101
|
Vilen ST, Suojanen J, Salas F, Risteli J, Ylipalosaari M, Itkonen O, Koistinen H, Baumann M, Stenman UH, Sorsa T, Salo T, Nyberg P. Trypsin-2 enhances carcinoma invasion by processing tight junctions and activating ProMT1-MMP. Cancer Invest 2012; 30:583-92. [PMID: 22909050 DOI: 10.3109/07357907.2012.716467] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Enhanced proteolysis and altered tight junction (TJ) proteins associate with carcinoma invasion. We hypothesized that trypsin-2, a tumor-associated serine proteinase, induces tongue carcinoma invasion by activating pro-membrane type-1 matrix metalloproteinase (MT1-MMP) and disturbing the TJs. The effects of invasion were analyzed using trypsin-2 over-expressing human tongue squamous cell carcinoma cells (Try2-HSC-3) in vitro and in vivo. The invasion of Try2-HSC-3 cells was increased in mouse xenografts and human organotypic model. Trypsin-2 activated proMT1-MMP, as well as altered the expression of TJ protein claudin-7. In conclusion, trypsin-2 over-expression enhanced tongue carcinoma cell invasion by various genetic and proteolytic mechanisms.
Collapse
Affiliation(s)
- Suvi-Tuuli Vilen
- Institute of Dentistry, University of Helsinki, and Department of Oral and Maxillofacial Diseases, Helsinki University Hospital, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Santos-Valle P, Guijarro-Muñoz I, Cuesta ÁM, Alonso-Camino V, Villate M, Álvarez-Cienfuegos A, Blanco FJ, Sanz L, Álvarez-Vallina L. The heterotrimeric laminin coiled-coil domain exerts anti-adhesive effects and induces a pro-invasive phenotype. PLoS One 2012; 7:e39097. [PMID: 22723936 PMCID: PMC3378518 DOI: 10.1371/journal.pone.0039097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 05/18/2012] [Indexed: 12/24/2022] Open
Abstract
Laminins are large heterotrimeric cross-shaped extracellular matrix glycoproteins with terminal globular domains and a coiled-coil region through which the three chains are assembled and covalently linked. Laminins are key components of basement membranes, and they serve as attachment sites for cell adhesion, migration and proliferation. In this work, we produced a recombinant fragment comprising the entire laminin coiled-coil of the α1-, β1-, and γ1-chains that assemble into a stable heterotrimeric coiled-coil structure independently of the rest of the molecule. This domain was biologically active and not only failed to serve as a substrate for cell attachment, spreading and focal adhesion formation but also inhibited cell adhesion to laminin when added to cells in a soluble form at the time of seeding. Furthermore, gene array expression profiling in cells cultured in the presence of the laminin coiled-coil domain revealed up-regulation of genes involved in cell motility and invasion. These findings were confirmed by real-time quantitative PCR and zymography assays. In conclusion, this study shows for the first time that the laminin coiled-coil domain displays anti-adhesive functions and has potential implications for cell migration during matrix remodeling.
Collapse
Affiliation(s)
- Patricia Santos-Valle
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Irene Guijarro-Muñoz
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Ángel M. Cuesta
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Vanesa Alonso-Camino
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Maider Villate
- Structural Biology Unit, CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
| | - Ana Álvarez-Cienfuegos
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Francisco J. Blanco
- Structural Biology Unit, CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
- * E-mail: (LS); (LA-V)
| | - Luis Álvarez-Vallina
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
- * E-mail: (LS); (LA-V)
| |
Collapse
|
103
|
Boudreau A, van't Veer LJ, Bissell MJ. An "elite hacker": breast tumors exploit the normal microenvironment program to instruct their progression and biological diversity. Cell Adh Migr 2012; 6:236-48. [PMID: 22863741 DOI: 10.4161/cam.20880] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The year 2011 marked the 40 year anniversary of Richard Nixon signing the National Cancer Act, thus declaring the beginning of the "War on Cancer" in the United States. Whereas we have made tremendous progress toward understanding the genetics of tumors in the past four decades, and in developing enabling technology to dissect the molecular underpinnings of cancer at unprecedented resolution, it is only recently that the important role of the stromal microenvironment has been studied in detail. Cancer is a tissue-specific disease, and it is becoming clear that much of what we know about breast cancer progression parallels the biology of the normal breast differentiation, of which there is still much to learn. In particular, the normal breast and breast tumors share molecular, cellular, systemic and microenvironmental influences necessary for their progression. It is therefore enticing to consider a tumor to be a "rogue hacker"--one who exploits the weaknesses of a normal program for personal benefit. Understanding normal mammary gland biology and its "security vulnerabilities" may thus leave us better equipped to target breast cancer. In this review, we will provide a brief overview of the heterotypic cellular and molecular interactions within the microenvironment of the developing mammary gland that are necessary for functional differentiation, provide evidence suggesting that similar biology--albeit imbalanced and exaggerated--is observed in breast cancer progression particularly during the transition from carcinoma in situ to invasive disease. Lastly we will present evidence suggesting that the multigene signatures currently used to model cancer heterogeneity and clinical outcome largely reflect signaling from a heterogeneous microenvironment-a recurring theme that could potentially be exploited therapeutically.
Collapse
Affiliation(s)
- Aaron Boudreau
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | | | | |
Collapse
|
104
|
Huber RJ, Suarez A, O'Day DH. CyrA, a matricellular protein that modulates cell motility in Dictyostelium discoideum. Matrix Biol 2012; 31:271-80. [PMID: 22391412 DOI: 10.1016/j.matbio.2012.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 02/02/2012] [Accepted: 02/14/2012] [Indexed: 01/16/2023]
Abstract
CyrA, an extracellular matrix (slime sheath), calmodulin (CaM)-binding protein in Dictyostelium discoideum, possesses four tandem EGF-like repeats in its C-terminus and is proteolytically cleaved during asexual development. A previous study reported the expression and localization of CyrA cleavage products CyrA-C45 and CyrA-C40. In this study, an N-terminal antibody was produced that detected the full-length 63kDa protein (CyrA-C63). Western blot analyses showed that the intracellular expression of CyrA-C63 peaked between 12 and 16h of development, consistent with the time that cells are developing into a motile, multicellular slug. CyrA immunolocalization and CyrA-GFP showed that the protein localized to the endoplasmic reticulum, particularly its perinuclear component. CyrA-C63 secretion began shortly after the onset of starvation peaking between 8 and 16h of development. A pharmacological analysis showed that CyrA-C63 secretion was dependent on intracellular Ca(2+) release and active CaM, PI3K, and PLA2. CyrA-C63 bound to CaM both intra- and extracellularly and both proteins were detected in the slime sheath deposited by migrating slugs. In keeping with its purported function, CyrA-GFP over-expression enhanced cAMP-mediated chemotaxis and CyrA-C45 was detected in vinculin B (VinB)-GFP immunoprecipitates, thus providing a link between the increase in chemotaxis and a specific cytoskeletal component. Finally, DdEGFL1-FITC was detected on the membranes of cells capped with concanavalin A suggesting that a receptor exists for this peptide sequence. Together with previous studies, the data presented here suggests that CyrA is a bona fide matricellular protein in D. discoideum.
Collapse
Affiliation(s)
- Robert J Huber
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON, Canada M5S 3G5.
| | | | | |
Collapse
|
105
|
Complex Regulation of the Pericellular Proteolytic Microenvironment during Tumor Progression and Wound Repair: Functional Interactions between the Serine Protease and Matrix Metalloproteinase Cascades. Biochem Res Int 2012; 2012:454368. [PMID: 22454771 PMCID: PMC3290807 DOI: 10.1155/2012/454368] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/21/2011] [Indexed: 01/08/2023] Open
Abstract
Spatial and temporal regulation of the pericellular proteolytic environment by local growth factors, such as EGF and TGF-β, initiates a wide repertoire of cellular responses coupled to a plasmin/matrix metalloproteinase (MMP) dependent stromal-remodeling axis. Cell motility and invasion, tumor metastasis, wound healing, and organ fibrosis, for example, represent diverse events controlled by expression of a subset of genes that encode various classes of tissue remodeling proteins. These include members of the serine protease and MMP families that functionally constitute a complex system of interacting protease cascades and titrated by their respective inhibitors. Several structural components of the extracellular matrix are upregulated by TGF-β as are matrix-active proteases (e.g., urokinase (uPA), plasmin, MMP-1, -3, -9, -10, -11, -13, -14). Stringent controls on serine protease/MMP expression and their topographic activity are essential for maintaining tissue homeostasis. Targeting individual elements in this highly interactive network may lead to novel therapeutic approaches for the treatment of cancer, fibrotic diseases, and chronic wounds.
Collapse
|
106
|
Ngora H, Galli UM, Miyazaki K, Zöller M. Membrane-bound and exosomal metastasis-associated C4.4A promotes migration by associating with the α(6)β(4) integrin and MT1-MMP. Neoplasia 2012; 14:95-107. [PMID: 22431918 PMCID: PMC3306255 DOI: 10.1593/neo.111450] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Revised: 11/28/2011] [Accepted: 11/29/2011] [Indexed: 12/19/2022]
Abstract
Metastasis-associated C4.4A, which becomes upregulated during wound healing and, in some tumors, during tumor progression, is known to be frequently associated with hypoxia. With the function of C4.4A still unknown, we explored the impact of hypoxia on C4.4A expression and functional activity. Metastatic rat and human tumor lines upregulate C4.4A expression when cultured in the presence of CoCl(2). Although hypoxia-inducible factor 1α (HIF-1α) becomes upregulated concomitantly, HIF-1α did not induce C4.4A transcription. Instead, hypoxia-induced C4.4A up-regulation promoted in vivo and in vitro wound healing, where increased migration on the C4.4A ligands laminin-111 and -332 was observed after a transient period of pronounced binding. Increased migration was accompanied by C4.4A associating with α(6)β(4), MT1-MMP1, and TACE and by laminin fragmentation. Hypoxia also promoted the release of C4.4A in exosomes and TACE-mediated C4.4A shedding. The association of C4.4A with α(6)β(4) and MT1-MMP1 was maintained in exosomes and exosomal α(6)β(4)- and MT1-MMP1-associated C4.4A but not shed C4.4A sufficient for laminin degradation. Hypoxia-induced recruitment of α(6)β(4) toward raft-located C4.4A, MT1-MMP, and TACE allows for a shift from adhesion to motility, which is supported by laminin degradation. These findings provide the first explanation for the C4.4A contribution to wound healing and metastasis.
