell migration and invasion in HCC.
Collapse
Affiliation(s)
- Xiaohong Gai
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Zhongtang Lu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Xin Zheng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
162
|
Alizadeh AM, Shiri S, Farsinejad S. Metastasis review: from bench to bedside. Tumour Biol 2014; 35:8483-523. [PMID: 25104089 DOI: 10.1007/s13277-014-2421-z] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer is the final result of uninhibited cell growth that involves an enormous group of associated diseases. One major aspect of cancer is when cells attack adjacent components of the body and spread to other organs, named metastasis, which is the major cause of cancer-related mortality. In developing this process, metastatic cells must successfully negotiate a series of complex steps, including dissociation, invasion, intravasation, extravasation, and dormancy regulated by various signaling pathways. In this review, we will focus on the recent studies and collect a comprehensive encyclopedia in molecular basis of metastasis, and then we will discuss some new potential therapeutics which target the metastasis pathways. Understanding the new aspects on molecular mechanisms and signaling pathways controlling tumor cell metastasis is critical for the development of therapeutic strategies for cancer patients that would be valuable for researchers in both fields of molecular and clinical oncology.
Collapse
Affiliation(s)
- Ali Mohammad Alizadeh
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, 1419733141, Iran,
| | | | | |
Collapse
|
163
|
Mori S, Kiuchi S, Ouchi A, Hase T, Murase T. Characteristic expression of extracellular matrix in subcutaneous adipose tissue development and adipogenesis; comparison with visceral adipose tissue. Int J Biol Sci 2014; 10:825-33. [PMID: 25076859 PMCID: PMC4115194 DOI: 10.7150/ijbs.8672] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 06/16/2014] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is a connective tissue specified for energy metabolism and endocrines, but functional differences between subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) have not been fully elucidated. To reveal the physiological role of SAT, we characterized in vivo tissue development and in vitro adipocyte differentiation. In a DNA microarray analysis of SAT and VAT in Wistar rats, functional annotation clusters of extracellular matrix (ECM)-related genes were found in SAT, and major ECM molecules expressed in adipose tissues were profiled. In a histological analysis and quantitative expression analysis, ECM expression patterns could be classified into two types: (i) a histogenesis-correlated type such as type IV and XV collagen, and laminin subunits, (ii) a high-SAT expression type such as type I, III, and V collagen and minor characteristic collagens. Type (i) was related to basal membrane and up-regulated in differentiated 3T3-L1 cells and in histogenesis at depot-specific timings. In contrast, type (ii) was related to fibrous forming and highly expressed in 3T3-L1 preadipocytes. Exceptionally, fibronectin was abundant in developed adipose tissue, although it was highly expressed in 3T3-L1 preadipocytes. The present study showed that adipose tissues site-specifically regulate molecular type and timing of ECM expression, and suggests that these characteristic ECM molecules provide a critical microenvironment, which may affect bioactivity of adipocyte itself and interacts with other tissues. It must be important to consider the depot-specific property for the treatment of obesity-related disorders, dermal dysfunction and for the tissue regeneration.
Collapse
Affiliation(s)
- Shinobu Mori
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-Machi, Haga-gun, Tochigi, 321-3497, Japan
| | - Satomi Kiuchi
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-Machi, Haga-gun, Tochigi, 321-3497, Japan
| | - Atsushi Ouchi
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-Machi, Haga-gun, Tochigi, 321-3497, Japan
| | - Tadashi Hase
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-Machi, Haga-gun, Tochigi, 321-3497, Japan
| | - Takatoshi Murase
- Biological Science Laboratories, Kao Corporation, 2606 Akabane, Ichikai-Machi, Haga-gun, Tochigi, 321-3497, Japan
| |
Collapse
|
164
|
Abstract
Intratumoral hypoxia is a common feature of solid tumors. Recent advances in cancer biology indicate that hypoxia is not only a consequence of unrestrained tumor growth, but also plays an active role in promoting tumor progression, malignancy, and resistance to therapy. Hypoxia signaling is mediated by the hypoxia-inducible factors (HIFs), which are not only stabilized under hypoxia, but also by activated oncogenes or inactivated tumor suppressors under normoxia. Hypoxia is a prominent feature of the tumor microenvironment of pancreatic tumors, also characterized by the presence of a fibrotic reaction that promotes, and is also modulated by, hypoxia. As the mechanisms by which hypoxia signaling impacts invasion and metastasis in pancreatic cancer are being elucidated, hypoxia is emerging as a key determinant of pancreatic cancer malignancy as well as an important target for therapy. Herein we present an overview of recent advances in the understanding of the impact that hypoxia has in pancreatic cancer invasion and metastasis.
Collapse
Affiliation(s)
- Angela Yuen
- Tumor Microenvironment and Metastasis Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Begoña Díaz
- Tumor Microenvironment and Metastasis Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
165
|
Golubkov VS, Prigozhina NL, Zhang Y, Stoletov K, Lewis JD, Schwartz PE, Hoffman RM, Strongin AY. Protein-tyrosine pseudokinase 7 (PTK7) directs cancer cell motility and metastasis. J Biol Chem 2014; 289:24238-49. [PMID: 25006253 DOI: 10.1074/jbc.m114.574459] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is well established that widely expressed PTK7 is essential for vertebrate tissue morphogenesis. In cancer, the functionality of PTK7 is selectively regulated by membrane type-1 matrix metalloproteinase (MT1-MMP), ADAMs (a disintegrin domain and metalloproteinases), and γ-secretase proteolysis. Here, we established that the full-length membrane PTK7, its Chuzhoi mutant with the two functional MT1-MMP cleavage sites, and its L622D mutant with the single inactivated MT1-MMP cleavage site differentially regulate cell motility in a two-dimensional versus three-dimensional environment. We also demonstrated that in polarized cancer cells, the levels of PTK7 expression and proteolysis were directly linked to the structure and kinetics of cell protrusions, including lamellipodia and invadopodia. In the functionally relevant and widely accepted animal models of metastasis, mouse and chick embryo models, both the overexpression and knock-out of PTK7 in HT1080 cells abrogated metastatic dissemination. Our analysis of human tissue specimens confirmed intensive proteolysis of PTK7 in colorectal cancer tumors, but not in matching normal tissue. Our results provide convincing evidence that both PTK7 expression and proteolysis, rather than the level of the cellular full-length PTK7 alone, contribute to efficient directional cell motility and metastasis in cancer.
Collapse
Affiliation(s)
- Vladislav S Golubkov
- From the Sanford-Burnham Medical Research Institute, La Jolla, California 92037,
| | | | - Yong Zhang
- AntiCancer, Inc., San Diego, California 92111
| | | | - John D Lewis
- the Department of Oncology, University of Alberta, Edmonton T6G 2E1, Canada
| | | | - Robert M Hoffman
- AntiCancer, Inc., San Diego, California 92111, the Department of Surgery, University of California, San Diego, California 92103
| | - Alex Y Strongin
- From the Sanford-Burnham Medical Research Institute, La Jolla, California 92037,
| |
Collapse
|
166
|
de Jong PR, Mo JH, Harris AR, Lee J, Raz E. STAT3: An Anti-Invasive Factor in Colorectal Cancer? Cancers (Basel) 2014; 6:1394-407. [PMID: 24995503 PMCID: PMC4190547 DOI: 10.3390/cancers6031394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/13/2014] [Accepted: 06/20/2014] [Indexed: 02/07/2023] Open
Abstract
Signal Transducer and Activator of Transcription 3 (STAT3) is activated in a majority of cancers, and promotes tumorigenesis and even metastasis through transcriptional activation of its target genes. Recently, we discovered that STAT3 suppresses epithelial-to-mesenchymal transition (EMT) and thus metastasis in a mouse model of colorectal cancer (CRC), while it did not affect the overall tumor burden. Furthermore, we found that STAT3 in intestinal epithelial cells (IEC) suppresses EMT by regulating stability of an EMT inducer, SNAI-1 (Snail-1). Here, STAT3 functions as an adaptor rather than a transcription factor in the post-translational modification of SNAI-1. In this review, we discuss the unexpected and contradictory role of STAT3 in metastasis of CRC and its clinical implications.