Collapse
Affiliation(s)
- Honoré Ngora
- Department of Tumor Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Uwe M Galli
- Department of Tumor Cell Biology, University of Heidelberg, Heidelberg, Germany
| | | | - Margot Zöller
- Department of Tumor Cell Biology, University of Heidelberg, Heidelberg, Germany
- German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
107
|
Ding BS, Nolan DJ, Guo P, Babazadeh AO, Cao Z, Rosenwaks Z, Crystal RG, Simons M, Sato TN, Worgall S, Shido K, Rabbany SY, Rafii S. Endothelial-derived angiocrine signals induce and sustain regenerative lung alveolarization. Cell 2011; 147:539-53. [PMID: 22036563 DOI: 10.1016/j.cell.2011.10.003] [Citation(s) in RCA: 369] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 08/15/2011] [Accepted: 10/05/2011] [Indexed: 12/31/2022]
Abstract
To identify pathways involved in adult lung regeneration, we employ a unilateral pneumonectomy (PNX) model that promotes regenerative alveolarization in the remaining intact lung. We show that PNX stimulates pulmonary capillary endothelial cells (PCECs) to produce angiocrine growth factors that induce proliferation of epithelial progenitor cells supporting alveologenesis. Endothelial cells trigger expansion of cocultured epithelial cells, forming three-dimensional angiospheres reminiscent of alveolar-capillary sacs. After PNX, endothelial-specific inducible genetic ablation of Vegfr2 and Fgfr1 in mice inhibits production of MMP14, impairing alveolarization. MMP14 promotes expansion of epithelial progenitor cells by unmasking cryptic EGF-like ectodomains that activate the EGF receptor (EGFR). Consistent with this, neutralization of MMP14 impairs EGFR-mediated alveolar regeneration, whereas administration of EGF or intravascular transplantation of MMP14(+) PCECs into pneumonectomized Vegfr2/Fgfr1-deficient mice restores alveologenesis and lung inspiratory volume and compliance function. VEGFR2 and FGFR1 activation in PCECs therefore increases MMP14-dependent bioavailability of EGFR ligands to initiate and sustain alveologenesis.
Collapse
Affiliation(s)
- Bi-Sen Ding
- Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Fibrous stroma is associated with poorer prognosis in lung squamous cell carcinoma patients. J Thorac Oncol 2011; 6:1460-7. [PMID: 21849853 DOI: 10.1097/jto.0b013e318229189d] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Cancer tissue is composed of various stromal cells forming cancer-specific microenvironments. Peritumoral stroma is reportedly composed of activated fibroblasts that can influence the biological properties of tumor cells, mainly their local aggressiveness and their ability. The aim of this study was to examine whether the histological properties of peritumoral stroma are correlated with squamous cell carcinoma (SqCC) aggressiveness and clinical outcome. METHODS A series of 220 pathological stage I lung SqCC were categorized into two types according to the histological properties of the peritumoral stroma, "fibrous stroma type" (n = 85), and "thin stroma type" (n = 135), and compared the prognostic significance. Furthermore, we compared the immunohistochemical properties of the SqCC cells surrounded by "fibrous stroma" with those of the SqCC cells surrounded by "thin stroma." RESULTS The prognosis of the patients with fibrous stroma-type tumors was significantly poorer than that of the thin stroma type with regard to both recurrence-free survival (p = 0.005) and overall survival (p = 0.008). A multivariate analysis showed that the presence of a fibrous stroma was an independent prognostic factor (p = 0.030). Compared with the SqCC cells with a thin stroma, the SqCC cells with a fibrous stroma exhibited reduced expression of E-cadherin (55.9 versus 126.0, p < 0.001) and an increased expression of laminin-5γ2 (94.6 versus 25.0, p = 0.001), matrix metalloproteinase-7 (26.0 versus 3.50, p = 0.009), and c-Met (64.0 versus 36.5, p = 0.033). CONCLUSION SqCC with a fibrous stroma displayed higher invasive phenotype and were associated with a significantly poor prognosis. The current results suggest that the microenvironment created by both SqCC cells and the peritumoral fibroblasts may facilitate cancer aggressiveness.
Collapse
|
109
|
Keely PJ. Mechanisms by which the extracellular matrix and integrin signaling act to regulate the switch between tumor suppression and tumor promotion. J Mammary Gland Biol Neoplasia 2011; 16:205-19. [PMID: 21822945 PMCID: PMC3885166 DOI: 10.1007/s10911-011-9226-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 07/19/2011] [Indexed: 12/21/2022] Open
Abstract
Cell adhesion to the extracellular matrix (ECM) is necessary for development of the mammary gland, and to maintain the normal architecture and function of the gland. Cells adhere to the ECM via the integrin family of trans-membrane receptors, which signal to control mammary-specific gene expression and regulate cell proliferation and survival. During tumor formation, the ECM is extensively remodeled and signaling through integrins is altered such that cells become proliferative and invasive. A key regulator of whether integrin-mediated adhesion will promote tumor suppression or tumor formation is the stiffness of the stromal ECM. The normal mammary gland is typically surrounded by a loose collagenous stroma. An increase in the deposition of collagen and other stromal components is associated with mammographic density, which is one of the greatest risk factors for developing breast carcinoma. Several groups have demonstrated that increased stromal ECM density results in a matrix that is stiffer. Cells sense the stiffness of their surrounding ECM by Rho-mediated contraction of the actin-myosin cytoskeleton. If the surrounding ECM is stiffer than the cell's ability to contract it, then the tensile forces that result are able to drive the clustering of integrins and assemble adhesion signaling complexes. The result is subsequent activation of signaling pathways including FAK, ERK, and PI3K that drive cell proliferation and survival. In contrast, focal complexes are not formed in a compliant matrix, and activation of FAK and pERK is diminished, resulting in control of proliferation. Signaling from FAK moreover regulates p53 and miR-200 members, which control apoptosis and epithelial phenotype, such that a compliant matrix is predicted to promote normal mammary gland architecture and suppress tumor formation.
Collapse
Affiliation(s)
- Patricia J Keely
- Department of Cell and Regenerative Biology, Laboratory for Cellular and Molecular Biology, & Laboratory for Optical and Computational Instrumentation, University of Wisconsin, 227D Bock Laboratories, 1525 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
110
|
Expression of laminin 5-γ2 chain in cutaneous squamous cell carcinoma and its role in tumour invasion. Br J Cancer 2011; 105:824-32. [PMID: 21829200 PMCID: PMC3171006 DOI: 10.1038/bjc.2011.283] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Laminin-5 (Ln5), a heterotrimer composed of three chains (α3, β3, and γ2), is a major component of the basement membrane in most adult tissues. One of the chains, Ln5-γ2, is a marker of invasive tumours because it is frequently expressed as a monomer in malignant tumours. Recent studies from our laboratories detected higher levels of Ln5-γ2 expression in basal cell carcinoma (BCC) than in trichoblastoma. Furthermore, Ln5-γ2 overexpression tended to correlate with aggressiveness in BCC. Methods: In this study, we compared the expression of Ln5-γ2 in invasive squamous cell carcinoma (SCC, n=62) of the skin to that in preinvasive Bowen’s disease (BD, n=51), followed by analysis of the role of Ln5-γ2 in cancer invasion in vitro. Results: Immunohistochemically, the proportion of SCC cases (86%) strongly positive for Ln5-γ2 expression was higher than that of BD (16%). Real-time RT–PCR showed Ln5-γ2 overexpression in SCC cell line, A431, compared with normal keratinocyte cell line, HaCaT. Ln5-γ2 monomer and proteolytically cleaved, biologically active fragments of Ln5-γ2 were identified in SCC tumour extracts. In in vitro raft cultures, which simulate in vivo conditions, Ln5-γ2 siRNA significantly suppressed epidermal growth factor (EGF)-stimulated A431 cell invasion. Conclusion: Our results indicate that Ln5-γ2 has a role in cutaneous SCC invasion.
Collapse
|
111
|
Visser MB, Pollitt CC. The timeline of lamellar basement membrane changes during equine laminitis development. Equine Vet J 2011; 43:471-7. [PMID: 21496071 DOI: 10.1111/j.2042-3306.2010.00292.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
REASONS FOR PERFORMING STUDY The timing of lamellar basement membrane (BM) changes occurring during laminitis development is incompletely understood. OBJECTIVES To determine the temporal progression of lamellar BM changes and whether laminin-332 (Ln-332) γ2 cleavage products are generated during laminitis development. METHODS Eight clinically normal Standardbred horses were allocated into treatment (n = 5) or sham (n = 3) groups. The treatment group received, via nasogastric intubation, an oligofructose (OF) bolus (10 g/kg bwt) while the sham group was given water. Laminitis induction proceeded for 48 h followed by euthanasia. Lamellar biopsies were obtained prior to dosing and at intervals during the treatment period for analysis (at 12, 18, 24, 30 and 36 h and at 48 h following euthanasia). RESULTS Changes in lamellar collagen type IV and Ln-332 were first observed at 12 h post dosing. A unique pattern of reactivity for the Ln-332 γ2 antibody D4B5 occurred, in which reactivity was observed only in lamellar tissue affected by laminitis. No bioactive Ln-332 γ2 proteolytic fragments were detected in lamellar samples. CONCLUSIONS Basement membrane changes occurred early during the laminitis process. Direct Ln-332 γ2 cleavage to release biologically active products did not appear to occur. Thus loss of stability or protein interaction of the BM is probably responsible for the γ2 specific reactivity observed. POTENTIAL RELEVANCE Basement membrane changes may a first step in lamellar failure occurring prior to detection with conventional methods. Thus, more sensitive detection methods of BM changes are required to study laminitis development.