Collapse
Affiliation(s)
- Petrus Rudolf de Jong
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr. MC 0663, La Jolla, CA 92093, USA.
| | - Ji-Hun Mo
- Department of Otorhinolaryngology, Dankook University College of Medicine, 16-5 Anseo-dong, Cheonan, Chungcheongnam-do 330-715, Korea.
| | - Alexandra R Harris
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr. MC 0663, La Jolla, CA 92093, USA.
| | - Jongdae Lee
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr. MC 0663, La Jolla, CA 92093, USA.
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr. MC 0663, La Jolla, CA 92093, USA.
| |
Collapse
|
167
|
Hong Y, Ku M, Heo D, Hwang S, Lee E, Park J, Choi J, Jung Lee H, Seo M, Jig Lee E, In Yook J, Haam S, Huh YM, Sung Yoon D, Suh JS, Yang J. Molecular recognition of proteolytic activity in metastatic cancer cells using fluorogenic gold nanoprobes. Biosens Bioelectron 2014; 57:171-8. [DOI: 10.1016/j.bios.2014.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/05/2014] [Accepted: 02/05/2014] [Indexed: 12/19/2022]
|
168
|
Abstract
Discoidin domain receptor 2 (DDR2) is an atypical receptor tyrosine kinase that binds to and is activated by collagen in the extracellular matrix. Recent exon sequencing studies have identified DDR2 to be mutated with a 3% to 4% incidence in squamous cell cancers of the lung. This article summarizes the current state of knowledge of DDR2 biology and signaling in lung squamous cell cancer. It also explores the context-dependent role of this receptor as both an oncogene and a tumor suppressor in cancer cells. Promising therapeutic opportunities based on existing and novel targeted small molecule inhibitors against DDR2 may provide new strategies for treating lung squamous cell cancer patients.
Collapse
Affiliation(s)
- Leo S. Payne
- Division of Cancer Biology, Institute of Cancer Research, London SW3 6JB, UK
| | - Paul H. Huang
- Division of Cancer Biology, Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
169
|
Ma YC, Fan WJ, Rao SM, Gao L, Bei ZY, Xu ST. Effect of Furin inhibitor on lung adenocarcinoma cell growth and metastasis. Cancer Cell Int 2014; 14:43. [PMID: 24876827 PMCID: PMC4037552 DOI: 10.1186/1475-2867-14-43] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/11/2014] [Indexed: 01/08/2023] Open
Abstract
Background To investigate the mechanisms of lung adenocarcinoma cell metastasis and provide a theoretical basis for the in-depth study of lung adenocarcinoma. Methods A549 cells are incubated with different concentrations of Furin inhibitor for indicated times. The proliferation and migration were confirmed with MTT, colony formation, wound Healing and Transwell assayes. Hochest 33342 / PI double staining was used to detect apoptosis. Cell migration and apoptosis associated proteins were analysed by enzyme-linked immunosorbent assay (ELISA) and western blot. Results We have found that Furin inhibitor play a significant role in inhibition A549 cell growth. And we also found cell migration was inhibited significantly upon Furin inhibitor treatment. Conclusion The proliferration and migration of A549 cell were inhibited by Furin inbitor through down-regulation the expression of migration and apoptosis related proteins.
Collapse
Affiliation(s)
- Yong-Chao Ma
- Luo He Medical Colledge, Daxue Road, #148, Luohe City, Henan Province 462002, P.R. China
| | - Wen-Juan Fan
- Luo He Medical Colledge, Daxue Road, #148, Luohe City, Henan Province 462002, P.R. China
| | - Shu-Mei Rao
- Luo He Medical Colledge, Daxue Road, #148, Luohe City, Henan Province 462002, P.R. China
| | - Li Gao
- Luo He Medical Colledge, Daxue Road, #148, Luohe City, Henan Province 462002, P.R. China
| | - Zhan-Yu Bei
- Luo He Medical Colledge, Daxue Road, #148, Luohe City, Henan Province 462002, P.R. China
| | - Song-Tao Xu
- Luo He Medical Colledge, Daxue Road, #148, Luohe City, Henan Province 462002, P.R. China
| |
Collapse
|
170
|
Williams KC, McNeilly RE, Coppolino MG. SNAP23, Syntaxin4, and vesicle-associated membrane protein 7 (VAMP7) mediate trafficking of membrane type 1-matrix metalloproteinase (MT1-MMP) during invadopodium formation and tumor cell invasion. Mol Biol Cell 2014; 25:2061-70. [PMID: 24807903 PMCID: PMC4072579 DOI: 10.1091/mbc.e13-10-0582] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The SNAREs SNAP23, Syntaxin4, and VAMP7 associate to target the delivery of MT1-MMP to sites of invadopodium formation in breast tumor cells. The interaction of these SNAREs correlates with decreased phosphorylation of Syntaxin4. The targeted delivery of MT1-MMP is required for efficient ECM degradation and cell invasion. Movement through the extracellular matrix (ECM) requires cells to degrade ECM components, primarily through the action of matrix metalloproteinases (MMPs). Membrane type 1–matrix metalloproteinase (MT1-MMP) has an essential role in matrix degradation and cell invasion and localizes to subcellular degradative structures termed invadopodia. Trafficking of MT1-MMP to invadopodia is required for the function of these structures, and here we examine the role of N-ethylmaleimide–sensitive factor–activating protein receptor (SNARE)–mediated membrane traffic in the transport of MT1-MMP to invadopodia. During invadopodium formation in MDA-MB-231 human breast cancer cells, increased association of SNAP23, Syntaxin4, and vesicle-associated membrane protein 7 (VAMP7) is detected by coimmunoprecipitation. Blocking the function of these SNAREs perturbs invadopodium-based ECM degradation and cell invasion. Increased level of SNAP23-Syntaxin4-VAMP7 interaction correlates with decreased Syntaxin4 phosphorylation. These results reveal an important role for SNARE-regulated trafficking of MT1-MMP to invadopodia during cellular invasion of ECM.
Collapse
Affiliation(s)
- Karla C Williams
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Rachael E McNeilly
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Marc G Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
171
|
Rossé C, Lodillinsky C, Fuhrmann L, Nourieh M, Monteiro P, Irondelle M, Lagoutte E, Vacher S, Waharte F, Paul-Gilloteaux P, Romao M, Sengmanivong L, Linch M, van Lint J, Raposo G, Vincent-Salomon A, Bièche I, Parker PJ, Chavrier P. Control of MT1-MMP transport by atypical PKC during breast-cancer progression. Proc Natl Acad Sci U S A 2014; 111:E1872-9. [PMID: 24753582 PMCID: PMC4020077 DOI: 10.1073/pnas.1400749111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Dissemination of carcinoma cells requires the pericellular degradation of the extracellular matrix, which is mediated by membrane type 1-matrix metalloproteinase (MT1-MMP). In this article, we report a co-up-regulation and colocalization of MT1-MMP and atypical protein kinase C iota (aPKCι) in hormone receptor-negative breast tumors in association with a higher risk of metastasis. Silencing of aPKC in invasive breast-tumor cell lines impaired the delivery of MT1-MMP from late endocytic storage compartments to the surface and inhibited matrix degradation and invasion. We provide evidence that aPKCι, in association with MT1-MMP-containing endosomes, phosphorylates cortactin, which is present in F-actin-rich puncta on MT1-MMP-positive endosomes and regulates cortactin association with the membrane scission protein dynamin-2. Thus, cell line-based observations and clinical data reveal the concerted activity of aPKC, cortactin, and dynamin-2, which control the trafficking of MT1-MMP from late endosome to the plasma membrane and play an important role in the invasive potential of breast-cancer cells.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adult
- Aged
- Biological Transport, Active
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Cell Line, Tumor
- Cortactin/metabolism
- Cytoplasmic Granules/metabolism
- Disease Progression
- Dynamin II/metabolism
- Endosomes/metabolism
- Extracellular Matrix/metabolism
- Female
- Humans
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Matrix Metalloproteinase 14/genetics
- Matrix Metalloproteinase 14/metabolism
- Middle Aged
- Neoplasm Invasiveness
- Phosphorylation
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/genetics
- Protein Kinase C/metabolism
- RNA Interference
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA, Small Interfering/genetics
- Up-Regulation
Collapse
Affiliation(s)
- Carine Rossé
- Research Center, Institut Curie, 75005 Paris, France
- Membrane and Cytoskeleton Dynamics, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Catalina Lodillinsky
- Research Center, Institut Curie, 75005 Paris, France
- Membrane and Cytoskeleton Dynamics, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | | | - Maya Nourieh
- Research Center, Institut Curie, 75005 Paris, France
| | - Pedro Monteiro
- Research Center, Institut Curie, 75005 Paris, France
- Membrane and Cytoskeleton Dynamics, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie, University of Paris VI, Institut de Formation Doctorale, 75252 Paris Cedex 5, France
| | - Marie Irondelle