Collapse
Affiliation(s)
- M B Visser
- The Australian Equine Laminitis Research Unit, School of Veterinary Science, The University of Queensland, St Lucia, Australia
| | | |
Collapse
|
112
|
Han GY, Park SA, Kim JH, Lee EK, Kim HJ, Seo YK, Park JK, Kim CW. Effects of vibration on the proteome expression of anterior cruciate ligament cells. Exp Biol Med (Maywood) 2011; 236:783-9. [DOI: 10.1258/ebm.2011.010358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent reports have suggested that vibration has beneficial effects on knee healing response; however, the biomechanism of these beneficial effects still need to be determined on the anterior cruciate ligament (ACL) cell level. In this study, we applied a 20 Hz vibration to ACL cells, which produced a 20% increase ( P < 0.001) in cell activity and 17% increase ( P < 0.001) in intracellular sulfated glycosaminoglycan levels. In the 20 Hz vibration-stimulated ACL cell group, eight up-regulated (100 ∼ 300%) protein spots were identified compared with the control group by proteomics analysis. Among these proteins, Annexin A2 and Prolyl 4 hydroxylase (PH4B) were shown to have a 71% and 16% higher expression, respectively, in the 20 Hz vibration-stimulated ACL cell group by Western blotting ( P < 0.001). These results indicate that vibration produces a positive cellular environment, and Annexin A2 and prolyl 4 hydroxylase are expected to help ligament repair and ACL cell proliferation by controlling cell membrane and extracellular matrix formation.
Collapse
Affiliation(s)
- Gi-Yeon Han
- School of Life Sciences and Biotechnology, Korea University, 1-5, Anam Dong, Seongbuk-Gu, Seoul 136-701
| | - Seung-Ah Park
- School of Life Sciences and Biotechnology, Korea University, 1-5, Anam Dong, Seongbuk-Gu, Seoul 136-701
| | - Ji-Hye Kim
- School of Life Sciences and Biotechnology, Korea University, 1-5, Anam Dong, Seongbuk-Gu, Seoul 136-701
| | - Eun-Kyung Lee
- School of Life Sciences and Biotechnology, Korea University, 1-5, Anam Dong, Seongbuk-Gu, Seoul 136-701
| | - Hyun-Jung Kim
- School of Life Sciences and Biotechnology, Korea University, 1-5, Anam Dong, Seongbuk-Gu, Seoul 136-701
| | - Young-Kwon Seo
- Dongguk University Research Institute of Biotechnology, Dongguk University3-26, Pil Dong, Choong-Gu, Seoul 100-715, Korea
| | - Jung-Keug Park
- Dongguk University Research Institute of Biotechnology, Dongguk University3-26, Pil Dong, Choong-Gu, Seoul 100-715, Korea
| | - Chan-Wha Kim
- School of Life Sciences and Biotechnology, Korea University, 1-5, Anam Dong, Seongbuk-Gu, Seoul 136-701
| |
Collapse
|
113
|
González LO, González-Reyes S, Marín L, González L, González JM, Lamelas ML, Merino AM, Rodríguez E, Pidal I, del Casar JM, Andicoechea A, Vizoso F. Comparative analysis and clinical value of the expression of metalloproteases and their inhibitors by intratumour stromal mononuclear inflammatory cells and those at the invasive front of breast carcinomas. Histopathology 2011; 57:862-76. [PMID: 21166700 DOI: 10.1111/j.1365-2559.2010.03723.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Matrix metalloproteases (MMPs) and their inhibitors (TIMPs) play an essential role in the degradation of stromal connective tissue and basement membrane components. The aim of this study was to determine whether the dynamic analysis of these components can help to predict tumour aggressiveness. METHODS AND RESULTS An immunohistochemical study was performed using tissue arrays and specific antibodies against MMPs -1, -2, -7, -9, -11, -13 and -14 and TIMPs -1, -2 and -3. More than 5000 determinations on cancer specimens from 124 patients with invasive breast cancer were performed on the tumour centre core as well as on the invasive front. Immunostaining for MMPs/TIMPs on mononuclear inflammatory cells (MICs) was evaluated. To identify specific groups of tumours with distinct expression profiles, data obtained from both MICs populations were analysed by unsupervised hierarchical cluster analysis. When compared with MICs at the invasive front, intratumour MICs more frequently showed expression of MMP-7 and -1 and TIMP-3, but less frequently expression of MMP-9 and -11 and TIMP-2. CONCLUSIONS Our data led us to consider the need of further studies in order to identify subsets of MICs and other protein elements of the microenvironment as attractive targets for new therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Luis O González
- Unidad de Investigación, Fundación Hospital de Jove, Gijón, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Khokha R, Werb Z. Mammary gland reprogramming: metalloproteinases couple form with function. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a004333. [PMID: 21106646 DOI: 10.1101/cshperspect.a004333] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The adult mammary structure provides for the rapid growth, development, and immunological protection of the live-born young of mammals through its production of milk. The dynamic remodeling of the branched epithelial structure of the mammary gland in response to physiological stimuli that allow its programmed branching morphogenesis at puberty, cyclical turnover during the reproductive cycle, differentiation into a secretory organ at parturition, postlactational involution, and ultimately, regression with age is critical for these processes. Extracellular metalloproteinases are essential for the remodeling programs that operate in the tissue microenvironment at the interface of the epithelium and the stroma, coupling form with function. Deregulated proteolytic activity drives the transition of a physiological mammary microenvironment into a tumor microenvironment, facilitating malignant transformation.
Collapse
Affiliation(s)
- Rama Khokha
- Ontario Cancer Institute/University Health Network, University of Toronto, Ontario, Canada.
| | | |
Collapse
|
115
|
Suarez A, Huber RJ, Myre MA, O'Day DH. An extracellular matrix, calmodulin-binding protein from Dictyostelium with EGF-like repeats that enhance cell motility. Cell Signal 2011; 23:1197-206. [PMID: 21402150 DOI: 10.1016/j.cellsig.2011.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Accepted: 03/07/2011] [Indexed: 01/16/2023]
Abstract
CyrA is a novel cysteine-rich protein with four EGFL repeats that was isolated using the calmodulin (CaM) binding overlay technique (CaMBOT), suggesting it is a CaM-binding protein (CaMBP). The full-length 63kDa cyrA is cleaved into two major C-terminal fragments, cyrA-C45 and cyrA-C40. A putative CaM-binding domain was detected and both CaM-agarose binding and CaM immunoprecipitation verified that cyrA-C45 and cyrA-C40 each bind to CaM in both a Ca(2+)-dependent and -independent manner. cyrA-C45 was present continuously throughout growth and development but was secreted at high levels during the multicellular slug stage of Dictyostelium development. At this time, cyrA localizes to the extracellular matrix (ECM). ECM purification verified the presence of cyrA-C45. An 18 amino acid peptide (DdEGFL1) from the first EGFL repeat sequence of cyrA (EGFL1) that is present in both cyrA-C45 and -C40 enhances both random cell motility and cAMP-mediated chemotaxis. Here we reveal that the dose-dependent enhancement of motility by DdEGFL1 is related to the time of cell starvation. Addition of DdEGFL1 also inhibits cyrA proteolysis. The status of cyrA as an extracellular CaMBP was further clarified by the demonstration that CaM is secreted during development. Antagonism of CaM with W7 resulted in enhanced cyrA proteolysis suggesting a functional role for extracellular CaM in protecting CaMBPs from proteolysis. cyrA is the first extracellular CaMBP identified in Dictyostelium and since it is an ECM protein with EGF-like repeats that enhance cell motility and it likely also represents the first matricellular protein identified in a lower eukaryote.
Collapse
Affiliation(s)
- Andres Suarez
- Department of Biology, University of Toronto at Mississauga, Mississauga, Ontario, Canada
| | | | | | | |
Collapse
|
116
|
Components of cell-matrix linkage as potential new markers for prostate cancer. Cancers (Basel) 2011; 3:883-96. [PMID: 24212644 PMCID: PMC3756394 DOI: 10.3390/cancers3010883] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 02/12/2011] [Accepted: 02/17/2011] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is one of the most common tumor diseases worldwide. Often being non-aggressive, prostate tumors in these cases do not need immediate treatment. However, about 20% of diagnosed prostate cancers tend to metastasize and require treatment. Existing diagnostic methods may fail to accurately recognize the transition of a dormant, non-aggressive tumor into highly malignant prostate cancer. Therefore, new diagnostic tools are needed to improve diagnosis and therapy of prostate carcinoma. This review evaluates existing methods to diagnose prostate carcinoma, such as the biochemical marker prostate-specific antigen (PSA), but also discusses the possibility to use the altered expression of integrins and laminin-332 in prostate carcinomas as diagnostic tools and therapeutic targets of prostate cancer.
Collapse
|
117
|
Tripathi M, Potdar AA, Yamashita H, Weidow B, Cummings PT, Kirchhofer D, Quaranta V. Laminin-332 cleavage by matriptase alters motility parameters of prostate cancer cells. Prostate 2011; 71:184-96. [PMID: 20672321 PMCID: PMC3669684 DOI: 10.1002/pros.21233] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Matriptase, a type II transmembrane serine protease, has been linked to initiation and promotion of epidermal carcinogenesis in a murine model, suggesting that deregulation of its role in epithelia contributes to transformation. In human prostate cancer, matriptase expression correlates with progression. It is therefore of interest to determine how matriptase may contribute to epithelial neoplastic progression. One approach for studying this is to identify potential matriptase substrates involved in epithelial integrity and/or transformation like the extracellular matrix macromolecule, laminin-332 (Ln-332), which is found in the basement membrane of many epithelia, including prostate. Proteolytic processing of Ln-332 regulates cell motility of both normal and transformed cells, which has implications in cancer progression. METHODS In vitro cleavage experiments were performed with purified Ln-332 protein and matriptase. Western blotting, enzyme inhibition assays, and mass spectrometry were used to confirm cleavage events. Matriptase overexpressing LNCaP prostate cancer cells were generated and included in Transwell migration assays and single cell motility assays, along with other prostate cells. RESULTS We report that matriptase proteolytically cleaves Ln-332 in the β3 chain. Substrate specificity was confirmed by blocking cleavage with the matriptase inhibitor, Kunitz domain-1. Transwell migration assays showed that DU145 cell motility was significantly enhanced when plated on matriptase-cleaved Ln-332. Similarly, Transwell migration of matriptase-overexpressing LNCaP cells was significantly increased on Ln-332 and, as determined by live single-cell microscopy, two motility parameters of this cell line, speed and directional persistence, were also higher. CONCLUSIONS Proteolytic processing of Ln-332 by matriptase enhances speed and directional persistence of prostate cancer cells.
Collapse
Affiliation(s)
- Manisha Tripathi
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alka A. Potdar
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee
| | - Hironobu Yamashita
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brandy Weidow
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Peter T. Cummings
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Daniel Kirchhofer
- Department of Protein Engineering, Genentech, Inc., South San Francisco, California
| | - Vito Quaranta
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Correspondence to: Vito Quaranta, MD, Department of Cancer Biology, Vanderbilt University School of Medicine, 771 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232-6840.,
| |
Collapse
|
118
|
Yurchenco PD. Basement membranes: cell scaffoldings and signaling platforms. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a004911. [PMID: 21421915 DOI: 10.1101/cshperspect.a004911] [Citation(s) in RCA: 625] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Basement membranes are widely distributed extracellular matrices that coat the basal aspect of epithelial and endothelial cells and surround muscle, fat, and Schwann cells. These extracellular matrices, first expressed in early embryogenesis, are self-assembled on competent cell surfaces through binding interactions among laminins, type IV collagens, nidogens, and proteoglycans. They form stabilizing extensions of the plasma membrane that provide cell adhesion and that act as solid-phase agonists. Basement membranes play a role in tissue and organ morphogenesis and help maintain function in the adult. Mutations adversely affecting expression of the different structural components are associated with developmental arrest at different stages as well as postnatal diseases of muscle, nerve, brain, eye, skin, vasculature, and kidney.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA.
| |
Collapse
|
119
|
Ulbrich SE, Meyer SU, Zitta K, Hiendleder S, Sinowatz F, Bauersachs S, Büttner M, Fröhlich T, Arnold GJ, Reichenbach HD, Wolf E, Meyer HHD. Bovine endometrial metallopeptidases MMP14 and MMP2 and the metallopeptidase inhibitor TIMP2 participate in maternal preparation of pregnancy. Mol Cell Endocrinol 2011; 332:48-57. [PMID: 20887771 DOI: 10.1016/j.mce.2010.09.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 09/18/2010] [Accepted: 09/21/2010] [Indexed: 01/08/2023]
Abstract
Early embryonic development is critically dependent on both maternal preparation and embryonic signalling of pregnancy. Matrix metallopeptidases (MMP) contribute to spatial and temporal matrix remodeling in the bovine endometrium. In this study we observed distinct changes in expression of MMP2, MMP14, and the metallopeptidase inhibitor TIMP2 between different phases of the estrous cycle indicating an endocrine regulation. An increase of TIMP2 protein abundance was ascertained in the uterine lumen during the time of embryo elongation. The expression pattern and cellular localization correlate well with the assumed effects of MMPs on release and activation of cytokines and growth factors directing cell migration, differentiation, and vascularization during this pivotal period of development. Specifically, active MMP2 in the endometrium may determine the allocation of growth factors supporting conceptus development. The presence of a day 18 conceptus in vivo and day 8 blastoysts in vitro induced endometrial TIMP2 mRNA expression. The results imply that TIMP2 is involved in very early local maternal recognition of pregnancy. Matrix metallopeptidases are likely to participate in remodeling processes preparing a receptive endometrium for a timely and precise regulation of embryo development.
Collapse
Affiliation(s)
- Susanne E Ulbrich
- Physiology Weihenstephan, Technical University Munich, Weihenstephaner Berg 3, 85354 Freising, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Wells A, Chao YL, Grahovac J, Wu Q, Lauffenburger DA. Epithelial and mesenchymal phenotypic switchings modulate cell motility in metastasis. Front Biosci (Landmark Ed) 2011; 16:815-37. [PMID: 21196205 DOI: 10.2741/3722] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The most ominous stage of cancer progression is metastasis, or the dissemination of carcinoma cells from the primary site into distant organs. Metastases are often resistant to current extirpative therapies and even the newest biological agents cure only a small subset of patients. Therefore a greater understanding of tumor biology that integrates properties intrinsic to carcinomas with tissue environmental modulators of behavior is needed. In no aspect of tumor progression is this more evident than the acquisition of cell motility that is critical for both escape from the primary tumor and colonization. In this overview, we discuss how this behavior is modified by carcinoma cell phenotypic plasticity that is evidenced by reversible switching between epithelial and mesenchymal phenotypes. The presence or absence of intercellular adhesions mediate these switches and dictate the receptivity towards signals from the extracellular milieu. These signals, which include soluble growth factors, cytokines, and extracellular matrix embedded with matrikines and matricryptines will be discussed in depth. Finally, we will describe a new mode of discerning the balance between epithelioid and mesenchymal movement.
Collapse
Affiliation(s)
- Alan Wells
- Department of Pathology, Pittsburgh VAMC and University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | |
Collapse
|
121
|
Tsubota Y, Ogawa T, Oyanagi J, Nagashima Y, Miyazaki K. Expression of laminin gamma2 chain monomer enhances invasive growth of human carcinoma cells in vivo. Int J Cancer 2010; 127:2031-41. [PMID: 20143393 DOI: 10.1002/ijc.25231] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Laminin gamma2 chain is a subunit of the heterotrimeric basement membrane protein laminin-332 (alpha3beta3gamma2). The gamma2 chain is highly expressed by human cancers at the invasion fronts and this expression correlates with poor prognosis of the cancers. Our previous study showed that the gamma2 chain is expressed as a monomer form in invading carcinoma cells. However, the role of the gamma2 protein in tumor invasion remains unknown. Here, we demonstrate that the monomeric gamma2 chain promotes invasive growth of human cancer cells in vivo. First, we analyzed regulatory factors for the gamma2 chain expression using 2 gastric carcinoma cell lines. It was found that tumor necrosis factor-alpha, by itself or in a combination with transforming growth factor-beta1, strongly induced the secretion of the monomeric gamma2 chain. In addition, epidermal growth factor families appeared to function as the gamma2 chain inducers in human cancers. Next, we established T-24 bladder carcinoma cell lines expressing the full-length or the short arm of the laminin gamma2 chain. When these cell lines were i.p. injected into nude mice, they produced larger tumors in the abdominal cavity and showed much stronger invasive growth onto the diaphragms than the control cell line. The gamma2-expressing T-24 cells often produced ascites fluid, but scarcely the control cells. In culture, the gamma2-expressing cells migrated through Matrigel more efficiently than the control cells. These findings imply that the gamma2 monomer is induced in human cancers by inflammatory and stromal cytokines and promotes their invasive growth in vivo.
Collapse
Affiliation(s)
- Yoshiaki Tsubota
- Division of Cell Biology, Kihara Institute for Biological Research, Yokohama City University, Totsuka-ku, Yokohama, Japan
| | | | | | | | | |
Collapse
|
122
|
Maller O, Martinson H, Schedin P. Extracellular matrix composition reveals complex and dynamic stromal-epithelial interactions in the mammary gland. J Mammary Gland Biol Neoplasia 2010; 15:301-18. [PMID: 20811805 DOI: 10.1007/s10911-010-9189-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 08/16/2010] [Indexed: 12/25/2022] Open
Abstract
The mammary gland is an excellent model system to study the interplay between stroma and epithelial cells because of the gland's unique postnatal development and its distinct functional states. This review focuses on the contribution of the extracellular matrix (ECM) to stromal-epithelial interactions in the mammary gland. We describe how ECM physical properties, protein composition, and proteolytic state impact mammary gland architecture as well as provide instructive cues that influence the function of mammary epithelial cells during pubertal gland development and throughout adulthood. Further, based on recent proteomic analyses of mammary ECM, we describe known mammary ECM proteins and their potential functions, as well as describe several ECM proteins not previously recognized in this organ. ECM proteins are discussed in the context of the morphologically-distinct stromal subcompartments: the basal lamina, the intra- and interlobular stroma, and the fibrous connective tissue. Future studies aimed at in-depth qualitative and quantitative characterization of mammary ECM within these various subcompartments is required to better elucidate the function of ECM in normal as well as in pathological breast tissue.
Collapse
Affiliation(s)
- Ori Maller
- Department of Medicine, Division of Medical Oncology, University of Colorado-Denver, 12801 E 17th Ave., Aurora, CO 80045, USA
| | | | | |
Collapse
|
123
|
Lysophosphatidic Acid Upregulates Laminin-332 Expression during A431 Cell Colony Dispersal. JOURNAL OF ONCOLOGY 2010; 2010. [PMID: 20862207 PMCID: PMC2938436 DOI: 10.1155/2010/107075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/11/2010] [Accepted: 07/02/2010] [Indexed: 01/01/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that affects various biological functions, such as cell proliferation, migration, survival, wound healing, and tumor invasion through LPA receptors. Previously, we reported that LPA induces A431 colony dispersal, accompanied by disruption of cell-cell contacts and cell migration. However, it remains unclear how LPA affects cell migration and gene expression during A431 colony dispersal. In this paper, we performed cDNA microarray analysis to investigate this question by comparing gene expression between untreated and LPA-treated A431 cells. Interestingly, these results revealed that LPA treatment upregulates several TGF-β1 target genes, including laminin-332 (Ln-332) components (α3, β3, and γ2 chains). Western blot analysis also showed that LPA increased phosphorylation of Smad2, an event that is carried out by TGF-β1 interactions. Among the genes upregulated, we further addressed the role of Ln-332. Real-time PCR analysis confirmed the transcriptional upregulation of all α3, β3, and γ2 chains of Ln-332 by LPA, corresponding to the protein level increases revealed by western blot. Further, the addition of anti-Ln-332 antibody prevented LPA-treated A431 colonies from dispersing. Taken together, our results suggest that LPA-induced Ln-332 plays a significant role in migration of individual cells from A431 colonies.