- Research Center, Institut Curie, 75005 Paris, France
- Membrane and Cytoskeleton Dynamics, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Emilie Lagoutte
- Research Center, Institut Curie, 75005 Paris, France
- Membrane and Cytoskeleton Dynamics, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - François Waharte
- Research Center, Institut Curie, 75005 Paris, France
- Cell and Tissue Imaging Facility, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Perrine Paul-Gilloteaux
- Research Center, Institut Curie, 75005 Paris, France
- Cell and Tissue Imaging Facility, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Maryse Romao
- Research Center, Institut Curie, 75005 Paris, France
- Structure and Membrane Compartments, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Lucie Sengmanivong
- Research Center, Institut Curie, 75005 Paris, France
- Cell and Tissue Imaging Facility, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
- Nikon Imaging Centre, Institut Curie, Centre National de la Recherche Scientifique, 75005 Paris, France
| | - Mark Linch
- Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, United Kingdom
| | - Johan van Lint
- Department of Molecular Cell Biology, Faculty of Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Graça Raposo
- Research Center, Institut Curie, 75005 Paris, France
- Structure and Membrane Compartments, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| | - Anne Vincent-Salomon
- Research Center, Institut Curie, 75005 Paris, France
- Department of Tumor Biology, Institut Curie, 75005 Paris, France
- Institut National de la Santé et de la Recherche Médicale U830, 75005 Paris, France; and
| | - Ivan Bièche
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Peter J. Parker
- Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, United Kingdom
- Division of Cancer Studies, King’s College London, Guy’s Campus, London WC2A 3LY, United Kingdom
| | - Philippe Chavrier
- Research Center, Institut Curie, 75005 Paris, France
- Membrane and Cytoskeleton Dynamics, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75005 Paris, France
| |
Collapse
|
172
|
Wang C, Tang Z, Zhao Y, Yao R, Li L, Sun W. Three-dimensional
in vitro
cancer models: a short review. Biofabrication 2014; 6:022001. [DOI: 10.1088/1758-5082/6/2/022001] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
173
|
Turturici G, Tinnirello R, Sconzo G, Geraci F. Extracellular membrane vesicles as a mechanism of cell-to-cell communication: advantages and disadvantages. Am J Physiol Cell Physiol 2014; 306:C621-33. [DOI: 10.1152/ajpcell.00228.2013] [Citation(s) in RCA: 326] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Microvesicles represent a newly identified mechanism of intercellular communication. Two different types of microvesicles have been identified: membrane-derived vesicles (EVs) and exosomes. EVs originate by direct budding from the plasma membrane, while exosomes arise from ectocytosis of multivesicular bodies. Recent attention has focused on the capacity of EVs to alter the phenotype of neighboring cells to make them resemble EV-producing cells. Stem cells are an abundant source of EVs, and the interaction between stem cells and the microenvironment (i.e., stem cell niche) plays a critical role in determining stem cell phenotype. The stem cell niche hypothesis predicts that stem cell number is limited by the availability of niches releasing the necessary signals for self-renewal and survival, and the niche thus provides a mechanism for controlling and limiting stem cell numbers. EVs may play a fundamental role in this context by transferring genetic information between cells. EVs can transfer mRNA and microRNA to target cells, both of which may be involved in the change in target-cell phenotype towards that of EV-producing cells. The exchange of genetic information may be bidirectional, and EV-mediated transfer of genetic information after tissue damage may reprogram stem cells to acquire the phenotypic features of the injured tissue cells. In addition, stem cell-derived EVs may induce the de-differentiation of cells that survive injury by promoting their reentry into the cell cycle and subsequently increasing the possibility of tissue regeneration.
Collapse
Affiliation(s)
- Giuseppina Turturici
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, University of Palermo, Palermo, Italy
| | - Rosaria Tinnirello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, University of Palermo, Palermo, Italy
| | - Gabriella Sconzo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, University of Palermo, Palermo, Italy
| | - Fabiana Geraci
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, University of Palermo, Palermo, Italy
| |
Collapse
|
174
|
Heo SH, Cho JY. ELK3 suppresses angiogenesis by inhibiting the transcriptional activity of ETS-1 on MT1-MMP. Int J Biol Sci 2014; 10:438-47. [PMID: 24719561 PMCID: PMC3979996 DOI: 10.7150/ijbs.8095] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 03/07/2014] [Indexed: 11/05/2022] Open
Abstract
Ets transcription factors play important roles in vasculogenesis and angiogenesis. Knockout of the Ets gene family members in mice resulted in disrupted angiogenesis and malformed vascular systems. In this study, the role and mechanism of ELK3, an Ets factor, in angiogenesis was investigated using ELK3-specific siRNA in human vascular endothelial cells (HUVECs) and in vivo implantation assay. The suppression of ELK3 expression resulted in the reinforcement of VEGF-induced tube formation in HUVECs. The in vivo Matrigel plug assay also showed that ELK3 knockdown resulted in increased angiogenesis. Luciferase activity of the MT1-MMP promoter induced by ETS-1 factor was attenuated ELK3 co-transfection. CHIP assay showed the binding of ELK3 on the MT1-MMP promoter. MT1-MMP knockdown in the ELK3 knockdowned cells resulted in the decrease of tube formation suggesting that MT1-MMP transcriptional repression is required for ELK3-mediated anti-angiogenesis effect. Our data also showed that the suppressive effect of ELK3 on the angiogenesis was partly due to the inhibitory effect of ELK3 to the ETS-1 transcriptional activity on the MT1-MMP promoter rather than direct suppression of ELK3 on the target gene, since the expression level of co-repressor Sin3A is low in endothelial cells. Our results suggest that ELK3 plays a negative role of VEGF-induced angiogenesis through indirectly inhibiting ETS-1 function.
Collapse
Affiliation(s)
- Sun-Hee Heo
- Department of Veterinary Biochemistry, BK21 Plus, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Je-Yoel Cho
- Department of Veterinary Biochemistry, BK21 Plus, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
175
|
Hayashi M, Nomoto S, Hishida M, Inokawa Y, Kanda M, Okamura Y, Nishikawa Y, Tanaka C, Kobayashi D, Yamada S, Nakayama G, Fujii T, Sugimoto H, Koike M, Fujiwara M, Takeda S, Kodera Y. Identification of the collagen type 1 α 1 gene (COL1A1) as a candidate survival-related factor associated with hepatocellular carcinoma. BMC Cancer 2014; 14:108. [PMID: 24552139 PMCID: PMC4015503 DOI: 10.1186/1471-2407-14-108] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 02/13/2014] [Indexed: 12/15/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the major causes of cancer-related death especially among Asian and African populations. It is urgent that we identify carcinogenesis-related genes to establish an innovative treatment strategy for this disease. Methods Triple-combination array analysis was performed using one pair each of HCC and noncancerous liver samples from a 68-year-old woman. This analysis consists of expression array, single nucleotide polymorphism array and methylation array. The gene encoding collagen type 1 alpha 1 (COL1A1) was identified and verified using HCC cell lines and 48 tissues from patients with primary HCC. Results Expression array revealed that COL1A1 gene expression was markedly decreased in tumor tissues (log2 ratio –1.1). The single nucleotide polymorphism array showed no chromosomal deletion in the locus of COL1A1. Importantly, the methylation value in the tumor tissue was higher (0.557) than that of the adjacent liver tissue (0.008). We verified that expression of this gene was suppressed by promoter methylation. Reactivation of COL1A1 expression by 5-aza-2′-deoxycytidine treatment was seen in HCC cell lines, and sequence analysis identified methylated CpG sites in the COL1A1 promoter region. Among 48 pairs of surgical specimens, 13 (27.1%) showed decreased COL1A1 mRNA expression in tumor sites. Among these 13 cases, 10 had promoter methylation at the tumor site. The log-rank test indicated that mRNA down-regulated tumors were significantly correlated with a poor overall survival rate (P = 0.013). Conclusions Triple-combination array analysis successfully identified COL1A1 as a candidate survival-related gene in HCCs. Epigenetic down-regulation of COL1A1 mRNA expression might have a role as a prognostic biomarker of HCC.