Collapse
|
124
|
Aokage K, Ishii G, Ohtaki Y, Yamaguchi Y, Hishida T, Yoshida J, Nishimura M, Nagai K, Ochiai A. Dynamic molecular changes associated with epithelial-mesenchymal transition and subsequent mesenchymal-epithelial transition in the early phase of metastatic tumor formation. Int J Cancer 2010; 128:1585-95. [DOI: 10.1002/ijc.25500] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 05/26/2010] [Indexed: 01/26/2023]
|
125
|
Zboralski D, Warscheid B, Klein-Scory S, Malas MB, Becker H, Böckmann M, Meyer HE, Schmiegel W, Simon-Assmann P, Schwarte-Waldhoff I. Uncoupled responses of Smad4-deficient cancer cells to TNFalpha result in secretion of monomeric laminin-gamma2. Mol Cancer 2010; 9:65. [PMID: 20307265 PMCID: PMC2853515 DOI: 10.1186/1476-4598-9-65] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 03/22/2010] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Functional loss of the tumor suppressor Smad4 is involved in pancreatic and colorectal carcinogenesis and has been associated with the acquisition of invasiveness. We have previously demonstrated that the heterotrimeric basement membrane protein laminin-332 is a Smad4 target. Namely, Smad4 functions as a positive transcriptional regulator of all three genes encoding laminin-332; its loss is thus implicated in the reduced or discontinuous deposition of the heterotrimeric basement membrane molecule as evident in carcinomas. Uncoupled expression of laminin genes, on the other hand, namely overexpression of the laminin-gamma2 chain is an impressive marker at invasive edges of carcinomas where tumor cells are maximally exposed to signals from stromal cell types like macrophages. As Smad4 is characterized as an integrator of multiple extracellular stimuli in a strongly contextual manner, we asked if loss of Smad4 may also be involved in uncoupled expression of laminin genes in response to altered environmental stimuli. Here, we address Smad4 dependent effects of the prominent inflammatory cytokine TNFalpha on tumor cells. RESULTS Smad4-reconstituted colon carcinoma cells like adenoma cells respond to TNFalpha with an increased expression of all three chains encoding laminin-332; coincubation with TGFbeta and TNFalpha leads to synergistic induction and to the secretion of large amounts of the heterotrimer. In contrast, in Smad4-deficient cells TNFalpha can induce expression of the gamma2 and beta3 but not the alpha3 chain. Surprisingly, this uncoupled induction of laminin-332 chains in Smad4-negative cells rather than causing intracellular accumulation is followed by the release of gamma2 into the medium, either in a monomeric form or in complexes with as yet unknown proteins. Soluble gamma2 is associated with increased cell migration. CONCLUSIONS Loss of Smad4 may lead to uncoupled induction of laminin-gamma2 in response to TNFalpha and may therefore represent one of the mechanisms which underlie accumulation of laminin-gamma2 at the invasive margin of a tumor. The finding, that gamma2 is secreted from tumor cells in significant amounts and is associated with increased cell migration may pave the way for further investigation to better understand its functional relevance for tumor progression.
Collapse
Affiliation(s)
- Dirk Zboralski
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
| | - Bettina Warscheid
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Susanne Klein-Scory
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
| | - M Bassel Malas
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
| | - Heiko Becker
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
| | - Miriam Böckmann
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Helmut E Meyer
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Wolff Schmiegel
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
- Abtlg. Gastroenterologie und Hepatologie, Kliniken Bergmannsheil, Ruhr-Universität Bochum, Bochum, Germany
| | - Patricia Simon-Assmann
- Inserm, U682, Strasbourg, F-67200 France; Univ Strasbourg, UMR-S682, Strasbourg, F-67081 France
| | - Irmgard Schwarte-Waldhoff
- Medizinische Universitätsklinik, Knappschaftskrankenhaus, IMBL, Ruhr-Universität Bochum, Bochum, Germany
| |
Collapse
|
126
|
Hamill KJ, Paller AS, Jones JCR. Adhesion and migration, the diverse functions of the laminin alpha3 subunit. Dermatol Clin 2010; 28:79-87. [PMID: 19945619 DOI: 10.1016/j.det.2009.10.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The laminins are a secreted family of heterotrimeric molecules essential for basement membrane formation, structure, and function. It is now well established that the alpha3 subunit of laminins-332, -321, and -311 plays an important role in mediating epidermal-dermal integrity and is essential for the skin to withstand mechanical stresses. These laminins also regulate cell migration and mechanosignal transduction. This article provides an overview of the gene, transcripts, and protein structures of laminin alpha3. Also discussed are the proposed functions for the alpha3 subunit-containing laminins.
Collapse
Affiliation(s)
- Kevin J Hamill
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Tarry 8-746, Chicago, IL 60611, USA.
| | | | | |
Collapse
|
127
|
O'Brien J, Lyons T, Monks J, Lucia MS, Wilson RS, Hines L, Man YG, Borges V, Schedin P. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1241-55. [PMID: 20110414 DOI: 10.2353/ajpath.2010.090735] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recent pregnancy correlates with decreased survival for breast cancer patients compared with non-pregnancy-associated breast cancer. We hypothesize that postpartum mammary involution induces metastasis through wound-healing programs known to promote cancer. It is unknown whether alternatively activated M2 macrophages, immune cells important in wound-healing and experimental tumorigenesis that also predict poor prognosis for breast cancer patients, are recruited to the normal involuting gland. Macrophage markers CD68, CSF-1R, and F4/80 were examined across the pregnancy and involution cycle in rodent and human mammary tissues. Quantitative immunohistochemistry revealed up to an eightfold increase in macrophage number during involution, which returned to nulliparous levels with full regression. The involution macrophages exhibit an M2 phenotype as determined by high arginase-1 and low inducible nitric oxide synthase staining in rodent tissue, and by mannose receptor expression in human breast tissue. M2 cytokines IL-4 and IL-13 also peaked during involution. Extracellular matrix (ECM) isolated from involuting rat mammary glands was chemotactic for macrophages compared with nulliparous mammary ECM. Fibrillar collagen levels and proteolysis increased dramatically during involution, and denatured collagen I acted as a strong chemoattractant for macrophages in cell culture, suggesting proteolyzed fibrillar collagen as a candidate ECM mediator of macrophage recruitment. M2 macrophages, IL-4, IL-13, fibrillar collagen accumulation, and proteolysis of collagen are all components of tumor promotional microenvironments, and thus may mediate promotion of breast cancers arising in the postpartum setting.
Collapse
Affiliation(s)
- Jenean O'Brien
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, Mail Stop 8117, RC-1 S, 8401K, 12801 E. 17 Avenue, UCD, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Lugassy C, Torres-Muñoz JE, Kleinman HK, Ghanem G, Vernon S, Barnhill RL. Overexpression of malignancy-associated laminins and laminin receptors by angiotropic human melanoma cells in a chick chorioallantoic membrane model. J Cutan Pathol 2009; 36:1237-43. [DOI: 10.1111/j.1600-0560.2009.01273.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
129
|
Abstract
The extracellular matrix (ECM) and ECM proteins are important in phenomena as diverse as developmental patterning, stem cell niches, cancer, and genetic diseases. The ECM has many effects beyond providing structural support. ECM proteins typically include multiple, independently folded domains whose sequences and arrangement are highly conserved. Some of these domains bind adhesion receptors such as integrins that mediate cell-matrix adhesion and also transduce signals into cells. However, ECM proteins also bind soluble growth factors and regulate their distribution, activation, and presentation to cells. As organized, solid-phase ligands, ECM proteins can integrate complex, multivalent signals to cells in a spatially patterned and regulated fashion. These properties need to be incorporated into considerations of the functions of the ECM.
Collapse
Affiliation(s)
- Richard O Hynes
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
130
|
Affiliation(s)
- Richard O. Hynes
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
131
|
Guess CM, Quaranta V. Defining the role of laminin-332 in carcinoma. Matrix Biol 2009; 28:445-55. [PMID: 19686849 PMCID: PMC2875997 DOI: 10.1016/j.matbio.2009.07.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 07/27/2009] [Accepted: 07/28/2009] [Indexed: 01/10/2023]
Abstract
The deadly feature of cancer, metastasis, requires invasion of cells through basement membranes (BM), which normally act as barriers between tissue compartments. In the case of many epithelially-derived cancers (carcinomas), laminin-332 (Ln-332) is a key component of the BM barrier. This review provides a historical examination of Ln-332 from its discovery through identification of its functions in BM and possible role in carcinomas. Current understanding points to distinct roles for the three Ln-332 subunits (alpha3, beta3, gamma2) in cell adhesion, extracellular matrix stability, and cell signaling processes in cancer. Given the large number of studies linking Ln-332 gamma2 subunit with cancer prognosis, particular attention is given to the crucial role of this subunit in cancer invasion and to the unanswered questions in this area.
Collapse
Affiliation(s)
- Cherise M Guess
- Meharry Medical College, Department of Microbial Pathogenesis & Immune Response; Nashville, TN 37232-6840, USA.
| | | |
Collapse
|
132
|
Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound Repair Regen 2009; 17:153-62. [PMID: 19320882 DOI: 10.1111/j.1524-475x.2009.00466.x] [Citation(s) in RCA: 713] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dynamic interactions between growth factors and extracellular matrix (ECM) are integral to wound healing. These interactions take several forms that may be categorized as direct or indirect. The ECM can directly bind to and release certain growth factors (e.g., heparan sulfate binding to fibroblast growth factor-2), which may serve to sequester and protect growth factors from degradation, and/or enhance their activity. Indirect interactions include binding of cells to ECM via integrins, which enables cells to respond to growth factors (e.g., integrin binding is necessary for vascular endothelial growth factor-induced angiogenesis) and can induce growth factor expression (adherence of monocytes to ECM stimulates synthesis of platelet-derived growth factor). Additionally, matrikines, or subcomponents of ECM molecules, can bind to cell surface receptors in the cytokine, chemokine, or growth factor families and stimulate cellular activities (e.g., tenascin-C and laminin bind to epidermal growth factor receptors, which enhances fibroblast migration). Growth factors such as transforming growth factor-beta also regulate the ECM by increasing the production of ECM components or enhancing synthesis of matrix degrading enzymes. Thus, the interactions between growth factors and ECM are bidirectional. This review explores these interactions, discusses how they are altered in difficult to heal or chronic wounds, and briefly considers treatment implications.