Collapse
Affiliation(s)
| | - Shuji Nomoto
- Gastroenterological Surgery (Department of Surgery II), Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Willis AL, Sabeh F, Li XY, Weiss SJ. Extracellular matrix determinants and the regulation of cancer cell invasion stratagems. J Microsc 2014; 251:250-60. [PMID: 23924043 DOI: 10.1111/jmi.12064] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/13/2013] [Indexed: 12/13/2022]
Abstract
During development, wound repair and disease-related processes, such as cancer, normal, or neoplastic cell types traffic through the extracellular matrix (ECM), the complex composite of collagens, elastin, glycoproteins, proteoglycans, and glycosaminoglycans that dictate tissue architecture. Current evidence suggests that tissue-invasive processes may proceed by protease-dependent or protease-independent strategies whose selection is not only governed by the characteristics of the motile cell population, but also by the structural properties of the intervening ECM. Herein, we review the mechanisms by which ECM dimensionality, elasticity, crosslinking, and pore size impact patterns of cell invasion. This summary should prove useful when designing new experimental approaches for interrogating invasion programs as well as identifying potential cellular targets for next-generation therapeutics.
Collapse
Affiliation(s)
- A L Willis
- Division of Molecular Medicine & Genetics, Department of Internal Medicine, and the Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
177
|
Matrix metalloproteinases: the gene expression signatures of head and neck cancer progression. Cancers (Basel) 2014; 6:396-415. [PMID: 24531055 PMCID: PMC3980592 DOI: 10.3390/cancers6010396] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/14/2014] [Accepted: 01/29/2014] [Indexed: 11/17/2022] Open
Abstract
Extracellular matrix degradation by matrix metalloproteinases (MMPs) plays a pivotal role in cancer progression by promoting motility, invasion and angiogenesis. Studies have shown that MMP expression is increased in head and neck squamous cell carcinomas (HNSCCs), one of the most common cancers in the world, and contributes to poor outcome. In this review, we examine the expression pattern of MMPs in HNSCC by microarray datasets and summarize the current knowledge of MMPs, specifically MMP-1, -3, -7 -10, -12, -13, 14 and -19, that are highly expressed in HNSCCs and involved cancer invasion and angiogenesis.
Collapse
|
178
|
Alcantara MB, Dass CR. Pigment epithelium-derived factor as a natural matrix metalloproteinase inhibitor: a comparison with classical matrix metalloproteinase inhibitors used for cancer treatment. J Pharm Pharmacol 2014; 66:895-902. [PMID: 24697787 DOI: 10.1111/jphp.12218] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/23/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVES In the 1990s, the discovery of the important role of matrix metalloproteinases (MMPs) in cancer angiogenesis, growth and metastasis galvanised research efforts to search for ways to inhibit these MMPs. To date, this has resulted in the investigation of approximately 50 MMPIs which have undergone various phases of clinical trials. However, despite a large body of research being devoted to discovery and development of MMPIs, results have largely not been supportive of this approach to anticancer treatment. KEY FINDINGS The reasons for the general failure of these drugs in clinical trials include various unwanted side-effects, the use of healthy volunteers to provide drug dosages which did not correctly reflect dosages for cancer patients, and the exclusion of patients with early stage cancer in clinical trials despite MMPs being determined to be critical for the angiogenic switch, a process associated with early tumour growth. In contrast, a naturally-occurring endogenous protein and a non-functional serine protease inhibitor (serpin), pigment epithelium-derived factor (PEDF), has been proposed for cancer therapy partly due to its ability to regulate specific MMPs central to cancer progression. SUMMARY PEDF has been found to specifically downregulate membrane-type I matrix metalloproteinase (MT1-MMP) and furthermore, potentially matrix metalloproteinase-2 (MMP-2), two of the most commonly implicated MMPs in neoplasia.
Collapse
Affiliation(s)
- Marice B Alcantara
- College of Health and Biomedicine, Victoria University, St Albans, Australia
| | | |
Collapse
|
179
|
Jujo T, Sakao S, Tsukahara M, Kantake M, Kantake S, Maruoka M, Tanabe N, Masuda M, Tatsumi K. The role of matrix metalloproteinase in the intimal sarcoma-like cells derived from endarterectomized tissues from a chronic thromboembolic pulmonary hypertension patient. PLoS One 2014; 9:e87489. [PMID: 24489925 PMCID: PMC3905027 DOI: 10.1371/journal.pone.0087489] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/21/2013] [Indexed: 01/26/2023] Open
Abstract
Sarcoma-like cells (SCLs) were derived from endarterectomized tissue of a single chronic thromboembolic pulmonary hypertension (CTEPH) patient during incubation of those thrombi at second passage as described at our previous report. These cells had malignant potential, with an increased expression of matrix metalloproteinase-14 (MMP-14), leading to tumor emboli within pulmonary arteries in in vivo studies. The purpose of this study was to perform a more detailed evaluation of the characteristics of SCLs, and to elucidate the role of the increased expression of MMP-14 expression in the growth and death of these cells. In order to elucidate the characteristics of SCLs and to confirm the protein expression of MMP-14, three-dimentional culture, invasion assays, a Western blot analysis and immunohistochemical studies were performed. To examine the role of MMP-14 in tumorigenesis, the metalloproteinase inhibitor, batimastat, was administered to SCID mice which were subcutaneously injected with SCLs. Those mice were sacrificed on day 14 and the tumor volume was evaluated. A Western blot analysis showed the increased expression of MMP-14 in comparison to the expression in lung adenocarcinoma cells (A549). Immunohistochemistry showed that SCLs were positive for vimentin, MMP-14, MMP-2 and CD44. However, endothelial markers, such as CD31 and von Willebrand factor (vWF), were negative. The in vivo studies demonstrated that batimastat could suppress the growth of the subcutaneous tumors formed by the SCLs. This study suggested that MMPs had critical roles on the pathological activities of SCLs and that batimastat might have anti-proliferative and anti-invasive effects on these cells.
Collapse
Affiliation(s)
- Takayuki Jujo
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, Inohana Chuo-Ku, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, Inohana Chuo-Ku, Chiba, Japan
| | - Masanori Tsukahara
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, Inohana Chuo-Ku, Chiba, Japan
| | | | - Seiji Kantake
- Respirology, Kimitsu Chuo Hospital, Sakurai, Kisarazu City, Japan
| | - Miki Maruoka
- Respirology, National Hospital Organization Chiba Medical Center, Tsubakimori, Chuo-ku, Chiba, Japan
| | - Nobuhiro Tanabe
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, Inohana Chuo-Ku, Chiba, Japan
| | - Masahisa Masuda
- Cardiovascular Surgery, National Hospital Organization Chiba Medical Center, Tsubakimori, Chuo-ku, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, Inohana Chuo-Ku, Chiba, Japan
| |
Collapse
|
180
|
Weaver SA, Wolters B, Ito N, Woskowicz AM, Kaneko K, Shitomi Y, Seiki M, Itoh Y. Basal localization of MT1-MMP is essential for epithelial cell morphogenesis in 3D collagen matrix. J Cell Sci 2014; 127:1203-13. [PMID: 24463815 PMCID: PMC4117704 DOI: 10.1242/jcs.135236] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The membrane-anchored collagenase membrane type 1 matrix metalloprotease (MT1-MMP) has been shown to play an essential role during epithelial tubulogenesis in 3D collagen matrices; however, its regulation during tubulogenesis is not understood. Here, we report that degradation of collagen in polarized epithelial cells is post-translationally regulated by changing the localization of MT1-MMP from the apical to the basal surface. MT1-MMP predominantly localizes at the apical surface in inert polarized epithelial cells, whereas treatment with HGF induced basal localization of MT1-MMP followed by collagen degradation. The basal localization of MT1-MMP requires the ectodomains of the enzyme because deletion of the MT-loop region or the hemopexin domain inhibited basal localization of the enzyme. TGFβ is a well-known inhibitor of tubulogenesis and our data indicate that its mechanism of inhibition is, at least in part, due to inhibition of MT1-MMP localization to the basal surface. Interestingly, however, the effect of TGFβ was found to be bi-phasic: at high doses it effectively inhibited basal localization of MT1-MMP, whereas at lower doses tubulogenesis and basal localization of MT1-MMP was promoted. Taken together, these data indicate that basal localization of MT1-MMP is a key factor promoting the degradation of extracellular matrix by polarized epithelial cells, and that this is an essential part of epithelial morphogenesis in 3D collagen.
Collapse
Affiliation(s)
- Sarah A Weaver
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, UK
| | | | | | | | | | | | | | | |
Collapse
|
181
|
Shiryaev SA, Remacle AG, Golubkov VS, Ingvarsen S, Porse A, Behrendt N, Cieplak P, Strongin AY. A monoclonal antibody interferes with TIMP-2 binding and incapacitates the MMP-2-activating function of multifunctional, pro-tumorigenic MMP-14/MT1-MMP. Oncogenesis 2013; 2:e80. [PMID: 24296749 PMCID: PMC3940861 DOI: 10.1038/oncsis.2013.44] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/29/2013] [Indexed: 01/01/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and, especially membrane type 1 (MT1)-MMP/MMP-14, are promising drug targets in malignancies. In contrast with multiple small-molecule and protein pan-inhibitors of MT1–MMP cleavage activity, the murine 9E8 monoclonal antibody targets the MMP-2-activating function of cellular MT1–MMP alone, rather than the general proteolytic activity and the pro-migratory function of MT1–MMP. Furthermore, the antibody does not interact in any detectable manner with other members of the membrane type (MT)-MMP family. The mechanism of this selectivity remained unknown. Using mutagenesis, binding and activity assays, and modeling in silico, we have demonstrated that the 9E8 antibody recognizes the MT-loop structure, an eight residue insertion that is specific for MT–MMPs and that is distant from the MT1–MMP active site. The binding of the 9E8 antibody to the MT-loop, however, prevents tissue inhibitor of metalloproteinases-2 (TIMP-2) association with MT1–MMP. As a result, the 9E8 antibody incapacitates the TIMP-2-dependent MMP-2-activating function alone rather than the general enzymatic activity of human MT1–MMP. The specific function of the 9E8 antibody we determined directly supports an essential, albeit paradoxical, role of the protein inhibitor (TIMP-2) in MMP-2 activation via a unique membrane-tethered mechanism. In this mechanism, the formation of a tri-molecular MT1–MMPTIMP-2MMP-2 complex is required for both the capture of the soluble MMP-2 proenzyme by cells and then its well-controlled conversion into the mature MMP-2 enzyme. In sum, understanding of the structural requirements for the 9E8 antibody specificity may pave the way for the focused design of the inhibitory antibodies against other individual MMPs.
Collapse
Affiliation(s)
- S A Shiryaev
- Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
182
|
Phosphoproteomics of collagen receptor networks reveals SHP-2 phosphorylation downstream of wild-type DDR2 and its lung cancer mutants. Biochem J 2013; 454:501-13. [PMID: 23822953 PMCID: PMC3893797 DOI: 10.1042/bj20121750] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Collagen is an important extracellular matrix component that directs many fundamental cellular processes including differentiation, proliferation and motility. The signalling networks driving these processes are propagated by collagen receptors such as the β1 integrins and the DDRs (discoidin domain receptors). To gain an insight into the molecular mechanisms of collagen receptor signalling, we have performed a quantitative analysis of the phosphorylation networks downstream of collagen activation of integrins and DDR2. Temporal analysis over seven time points identified 424 phosphorylated proteins. Distinct DDR2 tyrosine phosphorylation sites displayed unique temporal activation profiles in agreement with in vitro kinase data. Multiple clustering analysis of the phosphoproteomic data revealed several DDR2 candidate downstream signalling nodes, including SHP-2 (Src homology 2 domain-containing protein tyrosine phosphatase 2), NCK1 (non-catalytic region of tyrosine kinase adaptor protein 1), LYN, SHIP-2 [SH2 (Src homology 2)-domain-containing inositol phosphatase 2], PIK3C2A (phosphatidylinositol-4-phosphate 3-kinase, catalytic subunit type 2α) and PLCL2 (phospholipase C-like 2). Biochemical validation showed that SHP-2 tyrosine phosphorylation is dependent on DDR2 kinase activity. Targeted proteomic profiling of a panel of lung SCC (squamous cell carcinoma) DDR2 mutants demonstrated that SHP-2 is tyrosine-phosphorylated by the L63V and G505S mutants. In contrast, the I638F kinase domain mutant exhibited diminished DDR2 and SHP-2 tyrosine phosphorylation levels which have an inverse relationship with clonogenic potential. Taken together, the results of the present study indicate that SHP-2 is a key signalling node downstream of the DDR2 receptor which may have therapeutic implications in a subset of DDR2 mutations recently uncovered in genome-wide lung SCC sequencing screens.
Collapse
|
183
|
Hypoxia-inducible factor 1 regulation through cross talk between mTOR and MT1-MMP. Mol Cell Biol 2013; 34:30-42. [PMID: 24164895 DOI: 10.1128/mcb.01169-13] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) plays a key role in the cellular adaptation to hypoxia. Although HIF-1 is usually strongly suppressed by posttranslational mechanisms during normoxia, HIF-1 is active and enhances tumorigenicity in malignant tumor cells that express the membrane protease MT1-MMP. The cytoplasmic tail of MT1-MMP, which can bind a HIF-1 suppressor protein called factor inhibiting HIF-1 (FIH-1), promotes inhibition of FIH-1 by Mint3 during normoxia. To explore possible links between HIF-1 activation by MT1-MMP/Mint3 and tumor growth signals, we surveyed a panel of 252 signaling inhibitors. The mTOR inhibitor rapamycin was identified as a possible modulator, and it inhibited the mTOR-dependent phosphorylation of Mint3 that is required for FIH-1 inhibition. A mutant Mint3 protein that cannot be phosphorylated exhibited a reduced ability to inhibit FIH-1 and promoted tumor formation in mice. These data suggest a novel molecular link between the important hub proteins MT1-MMP and mTOR that contributes to tumor malignancy.
Collapse
|
184
|
Woskowicz AM, Weaver SA, Shitomi Y, Ito N, Itoh Y. MT-LOOP-dependent localization of membrane type I matrix metalloproteinase (MT1-MMP) to the cell adhesion complexes promotes cancer cell invasion. J Biol Chem 2013; 288:35126-37. [PMID: 24165131 DOI: 10.1074/jbc.m113.496067] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Localization of membrane type I matrix metalloproteinase (MT1-MMP) to the leading edge is thought to be a crucial step during cancer cell invasion. However, its mechanisms and functional impact on cellular invasion have not been clearly defined. In this report, we have identified the MT-LOOP, a loop region in the catalytic domain of MT1-MMP ((163)PYAYIREG(170)), as an essential region for MT1-MMP to promote cellular invasion. Deletion of the MT-LOOP effectively inhibited functions of MT1-MMP on the cell surface, including proMMP-2 activation, degradation of gelatin and collagen films, and cellular invasion into a collagen matrix. This is not due to loss of the catalytic function of MT1-MMP but due to inefficient localization of the enzyme to β1-integrin-rich cell adhesion complexes at the plasma membrane. We also found that an antibody that specifically recognizes the MT-LOOP region of MT1-MMP (LOOPAb) inhibited MT1-MMP functions, fully mimicking the phenotype of the MT-LOOP deletion mutant. We therefore propose that the MT-LOOP region is an interface for molecular interactions that mediate enzyme localization to cell adhesion complexes and regulate MT1-MMP functions. Our findings have revealed a novel mechanism regulating MT1-MMP during cellular invasion and have identified the MT-LOOP as a potential exosite target region to develop selective MT1-MMP inhibitors.
Collapse
Affiliation(s)
- Anna M Woskowicz
- From the Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | | | | | | | | |
Collapse
|
185
|
Wawrzyniak JA, Bianchi-Smiraglia A, Bshara W, Mannava S, Ackroyd J, Bagati A, Omilian AR, Im M, Fedtsova N, Miecznikowski JC, Moparthy KC, Zucker SN, Zhu Q, Kozlova NI, Berman AE, Hoek KS, Gudkov AV, Shewach DS, Morrison CD, Nikiforov MA. A purine nucleotide biosynthesis enzyme guanosine monophosphate reductase is a suppressor of melanoma invasion. Cell Rep 2013; 5:493-507. [PMID: 24139804 DOI: 10.1016/j.celrep.2013.09.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/20/2013] [Accepted: 09/11/2013] [Indexed: 01/02/2023] Open
Abstract
Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.