Collapse
Affiliation(s)
- Gregory S Schultz
- Department of Obstetrics and Gynecology, University of Florida, Gainesville, 32610-0294, USA.
| | | |
Collapse
|
133
|
Isolation and characterization of a secreted, cell-surface glycoprotein SCUBE2 from humans. Biochem J 2009; 422:119-28. [DOI: 10.1042/bj20090341] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SCUBE2 [signal peptide, CUB domain, EGF (epidermal growth factor)-like protein 2] belongs to an evolutionarily conserved SCUBE protein family, which possesses domain organization characteristic of an N-terminal signal peptide sequence followed by nine EGF-like repeats, a spacer region, three cysteine-rich repeat motifs, and one CUB domain at the C-terminus. Despite several genetic analyses suggesting that the zebrafish orthologue of the mammalian SCUBE2 gene participates in HH (Hedgehog) signalling, the complete full-length cDNA and biochemical function for mammalian SCUBE2 on HH signalling remains uninvestigated. In the present study, we isolated the full-length cDNA and studied the role of human SCUBE2 in the HH signalling cascade. When overexpressed, recombinant human SCUBE2 manifests as a secreted surface-anchored glycoprotein. Deletion mapping analysis defines the critical role of the spacer region and/or cysteine-rich repeats for membrane association. Further biochemical analyses and functional reporter assays demonstrated that human SCUBE2 can specifically interact with SHH (Sonic Hedgehog) and SHH receptor PTCH1 (Patched-1), and enhance the SHH signalling activity within the cholesterol-rich raft microdomains of the plasma membranes. Together, our results reveal that human SCUBE2 is a novel positive component of the HH signal, acting upstream of ligand binding at the plasma membrane. Thus human SCUBE2 could play important roles in HH-related biology and pathology, such as during organ development and tumour progression.
Collapse
|
134
|
Lozito TP, Kuo CK, Taboas JM, Tuan RS. Human mesenchymal stem cells express vascular cell phenotypes upon interaction with endothelial cell matrix. J Cell Biochem 2009; 107:714-22. [PMID: 19415687 PMCID: PMC5543930 DOI: 10.1002/jcb.22167] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Mesenchymal stem cells (MSCs) are thought to occupy a perivascular niche where they are exposed to signals originating from vascular cells. This study focused on the effects of endothelial cell (EC)-derived signals on MSC differentiation toward vascular cell lineages. Upon co-culture with two types of ECs, macrovascular (macro) ECs and microvascular (micro) ECs, the former caused MSCs to increase expression of both EC and smooth muscle cell (SMC) markers, while the latter induced expression of EC markers only. These marker changes in MSCs were linked to the extracellular matrixes secreted by the ECs (EC-matrix) rather than soluble EC-secreted factors. Beyond enhanced marker expression, EC-matrix also induced functional changes in MSCs indicative of development of a genuine vascular cell phenotype. These included enhanced incorporation into vessels and cytoskeletal localization of vascular SMC-specific contractile elements. The bioactivity of EC-matrix was sensitive to EDTA washes and required sulfated glycosaminoglycans. However, neither soluble VEGF nor substrate surfaces coated with fibronectin, collagen type IV, or laminin recreated the effects of EC-matrix on MSC vascular differentiation. In conclusion, these results identified EC-matrix as a critical regulator of vascular cell differentiation of MSCs. Elucidating these MSC-EC-matrix interactions and identifying the specific EC-matrix components involved will shed light on the perivascular signals seen by MSCs in vivo.
Collapse
Affiliation(s)
- Thomas P. Lozito
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| | - Catherine K. Kuo
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| | - Juan M. Taboas
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| |
Collapse
|
135
|
Zhang Z, Song T, Jin Y, Pan J, Zhang L, Wang L, Li P. Epidermal Growth Factor Receptor Regulates MT1-MMP and MMP-2 Synthesis in SiHa Cells via Both PI3-K/AKT and MAPK/ERK Pathways. Int J Gynecol Cancer 2009; 19:998-1003. [DOI: 10.1111/igc.0b013e3181a83749] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Matrix metalloproteinase 2 (MMP-2) and membrane type 1 matrix metalloproteinase (MT1-MMP) have been identified as important participants in tumor invasion, metastasis, and angiogenesis. Membrane type 1 matrix metalloproteinase has also been recognized as a major activator of MMP-2. The purpose of this study was to investigate epidermal growth factor (EGF) mediating signal pathways in the regulation of MMP-2 and MT1-MMP in SiHa cells, a cervical cancer cell line. We showed here that EGF induced the expression of MT1-MMP and inhibited the expression of MMP-2 at both the mRNA and protein levels. Membrane type 1 matrix metalloproteinase induction was blocked by mitogen-activated protein kinase or extracellular signal-regulated kinase inhibitors PD98059 and U0126 but not by phosphatidylinositol-3 kinase (PI3-K) inhibitors LY294002 and wortmannin. Interestingly, the mitogen-activated protein kinase or extracellular signal-regulated kinase inhibitors PD98059 and U0126 actually increased MMP-2 mRNA and protein synthesis, whereas the PI3-K inhibitors LY294002 and wortmannin further suppressed the expression of MMP-2. Our results suggest that EGF receptor up-regulated the expression of MT1-MMP and down-regulated the synthesis of MMP-2 through the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway while concomitantly transmitting a mild positive regulatory signal to the expression of MMP-2 via the PI3-K/AKT pathway in SiHa cells. Furthermore, we found that EGF elevated the activity of MMP-2 in culture media.
Collapse
|
136
|
Yamamoto H, Kitadai Y, Yamamoto H, Oue N, Ohdan H, Yasui W, Kikuchi A. Laminin gamma2 mediates Wnt5a-induced invasion of gastric cancer cells. Gastroenterology 2009; 137:242-52, 252.e1-6. [PMID: 19582886 DOI: 10.1053/j.gastro.2009.02.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND & AIMS Wnt5a expression stimulates in vitro migration and invasion of cultured gastric cancer cells by an unknown mechanism and is also correlated with aggressiveness of gastric tumors. The aim of this study was to show that Wnt5a is involved in metastasis of gastric cancer cells in vivo and to explore the molecular mechanism by which Wnt5a regulates migration and invasion. METHODS In an experimental liver metastasis assay, Wnt5a-knockdown gastric cancer cells were injected into the spleens of nude mice. Microarray analyses were used to compare expression patterns between mouse fibroblast L cells that stably express wild-type and a mutant form of Wnt5a to investigate Wnt5a-dependent gene expression. The expression of genes found to be regulated by Wnt5a was investigated in cultured gastric cancer cells. Immunohistochemical analyses were performed to measure levels of Wnt-regulated gene products in 153 gastric cancer samples. RESULTS Knockdown of Wnt5a in gastric cancer cells reduced the number of liver metastases that formed in nude mice. Microarray analyses indicated that Wnt5a activity induced expression of the gene encoding laminin gamma2, a subunit of the epithelial basement membrane protein laminin-5. Wnt5a induced the expression of laminin gamma2 through the activation of protein kinase C and c-Jun-N-terminal kinase. The invasive activity of gastric cancer cells depended on laminin gamma2; Wnt5a expression levels correlated with those of laminin gamma2 in diffuse-scattered type gastric tumor samples from patients. CONCLUSIONS Wnt5a contributes to gastric cancer progression by increasing metastatic potential. Wnt5a up-regulates laminin gamma2 to mediate gastric cancer cell aggressiveness.
Collapse
Affiliation(s)
- Hideki Yamamoto
- Department of Biochemistry, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | |
Collapse
|
137
|
Guess CM, LaFleur BJ, Weidow BL, Quaranta V. A decreased ratio of laminin-332 beta3 to gamma2 subunit mRNA is associated with poor prognosis in colon cancer. Cancer Epidemiol Biomarkers Prev 2009; 18:1584-90. [PMID: 19383890 PMCID: PMC2869450 DOI: 10.1158/1055-9965.epi-08-1027] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Laminin-332 (Ln-332) is a heterotrimeric glycoprotein (alpha3beta3gamma2) unique to epithelial cells with crucial roles in signaling, adhesion, and migration. Altered localization or expression levels of Ln-332, particularly its gamma2 subunit, are of prognostic value in a variety of cancers. However, the lack of standardized methodology and the limited quantification of previous study results have left unanswered questions, including the role of gamma2 transcript variants and whether differential expression of this chain represents dysregulation of the whole heterotrimer. Herein, we test the hypothesis that mRNA changes in one or more Ln-332 encoding genes can be used to distinguish between early- and advanced-stage cancer specimens and shed light on mechanistic questions raised by previous studies. Statistical analyses of human microarray data from the publicly available expression project in Oncology (expO) dataset, including examination of the distributions of Ln-332 subunit mRNA levels, identified a significant decrease in the Ln-332 beta3:gamma2 mRNA ratio between normal (n = 10) and early-stage colon cancer (n = 29) specimens. The beta3:gamma2 ratio was further decreased in metastatic colon cancer (n = 41) compared with early-stage samples. Our findings raise the possibility that Ln-332 gamma2 may be a therapeutic target against metastatic colon cancer because a lowered beta3:gamma2 ratio would reduce expression of heterotrimeric Ln-332 and increase monomeric gamma2 secretion. Further, standardized, quantitative methods for patient prognosis and therapeutic choice could be developed based upon the Ln-332 mRNA changes we uncovered.
Collapse
Affiliation(s)
- Cherise M. Guess
- Department of Microbial Pathogenesis and Immune Response, Meharry Medical College, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bonnie J. LaFleur
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brandy L. Weidow
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Integrative Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vito Quaranta
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Integrative Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
138
|
Chang YC, Sabourin CLK, Lu SE, Sasaki T, Svoboda KKH, Gordon MK, Riley DJ, Casillas RP, Gerecke DR. Upregulation of gamma-2 laminin-332 in the mouse ear vesicant wound model. J Biochem Mol Toxicol 2009; 23:172-84. [PMID: 19526566 PMCID: PMC4465420 DOI: 10.1002/jbt.20275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epithelial cell migration during wound healing is regulated in part by enzymatic processing of laminin-332 (formerly LN-5), a heterodimer formed from alpha, beta, and gamma polypeptide chains. Under static conditions, laminin-332 is secreted into the extracellular matrix as a proform and has two chains processed to smaller forms, allowing it to anchor epithelial cells to the basement membrane of the dermis. During incisional wounding, laminin gamma2 chains in particular are processed to smaller sizes and function to promote epithelial sheet migration over the wound bed. The present study examines whether this same function occurs following chemical injury. The mouse ear vesicant model (MEVM) was used to follow the pathology in the ear and test whether processed laminin-332 enhances epithelial cell migration. Skin biopsies of sulfur mustard (SM) exposed ears for several time points were analyzed by histology, immunohistochemistry, real-time PCR, and Western blot analysis. SM exposure greatly increased mRNA levels for laminin-gamma2 in comparison to the other two chains. Protein production of laminin-gamma2 was upregulated, and there was an increase in the processed forms. Protein production was in excess of the amount required to form heterotrimeric laminin-332 and was associated with the migrating epithelial sheet, suggesting a potential role in wound healing for monomeric laminin-gamma2.