Collapse
Affiliation(s)
- Joseph A Wawrzyniak
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
DcR3 regulates the growth and metastatic potential of SW480 colon cancer cells. Oncol Rep 2013; 30:2741-8. [PMID: 24101127 DOI: 10.3892/or.2013.2769] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/02/2013] [Indexed: 11/05/2022] Open
Abstract
Decoy receptor 3 (DcR3) is considered to have anti‑apoptotic and pro-metastatic functions, suggesting it might be a therapeutic target. We examined the role and mechanisms of DcR3 on growth and the metastatic ability of SW480 colon cancer cells to provide therapeutic information for targeting DcR3 by RNA interference (RNAi) technology. Growth and the metastatic ability were inhibited, apoptosis was induced and cell cycle profile was changed after decreasing DcR3 expression, with lower levels of vascular endothelial growth factors (VEGFs) and matrix metalloproteinases (MMPs) expression. Our results implied the therapeutic potential of silencing DcR3 expression by RNAi in colon cancer.
Collapse
|
187
|
Christian L, Bahudhanapati H, Wei S. Extracellular metalloproteinases in neural crest development and craniofacial morphogenesis. Crit Rev Biochem Mol Biol 2013; 48:544-60. [PMID: 24066766 DOI: 10.3109/10409238.2013.838203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The neural crest (NC) is a population of migratory stem/progenitor cells that is found in early vertebrate embryos. NC cells are induced during gastrulation, and later migrate to multiple destinations and contribute to many types of cells and tissues, such as craniofacial structures, cardiac tissues, pigment cells and the peripheral nervous system. Recently, accumulating evidence suggests that many extracellular metalloproteinases, including matrix metalloproteinases (MMPs), a disintegrin and metalloproteinases (ADAMs), and ADAMs with thrombospondin motifs (ADAMTSs), play important roles in various stages of NC development. Interference with metalloproteinase functions often causes defects in craniofacial structures, as well as in other cells and tissues that are contributed by NC cells, in humans and other vertebrates. In this review, we summarize the current state of the field concerning the roles of these three families of metalloproteinases in NC development and related tissue morphogenesis, with a special emphasis on craniofacial morphogenesis.
Collapse
Affiliation(s)
- Laura Christian
- Department of Biology, West Virginia University , Morgantown, WV , USA
| | | | | |
Collapse
|
188
|
Wolf K, Te Lindert M, Krause M, Alexander S, Te Riet J, Willis AL, Hoffman RM, Figdor CG, Weiss SJ, Friedl P. Physical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction force. ACTA ACUST UNITED AC 2013; 201:1069-84. [PMID: 23798731 PMCID: PMC3691458 DOI: 10.1083/jcb.201210152] [Citation(s) in RCA: 990] [Impact Index Per Article: 82.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell migration through 3D tissue depends on a physicochemical balance between cell deformability and physical tissue constraints. Migration rates are further governed by the capacity to degrade ECM by proteolytic enzymes, particularly matrix metalloproteinases (MMPs), and integrin- and actomyosin-mediated mechanocoupling. Yet, how these parameters cooperate when space is confined remains unclear. Using MMP-degradable collagen lattices or nondegradable substrates of varying porosity, we quantitatively identify the limits of cell migration by physical arrest. MMP-independent migration declined as linear function of pore size and with deformation of the nucleus, with arrest reached at 10% of the nuclear cross section (tumor cells, 7 µm²; T cells, 4 µm²; neutrophils, 2 µm²). Residual migration under space restriction strongly depended upon MMP-dependent ECM cleavage by enlarging matrix pore diameters, and integrin- and actomyosin-dependent force generation, which jointly propelled the nucleus. The limits of interstitial cell migration thus depend upon scaffold porosity and deformation of the nucleus, with pericellular collagenolysis and mechanocoupling as modulators.
Collapse
Affiliation(s)
- Katarina Wolf
- Department of Cell Biology, Radboud University Nijmegen Medical Centre, 6500 HB Nijmegen, Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Lengyel E, Burdette JE, Kenny HA, Matei D, Pilrose J, Haluska P, Nephew KP, Hales DB, Stack MS. Epithelial ovarian cancer experimental models. Oncogene 2013; 33:3619-33. [PMID: 23934194 DOI: 10.1038/onc.2013.321] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 12/13/2022]
Abstract
Epithelial ovarian cancer (OvCa) is associated with high mortality and, as the majority (>75%) of women with OvCa have metastatic disease at the time of diagnosis, rates of survival have not changed appreciably over 30 years. A mechanistic understanding of OvCa initiation and progression is hindered by the complexity of genetic and/or environmental initiating events and lack of clarity regarding the cell(s) or tissue(s) of origin. Metastasis of OvCa involves direct extension or exfoliation of cells and cellular aggregates into the peritoneal cavity, survival of matrix-detached cells in a complex ascites fluid phase and subsequent adhesion to the mesothelium lining covering abdominal organs to establish secondary lesions containing host stromal and inflammatory components. Development of experimental models to recapitulate this unique mechanism of metastasis presents a remarkable scientific challenge, and many approaches used to study other solid tumors (for example, lung, colon and breast) are not transferable to OvCa research given the distinct metastasis pattern and unique tumor microenvironment (TME). This review will discuss recent progress in the development and refinement of experimental models to study OvCa. Novel cellular, three-dimensional organotypic, and ex vivo models are considered and the current in vivo models summarized. The review critically evaluates currently available genetic mouse models of OvCa, the emergence of xenopatients and the utility of the hen model to study OvCa prevention, tumorigenesis, metastasis and chemoresistance. As these new approaches more accurately recapitulate the complex TME, it is predicted that new opportunities for enhanced understanding of disease progression, metastasis and therapeutic response will emerge.
Collapse
Affiliation(s)
- E Lengyel
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - J E Burdette
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois/Chicago, Chicago, IL, USA
| | - H A Kenny
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - D Matei
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J Pilrose
- Medical Sciences, Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Bloomington, IN, USA
| | - P Haluska
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - K P Nephew
- Medical Sciences, Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Bloomington, IN, USA
| | - D B Hales
- Department of Physiology, Southern Illinois University, Carbondale, IL, USA
| | - M S Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| |
Collapse
|
190
|
Collaborative overexpression of matrix metalloproteinase-1 and vascular endothelial growth factor-C predicts adverse prognosis in patients with gliomas. Cancer Epidemiol 2013; 37:697-702. [PMID: 23870768 DOI: 10.1016/j.canep.2013.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/16/2013] [Accepted: 06/18/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIM Matrix metalloproteinase-1 (MMP-1), a member of the MMP family of zinc-dependent endopeptidases, has been detected to be strongly expressed in gliomas with high tumor grade and to be correlated with increased tumor invasiveness. Vascular endothelial growth factor-C (VEGF-C), which is able to induce MMP-1 transcription, has been found to be upregulated in glioblastoma compared to low grade gliomas and non-neoplastic brain. The aim of the present study was to investigate the clinical significance of the co-expression of MMP-1 and VEGF-C in glioma patients on determining the prognosis. METHODS One hundred and sixteen glioma patients (26 World Health Organization (WHO) grade I, 30 WHO grade II, 30 WHO grade III, and 30 WHO grade IV) and 15 non-neoplastic brain specimens acquired from 15 patients undergoing surgery for epilepsy as control were collected. Immunohistochemistry was used to evaluate the expression of MMP-1 and VEGF-C in glioma and non-neoplastic brain tissues. The correlations of collaborative MMP-1 and VEGF-C expression with selected clinicopathologic parameters and clinical outcome of glioma patients were also assessed. RESULTS Both MMP-1 and VEGF-C expression were significantly higher in glioma tissues compared to non-neoplastic brain tissues (both P<0.001). Of 116 glioma patients, 68 (58.62%) overexpressed MMP-1 and VEGF-C simultaneously. In addition, combined MMP-1 and VEGF-C expression was significantly associated with WHO grade (P<0.001) and Karnofsky performance status (KPS) score (P=0.01). Moreover, glioma patients expressing both MMP-1 and VEGF-C exhibited markedly poorer overall survival (P<0.001). According to the multivariate analyses, collaborative overexpression of MMP-1 and VEGF-C was found to be an independent prognostic factor for overall survival (P=0.009). CONCLUSIONS Our data demonstrated for the first time that overexpression of both MMP-1 and VEGF-C may be an independent poor prognostic factor in gliomas, suggesting the interaction between MMP-1 and VEGF-C collaboratively stimulated advanced tumor progression and adverse outcome. Inhibiting both MMP-1 and VEGF-C could be a novel therapeutic approach for gliomas.
Collapse
|
191
|
Leight JL, Alge DL, Maier AJ, Anseth KS. Direct measurement of matrix metalloproteinase activity in 3D cellular microenvironments using a fluorogenic peptide substrate. Biomaterials 2013; 34:7344-52. [PMID: 23830581 DOI: 10.1016/j.biomaterials.2013.06.023] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 06/12/2013] [Indexed: 12/21/2022]
Abstract
Incorporation of degradable moieties into synthetic hydrogels has greatly increased the utility of these three-dimensional matrices for in vitro cell culture as well as tissue engineering applications. A common method for introducing degradability is the inclusion of oligopeptides sensitive to cleavage by matrix metalloproteinases (MMPs), enabling cell-mediated remodeling and migration within the material. While this strategy has been effective, characterization and measurement of cell-mediated degradation in these materials has remained challenging. There are 20+ MMP family members whose activity is regulated in space and time by a number of biochemical and biophysical cues. Thus, the typical approach of characterizing cleavage of degradable moieties in solution with recombinant enzymes does not easily translate to three-dimensional cell-mediated matrix remodeling. To address this challenge, we report here the synthesis of a cell-laden hydrogel matrix functionalized with a fluorogenic peptide substrate to provide real-time, quantitative monitoring of global MMP activity. Using this system, stimulation of MMP activity was observed with growth factor treatment in mammary epithelial cells and compared to classical zymography results. Further, the effect of biophysical cues on MMP activity of human mesenchymal stem cells was also investigated where more rigid hydrogels were observed to increase MMP activity. The regulation of MMP activity by these biochemical and biophysical cues highlights the need for in situ, real-time measurement of hydrogel degradation, and use of these functionalized hydrogels will aid in future rational design of degradable synthetic hydrogels for in vitro cell studies and tissue engineering applications.
Collapse
Affiliation(s)
- Jennifer L Leight
- Howard Hughes Medical Institute and the BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, USA
| | | | | | | |
Collapse
|
192
|
Sugiyama N, Gucciardo E, Tatti O, Varjosalo M, Hyytiäinen M, Gstaiger M, Lehti K. EphA2 cleavage by MT1-MMP triggers single cancer cell invasion via homotypic cell repulsion. ACTA ACUST UNITED AC 2013; 201:467-84. [PMID: 23629968 PMCID: PMC3639392 DOI: 10.1083/jcb.201205176] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Metalloproteinase-mediated cleavage of EphA2 induces breast tumor cells to shift from collective invasion to single-cell invasion. Changes in EphA2 signaling can affect cancer cell–cell communication and motility through effects on actomyosin contractility. However, the underlying cell–surface interactions and molecular mechanisms of how EphA2 mediates these effects have remained unclear. We demonstrate here that EphA2 and membrane-anchored membrane type-1 matrix metalloproteinase (MT1-MMP) were selectively up-regulated and coexpressed in invasive breast carcinoma cells, where, upon physical interaction in same cell–surface complexes, MT1-MMP cleaved EphA2 at its Fibronectin type-III domain 1. This cleavage, coupled with EphA2-dependent Src activation, triggered intracellular EphA2 translocation, as well as an increase in RhoA activity and cell junction disassembly, which suggests an overall repulsive effect between cells. Consistent with this, cleavage-prone EphA2-D359I mutant shifted breast carcinoma cell invasion from collective to rounded single-cell invasion within collagen and in vivo. Up-regulated MT1-MMP also codistributed with intracellular EphA2 in invasive cells within human breast carcinomas. These results reveal a new proteolytic regulatory mechanism of cell–cell signaling in cancer invasion.
Collapse
Affiliation(s)
- Nami Sugiyama
- Research Programs Unit, Genome-Scale Biology, Haartman Institute, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
193
|
Lee H, Chang KW, Yang HY, Lin PW, Chen SU, Huang YL. MT1-MMP regulates MMP-2 expression and angiogenesis-related functions in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2013; 437:232-8. [PMID: 23796708 DOI: 10.1016/j.bbrc.2013.06.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 01/06/2023]
Abstract
Membrane type 1 (MT1)-MMP is a member of matrix metalloproteinases (MMPs) that regulates extracellular matrix remodeling. In addition, MT1-MMP also serves as a multi-functional protein. However, the functional role of MT1-MMP in human endothelial cells remains unclear. In this study we use real-time PCR and Western blotting to demonstrate for the first time that MMP-2 expression is regulated by MT1-MMP in human endothelial cells. Moreover, MMP-2 activity is also modulated by MT1-MMP. In addition we found that endothelial cells, ECM adhesion and human endothelial cell tube formation, which are known to be regulated by MMP-2, are blocked by MT1-MMP siRNA. These results suggest that MT1-MMP plays an important role in regulating angiogenesis in human endothelial cells.
Collapse
Affiliation(s)
- Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
194
|
Goh SK, Bertera S, Olsen P, Candiello JE, Halfter W, Uechi G, Balasubramani M, Johnson SA, Sicari BM, Kollar E, Badylak SF, Banerjee I. Perfusion-decellularized pancreas as a natural 3D scaffold for pancreatic tissue and whole organ engineering. Biomaterials 2013; 34:6760-72. [PMID: 23787110 DOI: 10.1016/j.biomaterials.2013.05.066] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/24/2013] [Indexed: 01/13/2023]
Abstract
Approximately 285 million people worldwide suffer from diabetes, with insulin supplementation as the most common treatment measure. Regenerative medicine approaches such as a bioengineered pancreas has been proposed as potential therapeutic alternatives. A bioengineered pancreas will benefit from the development of a bioscaffold that supports and enhances cellular function and tissue development. Perfusion-decellularized organs are a likely candidate for use in such scaffolds since they mimic compositional, architectural and biomechanical nature of a native organ. In this study, we investigate perfusion-decellularization of whole pancreas and the feasibility to recellularize the whole pancreas scaffold with pancreatic cell types. Our result demonstrates that perfusion-decellularization of whole pancreas effectively removes cellular and nuclear material while retaining intricate three-dimensional microarchitecture with perfusable vasculature and ductal network and crucial extracellular matrix (ECM) components. To mimic pancreatic cell composition, we recellularized the whole pancreas scaffold with acinar and beta cell lines and cultured up to 5 days. Our result shows successful cellular engraftment within the decellularized pancreas, and the resulting graft gave rise to strong up-regulation of insulin gene expression. These findings support biological utility of whole pancreas ECM as a biomaterials scaffold for supporting and enhancing pancreatic cell functionality and represent a step toward bioengineered pancreas using regenerative medicine approaches.
Collapse
Affiliation(s)
- Saik-Kia Goh
- Department of Bioengineering, University of Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Takino T, Guo L, Domoto T, Sato H. MT1-MMP prevents growth inhibition by three dimensional fibronectin matrix. Biochem Biophys Res Commun 2013; 436:503-8. [PMID: 23756810 DOI: 10.1016/j.bbrc.2013.05.134] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 05/30/2013] [Indexed: 11/18/2022]
Abstract
The extracellular microenvironment plays a key role in regulation of cellular functions and growth control. We show here that membrane-type 1 matrix metalloproteinase (MT1-MMP) acts as a growth promoter in confluent culture. When MT1-MMP was silenced in HT1080 fibrosarcoma cells, cells created three dimensional (3D) fibronectin matrix in a confluent culture, and growth of cells embedded within it was retarded. Formation of 3D fibronectin matrix initiated by MT1-MMP silencing was impeded by knockdown of either FN or integrin β₁, which resulted in restoration of cell growth. When cells in 3D fibronectin matrix were treated with integrin β₁ inhibitory antibody, cells underwent S phase entry. These results suggest that MT1-MMP prevents growth suppression by 3D fibronectin matrix, which is mediated through integrin β₁.