Collapse
Affiliation(s)
- Yoke-Chen Chang
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, EOHSI, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | - Shou-En Lu
- Department of Biostatistics, UMDNJ School of Public Health, 683 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Takako Sasaki
- Department of Molecular Biology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kathy K. H. Svoboda
- Biomedical Sciences, Texas A&M Health Science Center, Baylor College of Dentistry, Dallas, TX 75246, USA
| | - Marion K. Gordon
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, EOHSI, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - David J. Riley
- Department of Medicine, UMDNJ–Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Robert P. Casillas
- Battelle Memorial Institute, 505 King Avenue, Columbus, OH 43201-2693, USA
| | - Donald R. Gerecke
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, EOHSI, 170 Frelinghuysen Road, Piscataway, NJ 08854, USA
| |
Collapse
|
139
|
Méndez E, Houck JR, Doody DR, Fan W, Lohavanichbutr P, Rue TC, Yueh B, Futran ND, Upton MP, Farwell DG, Heagerty PJ, Zhao LP, Schwartz SM, Chen C. A genetic expression profile associated with oral cancer identifies a group of patients at high risk of poor survival. Clin Cancer Res 2009; 15:1353-61. [PMID: 19228736 DOI: 10.1158/1078-0432.ccr-08-1816] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE To determine if gene expression signature of invasive oral squamous cell carcinoma (OSCC) can subclassify OSCC based on survival. EXPERIMENTAL DESIGN We analyzed the expression of 131 genes in 119 OSCC, 35 normal, and 17 dysplastic mucosa to identify cluster-defined subgroups. Multivariate Cox regression was used to estimate the association between gene expression and survival. By stepwise Cox regression, the top predictive models of OSCC-specific survival were determined and compared by receiver operating characteristic analysis. RESULTS The 3-year overall mean+/-SE survival for a cluster of 45 OSCC patients was 38.7+/-0.09% compared with 69.1+/-0.08% for the remaining patients. Multivariate analysis adjusted for age, sex, and stage showed that the 45 OSCC patient cluster had worse overall and OSCC-specific survival (hazard ratio, 3.31; 95% confidence interval, 1.66-6.58 and hazard ratio, 5.43; 95% confidence interval, 2.32-12.73, respectively). Stepwise Cox regression on the 131 probe sets revealed that a model with a term for LAMC2 (laminin gamma2) gene expression best identified patients with worst OSCC-specific survival. We fit a Cox model with a term for a principal component analysis-derived risk score marker and two other models that combined stage with either LAMC2 or PCA. The area under the curve for models combining stage with either LAMC2 or PCA was 0.80 or 0.82, respectively, compared with 0.70 for stage alone (P=0.013 and 0.008, respectively). CONCLUSIONS Gene expression and stage combined predict survival of OSCC patients better than stage alone.
Collapse
Affiliation(s)
- Eduardo Méndez
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, WA 98109-1024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Tamoxifen induces pleiotrophic changes in mammary stroma resulting in extracellular matrix that suppresses transformed phenotypes. Breast Cancer Res 2009; 11:R5. [PMID: 19173736 PMCID: PMC2687708 DOI: 10.1186/bcr2220] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 12/16/2008] [Accepted: 01/27/2009] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION The functional unit of the mammary gland has been defined as the epithelial cell plus its microenvironment, a hypothesis that predicts changes in epithelial cell function will be accompanied by concurrent changes in mammary stroma. To test this hypothesis, the question was addressed of whether mammary stroma is functionally altered by the anti-oestrogen drug tamoxifen. METHODS Forty female rats at 70 days of age were randomised to two groups of 20 and treated with 1.0 mg/kg tamoxifen or vehicle subcutaneously daily for 30 days, followed by a three-day wash out period. Mammary tissue was harvested and effects of tamoxifen on mammary epithelium and stroma determined. RESULTS As expected, tamoxifen suppressed mammary alveolar development and mammary epithelial cell proliferation. Primary mammary fibroblasts isolated from tamoxifen-treated rats displayed a three-fold decrease in motility and incorporated less fibronectin in their substratum in comparison to control fibroblasts; attributes indicative of fibroblast quiescence. Immunohistochemistry analysis of CD68, a macrophage lysosomal marker, demonstrated a reduction in macrophage infiltration in mammary glands of tamoxifen-treated rats. Proteomic analyses by mass spectrometry identified several extracellular matrix (ECM) proteins with expression levels with tamoxifen treatment that were validated by Western blot. Mammary tissue from tamoxifen-treated rats had decreased fibronectin and increased collagen 1 levels. Further, ECM proteolysis was reduced in tamoxifen-treated rats as detected by reductions in fibronectin, laminin 1, laminin 5 and collagen 1 cleavage fragments. Consistent with suppression in ECM proteolysis with tamoxifen treatment, matrix metalloproteinase-2 levels and activity were decreased. Biochemically extracted mammary ECM from tamoxifen-treated rats suppressed in vitro macrophage motility, which was rescued by the addition of proteolysed collagen or fibronectin. Mammary ECM from tamoxifen-treated rats also suppressed breast tumour cell motility, invasion and haptotaxis, reduced organoid size in 3-dimensional culture and blocked tumour promotion in an orthotopic xenograft model; effects which could be partially reversed by the addition of exogenous fibronectin. CONCLUSIONS These data support the hypothesis that mammary stroma responds to tamoxifen treatment in concert with the epithelium and remodels to a microenvironment inhibitory to tumour cell progression. Reduced fibronectin levels and reduced ECM turnover appear to be hallmarks of the quiescent mammary microenvironment. These data may provide insight into attributes of a mammary microenvironment that facilitate tumour dormancy.
Collapse
|
141
|
Involvement of caspases and transmembrane metalloproteases in sulphur mustard-induced microvesication in adult human skin in organ culture: directions for therapy. Toxicology 2009; 258:39-46. [PMID: 19167455 DOI: 10.1016/j.tox.2009.01.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 12/23/2008] [Accepted: 01/05/2009] [Indexed: 11/24/2022]
Abstract
While skin is a major target for sulphur mustard (HD), a therapy to limit HD-induced vesication is currently not available. Since it is supposed that apoptotic cell death and proteolytic digestion of extracellular matrix proteins by metalloproteases are initiating factors for blister formation, we have explored whether inhibition of these processes could prevent HD-induced epidermal-dermal separation using adult human skin in organ culture. Involvement of the caspase and the metalloprotease families was confirmed by the observation that their respective broad spectrum inhibitors, Z-VAD-fmk and GM6001, each suppressed HD-induced microvesication. The lowest effective concentrations were 10 and 100microM, respectively. Using specific inhibitors for caspase-8 (> or =10microM) and caspase-9 (> or =10microM) we learned that HD-induced apoptosis is initiated by the death receptor pathway as well as by the mitochondrial pathway. Remarkably, blocking caspase-8 activity resulted in morphologically better conserved cells than blocking caspase-9 activity. We zoomed in on the role of metalloproteases in HD-induced microvesication by testing the effects of two inhibitors: dec-RVKR-cmk and TAPI-2. Dec-RVKR-cmk is an inhibitor of furin, which activates transmembrane enzymes of the 'a disintegrin and metalloproteinase' (ADAM)-family as well as the membrane-type metalloproteases (MTx-MMP). TAPI-2 specifically inhibits TNFalpha-converting enzyme (TACE/ADAM17), which is involved in pericellular proteolysis. Both inhibitors prevented microvesication at concentrations of > or =500 and > or =20microM, respectively. This confirms that ADAMs and MT-MMPs play a role in HD-induced epidermal-dermal separation, with a particular role for TACE/ADAM17. Since TACE is involved not only in degradation of cell-matrix adhesion structures, but also in ectodomain shedding of ligands for epidermal growth factor receptor (EGFR) and in release of TNFalpha, these results imply TACE-mediated pathways as a new concept in HD toxicity. In conclusion, transmembrane metalloproteases probably form a main target for treatment of blisters in HD casualties. The observation that microvesication in the ex vivo human skin model still could be prevented when the metalloprotease inhibitor GM6001 was applied up to 8h after exposure to HD opens perspectives for non-urgent cure of HD casualties.
Collapse
|
142
|
Liu A, Garg P, Yang S, Gong P, Pallero MA, Annis DS, Liu Y, Passaniti A, Mann D, Mosher DF, Murphy-Ullrich JE, Goldblum SE. Epidermal growth factor-like repeats of thrombospondins activate phospholipase Cgamma and increase epithelial cell migration through indirect epidermal growth factor receptor activation. J Biol Chem 2009; 284:6389-402. [PMID: 19129184 DOI: 10.1074/jbc.m809198200] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Thrombospondin (TSP) 1 is a trimeric multidomain protein that contains motifs that recognize distinct host cell receptors coupled to multiple signaling pathways. Selected TSP1-induced cellular responses are tyrosine kinase-dependent, and TSP1 contains epidermal growth factor (EGF)-like repeats. Specific receptor interactions or functions for the EGF-like repeats have not been identified. We asked whether one or more biological responses to TSP1 might be explained through EGF receptor (EGFR) activation. In A431 cells, TSP1 increased autophosphorylation of Tyr-1068 of EGFR in a dose- and time-dependent manner. The ability of TSP1 to activate EGFR was replicated by the tandem EGF-like repeats as a recombinant protein. The three EGF-like repeats alone produced a high level of Tyr-1068 phosphorylation. EGF-like repeats from TSP2 and TSP4 also activated EGFR. Tyr-1068 phosphorylation was less when individual EGF-like repeats were tested or flanking sequences were added to the three EGF-like repeats. TSP1 and its EGF-like repeats also increased phosphorylation of EGFR Tyr-845, Tyr-992, Tyr-1045, Tyr-1086, and Tyr-1173, activated phospholipase Cgamma, and increased cell migration. No evidence was found for binding of the EGF-like repeats to EGFR. Instead, EGFR activation in response to TSP1 or its EGF-like repeats required matrix metalloprotease activity, including activity of matrix metalloprotease 9. Access to the ligand-binding portion of the EGFR ectodomain was also required. These findings suggest release of an endogenous EGFR ligand in response to ligation of a second unknown receptor by the TSPs.