Collapse
Affiliation(s)
- Takahisa Takino
- Department of Molecular Virology and Oncology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| | | | | | | |
Collapse
|
196
|
Remacle AG, Shiryaev SA, Golubkov VS, Freskos JN, Brown MA, Karwa AS, Naik AD, Howard CP, Sympson CJ, Strongin AY. Non-destructive and selective imaging of the functionally active, pro-invasive membrane type-1 matrix metalloproteinase (MT1-MMP) enzyme in cancer cells. J Biol Chem 2013; 288:20568-80. [PMID: 23733191 DOI: 10.1074/jbc.m113.471508] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Proteolytic activity of cell surface-associated MT1-matrix metalloproteinase (MMP) (MMP-14) is directly related to cell migration, invasion, and metastasis. MT1-MMP is regulated as a proteinase by activation and conversion of the latent proenzyme into the active enzyme, and also via inhibition by tissue inhibitors of MMPs (TIMPs) and self-proteolysis. MT1-MMP is also regulated as a membrane protein through its internalization and recycling. Routine immunohistochemistry, flow cytometry, reverse transcription-PCR, and immunoblotting methodologies do not allow quantitative imaging and assessment of the cell-surface levels of the active, TIMP-free MT1-MMP enzyme. Here, we developed a fluorescent reporter prototype that targets the cellular active MT1-MMP enzyme alone. The reporter (MP-3653) represents a liposome tagged with a fluorochrome and functionalized with a PEG chain spacer linked to an inhibitory hydroxamate warhead. Our studies using the MP-3653 reporter and its inactive derivative demonstrated that MP-3653 can be efficiently used not only to visualize the trafficking of MT1-MMP through the cell compartment, but also to quantify the femtomolar range amounts of the cell surface-associated active MT1-MMP enzyme in multiple cancer cell types, including breast carcinoma, fibrosarcoma, and melanoma. Thus, the levels of the naturally expressed, fully functional, active cellular MT1-MMP enzyme are roughly equal to 1 × 10(5) molecules/cell, whereas these levels are in a 1 × 10(6) range in the cells with the enforced MT1-MMP expression. We suggest that the reporter we developed will contribute to the laboratory studies of MT1-MMP and then, ultimately, to the design of novel, more efficient prognostic approaches and personalized cancer therapies.
Collapse
Affiliation(s)
- Albert G Remacle
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Shin Y, Kim H, Han S, Won J, Jeong HE, Lee E, Kamm RD, Kim J, Chung S. Extracellular matrix heterogeneity regulates three-dimensional morphologies of breast adenocarcinoma cell invasion. Adv Healthc Mater 2013. [PMID: 23184641 DOI: 10.1002/adhm.201200320] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Plasticity and reciprocity of breast cancer cells to various extracellular matrice (ECMs) are three-dimensionally analyzed in quantitative way in a novel and powerful microfluidic in vitro platform. This successfully demonstrates the metastatic potential of cancer cells and their effective strategies of ECM proteolytic remodeling and morphological change, while interacting with other cells and invading into heterogeneous ECMs.
Collapse
Affiliation(s)
- Yoojin Shin
- School of Mechanical Engineering, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐713, Korea
| | - Hyunju Kim
- College of Life Science and Biotechnology, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐701, Korea
| | - Sewoon Han
- School of Mechanical Engineering, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐713, Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐713, Korea
| | - Hyo Eun Jeong
- School of Mechanical Engineering, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐713, Korea
| | - Eun‐Sook Lee
- National Cancer Center, Madu 1‐dong, Ilsandong‐gu, Goyang‐si, Gyeonggi‐do 410‐769, Korea
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jae‐Hong Kim
- College of Life Science and Biotechnology, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐701, Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Anam‐Dong, Seongbuk‐Gu, Seoul 136‐713, Korea
| |
Collapse
|
198
|
Li CY, Wood DK, Huang JH, Bhatia SN. Flow-based pipeline for systematic modulation and analysis of 3D tumor microenvironments. LAB ON A CHIP 2013; 13:1969-78. [PMID: 23563587 PMCID: PMC3812960 DOI: 10.1039/c3lc41300d] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 03/07/2013] [Indexed: 05/05/2023]
Abstract
The cancer microenvironment, which incorporates interactions with stromal cells, extracellular matrix (ECM), and other tumor cells in a 3-dimensional (3D) context, has been implicated in every stage of cancer development, including growth of the primary tumor, metastatic spread, and response to treatment. Our understanding of the tumor microenvironment and our ability to develop new therapies would greatly benefit from tools that allow us to systematically probe microenvironmental cues within a 3D context. Here, we leveraged recent advances in microfluidic technology to develop a platform for high-throughput fabrication of tunable cellular microniches ("microtissues") that allow us to probe tumor cell response to a range of microenvironmental cues, including ECM, soluble factors, and stromal cells, all in 3D. We further combine this tunable microniche platform with rapid, flow-based population level analysis (n > 500), which permits analysis and sorting of microtissue populations both pre- and post-culture by a range of parameters, including proliferation and homotypic or heterotypic cell density. We used this platform to demonstrate differential responses of lung adenocarcinoma cells to a selection of ECM molecules and soluble factors. The cells exhibited enhanced or reduced proliferation when encapsulated in fibronectin- or collagen-1-containing microtissues, respectively, and they showed reduced proliferation in the presence of TGF-β, an effect that we did not observe in monolayer culture. We also measured tumor cell response to a panel of drug targets and found, in contrast to monolayer culture, specific sensitivity of tumor cells to TGFβR2 inhibitors, implying that TGF-β has an anti-proliferative affect that is unique to the 3D context and that this effect is mediated by TGFβR2. These findings highlight the importance of the microenvironmental context in therapeutic development and that the platform we present here allows the high-throughput study of tumor response to drugs as well as basic tumor biology in well-defined microenvironmental niches.
Collapse
Affiliation(s)
- Cheri Y. Li
- Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - David K. Wood
- Harvard-MIT Division of Health Sciences and Technology , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States .
| | - Joanne H. Huang
- Biology , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Sangeeta N. Bhatia
- Harvard-MIT Division of Health Sciences and Technology , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States .
- Broad Institute , Cambridge , Massachusetts 02142 , United States
- Department of Medicine , Brigham and Women's Hospital , Boston , Massachusetts 02115 , United States
- Electrical Engineering and Computer Science , David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
- Howard Hughes Medical Institute , Chevy Chase , Maryland 20815 , United States
| |
Collapse
|
199
|
Juncker-Jensen A, Deryugina EI, Rimann I, Zajac E, Kupriyanova TA, Engelholm LH, Quigley JP. Tumor MMP-1 activates endothelial PAR1 to facilitate vascular intravasation and metastatic dissemination. Cancer Res 2013; 73:4196-211. [PMID: 23687338 DOI: 10.1158/0008-5472.can-12-4495] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Intravasation, the active entry of primary tumor cells into the vasculature, remains the least studied step in the metastatic cascade. Protease-mediated escape and stromal invasion of tumor cells represent widely accepted processes leading up to the intravasation step. However, molecular factors that contribute directly to tumor cell vascular penetration have not been identified. In this study, the in vivo role of the collagenolytic protease, MMP-1, in cancer cell intravasation and metastasis was analyzed by using a highly disseminating variant of human HEp3 epidermoid carcinoma, HEp3-hi/diss. Although naturally acquired or experimentally induced MMP-1 deficiency substantially suppressed HEp3-hi/diss intravasation, supplementation of recombinant MMP-1 to MMP-1-silenced primary tumors restored their impaired vascular dissemination. Surprisingly, abrogation of MMP-1 production and activity did not significantly affect HEp3-hi/diss migration or matrix invasion, suggesting noncollagenolytic mechanisms underlying MMP-1-dependent cell intravasation. In support of such noncollagenolytic mechanisms, MMP-1 silencing in HEp3-hi/diss cells modulated the microarchitecture and integrity of the angiogenic vasculature in a novel microtumor model. Concomitantly, MMP-1 deficiency led to decreased levels of intratumoral vascular permeability, tumor cell intravasation, and metastatic dissemination. Taking advantage of PAR1 deficiency of HEp3-hi/diss cells, we further show that endothelial PAR1 is a putative nontumor-cell/nonmatrix target, activation of which by carcinoma-produced MMP-1 regulates endothelial permeability and transendothelial migration. The inhibitory effects of specific PAR1 antagonists in live animals have also indicated that the mechanisms of MMP-1-dependent vascular permeability in tumors involve endothelial PAR1 activation. Together, our findings mechanistically underscore the contribution of a tumor MMP-1/endothelial PAR1 axis to actual intravasation events manifested by aggressive carcinoma cells.
Collapse
Affiliation(s)
- Anna Juncker-Jensen
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
200
|
Aricò A, Giantin M, Gelain M, Riondato F, Mortarino M, Comazzi S, Dacasto M, Castagnaro M, Aresu L. Matrix metalloproteinases and vascular endothelial growth factor expression in canine leukaemias. Vet J 2013; 196:260-2. [DOI: 10.1016/j.tvjl.2012.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 09/14/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
|