Collapse
Affiliation(s)
- Anguo Liu
- Mucosal Biology Research Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Liu A, Mosher DF, Murphy-Ullrich JE, Goldblum SE. The counteradhesive proteins, thrombospondin 1 and SPARC/osteonectin, open the tyrosine phosphorylation-responsive paracellular pathway in pulmonary vascular endothelia. Microvasc Res 2009; 77:13-20. [PMID: 18952113 PMCID: PMC3022346 DOI: 10.1016/j.mvr.2008.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 08/28/2008] [Indexed: 11/30/2022]
Abstract
Counteradhesive proteins are a group of genetically and structurally distinct multidomain proteins that have been grouped together for their ability to inhibit cell-substrate interactions. Three counteradhesive proteins that influence endothelial cell behavior include thrombospondin (TSP)1, (SPARC) (Secreted Protein Acidic and Rich in Cysteine), also known as osteonectin, and tenascin. More recently, these proteins have been shown to regulate not only cell-matrix interactions but cell-cell interactions as well. TSP1 increases tyrosine phosphorylation of components of the cell-cell adherens junctions or zonula adherens (ZA) and opens the paracellular pathway in human lung microvascular endothelia. The epidermal growth factor (EGF)-repeats of TSP1 activate the (EGF) receptor (EGFR) and ErbB2, and these two receptor protein tyrosine kinases (PTK)s participate in ZA protein tyrosine phosphorylation and barrier disruption in response to the TSP1 stimulus. For the barrier response to TSP1, EGFR/ErbB2 activation is necessary but insufficient. Protein tyrosine phosphatase (PTP)mu counter-regulates phosphorylation of selected tyrosine residues within the cytoplasmic domain of EGFR. Although tenascin, like TSP1, also contains EGF-like repeats and is known to activate EGFR, whether it also opens the paracellular pathway is unknown. In addition to TSP1, tenascin, and the other TSP family members, there are numerous other proteins that also contain EGF-like repeats and participate in hemostasis, wound healing, and tissue remodeling. EGFR not only responds to direct binding of EGF motif-containing ligands but can also be transactivated by a wide range of diverse stimuli. In fact, several established mediators of increased vascular permeability and/or lung injury, including thrombin, tumor necrosis factor-alpha, platelet-activating factor, bradykinin, angiopoietin, and H(2)O(2), transactivate EGFR. It is conceivable that EGFR serves a pivotal signaling role in a final common pathway for the pulmonary response to selected injurious stimuli. SPARC/Osteonectin also increases tyrosine phosphorylation of ZA proteins and opens the endothelial paracellular pathway in a PTK-dependent manner. The expression of the counteradhesive proteins is increased in response to a wide range of injurious stimuli. It is likely that these same molecules participate in the host response to acute lung injury and are operative during the barrier response within the pulmonary microvasculature.
Collapse
Affiliation(s)
- Anguo Liu
- University of Maryland School of Medicine, Mucosal Biology Research Center, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
144
|
Le Bitoux MA, Stamenkovic I. Tumor-host interactions: the role of inflammation. Histochem Cell Biol 2008; 130:1079-90. [PMID: 18953558 DOI: 10.1007/s00418-008-0527-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2008] [Indexed: 12/20/2022]
Abstract
It is well established that interactions between tumor cells and the host tissue stroma play a key role in determining whether and how any given solid malignancy will develop. In most cases, tumor cells hijack stromal cell functions for their own benefit and ultimately dictate the rules of engagement to the host tissue microenvironment. However, the contribution of the different stromal cell components to tumor growth remains to be clarified. Because most solid tumors are accompanied by a local inflammatory response, it has long been thought that inflammation and carcinogenesis are related. If formal proof that cancer can be initiated by inflammation in the absence of exogenous carcinogens is still lacking, there is abundant evidence that the inflammatory response can play a central role in modulating tumor growth and progression. This review will discuss some of the mechanisms whereby inflammation can both enhance and inhibit tumor growth.
Collapse
Affiliation(s)
- Marie-Aude Le Bitoux
- Division of Experimental Pathology, Institute of Pathology, CHUV, Lausanne, Switzerland
| | | |
Collapse
|
145
|
Abstract
The extracellular matrix (ECM) was long thought to be merely a structural tissue support and/or a filter. However, recent studies have suggested that ECM proteins regulate many intracellular and extracellular events, including cell growth, cell adhesion, cell division, cell movement, and apoptosis. They do so through activation of several families of cell surface receptor, including the integrins and syndecans. The focus of this review is on two laminin isoforms expressed in the skin. Laminins are an important molecular component of the basement membranes in a variety of tissue types. They have a cruciform shape, and are composed of three chains-alpha, beta, and gamma. Keratinocytes of the skin secrete numerous laminin isoforms, including laminin-511 and laminin-332. The latter are known to affect the behaviour of keratinocytes through binding to membrane-penetrating receptors (outside-in signal transduction). Conversely, the expression, secretion and assembly of laminin-rich matrices is regulated by cell surface receptors through inside-out signal transduction. We will review how integrins regulate laminin matrix assembly and the signals elicited by laminins that support either migration or stable adhesion of keratinocytes. We will also discuss recent data indicating that laminins plays key regulatory roles in the development of skin appendages and contribute to the pathogenesis of skin cancer.
Collapse
Affiliation(s)
- Koji Sugawara
- Department of Dermatology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | |
Collapse
|
146
|
Mydel P, Shipley JM, Adair-Kirk TL, Kelley DG, Broekelmann TJ, Mecham RP, Senior RM. Neutrophil elastase cleaves laminin-332 (laminin-5) generating peptides that are chemotactic for neutrophils. J Biol Chem 2008; 283:9513-22. [PMID: 18178964 PMCID: PMC2442305 DOI: 10.1074/jbc.m706239200] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 12/14/2007] [Indexed: 11/06/2022] Open
Abstract
Proteolytic processing of laminin-332 by matrix metalloproteinase (MMP)-2 and MMP-14 has been shown to yield fragments that are promigratory for epithelial cells. During acute and chronic inflammation, proteases are elaborated by neutrophils and macrophages that can degrade basement membranes. We investigated the susceptibility of laminin-332 to degradation by the following neutrophil and macrophage proteases: neutrophil elastase (NE), cathepsin G, proteinase-3, and MMPs-2, -8, -9, and -12. Protease-specific differences were seen in the capacity to cleave the individual chains of laminin-332. NE and MMP-12 showed the greatest activity toward the gamma2 chain, generating a fragment similar in size to the gamma2x fragment generated by MMP-2. The digestion pattern of laminin-332 by degranulated neutrophils was nearly identical to that generated with NE alone. Digestion by supernatants of degranulated neutrophils was blocked by an inhibitor of NE, and NE-deficient neutrophils were essentially unable to digest laminin-332, suggesting that NE is the major neutrophil-derived protease that degrades laminin-332. In vivo, laminin gamma2 fragments were found in the bronchoalveolar lavage fluid of wild-type mice treated with lipopolysaccharide, whereas that obtained from NE-deficient mice showed a different cleavage pattern. In addition, NE cleaved a synthetic peptide derived from the region of human laminin gamma2 containing the MMP-2 cleavage site, suggesting that NE may generate laminin-332 fragments that are also promigratory. Both laminin-332 fragments generated by NE digestion and NE-digested laminin gamma2 peptide were found to be chemotactic for neutrophils. Collectively, these data suggest that degradation of laminin-332 by NE generates fragments with important biological activities.
Collapse
Affiliation(s)
- Piotr Mydel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
147
|
Hojilla CV, Wood GA, Khokha R. Inflammation and breast cancer: metalloproteinases as common effectors of inflammation and extracellular matrix breakdown in breast cancer. Breast Cancer Res 2008; 10:205. [PMID: 18394187 PMCID: PMC2397522 DOI: 10.1186/bcr1980] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Two rapidly evolving fields are converging to impact breast cancer: one has identified novel substrates of metalloproteinases that alter immune cell function, and the other has revealed a role for inflammation in human cancers. Evidence now shows that the mechanisms underlying these two fields interact in the context of breast cancer, providing new opportunities to understand this disease and uncover novel therapeutic strategies. The metalloproteinase class of enzymes is well studied in mammary gland development and physiology, but mostly in the context of extracellular matrix modification. Aberrant metalloproteinase expression has also been implicated in breast cancer progression, where these genes act as tumor modifiers. Here, we review how the metalloproteinase axis impacts mammary physiology and tumorigenesis and is associated with inflammatory cell influx in human breast cancer, and evaluate its potential as a regulator of inflammation in the mammary gland.
Collapse
Affiliation(s)
- Carlo V Hojilla
- Department of Medical Biophysics, Ontario Cancer Institute, Toronto, M5G 2M9 Canada
| | | | | |
Collapse
|
148
|
Baba Y, Iyama KI, Hirashima K, Nagai Y, Yoshida N, Hayashi N, Miyanari N, Baba H. Laminin-332 promotes the invasion of oesophageal squamous cell carcinoma via PI3K activation. Br J Cancer 2008; 98:974-80. [PMID: 18283320 PMCID: PMC2266844 DOI: 10.1038/sj.bjc.6604252] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 01/15/2008] [Accepted: 01/16/2008] [Indexed: 01/16/2023] Open
Abstract
Laminin-332 is major component of epithelial basement membrane, and has an important role in cell migration and tumour invasion. Recently, the phosphatidylinositol 3-kinase (PI3K) activation induced by laminin-332 during carcinogenesis or tumour invasion has been highlighted in skin squamous cell carcinoma. The expression of laminin-332 in 126 resected oesophageal squamous cell carcinoma (ESCC) specimens was immunohistochemically examined to determine its associations with the clinicopathological characteristics, and the effect of laminin-332 on the invasiveness and the PI3K activation was assessed by in vitro experiments using ESCC cell lines (ESCCs). Sections with immunostaining signals in >30% cancer cells, which were observed in 55 of 126 cases, were judged to be positive for laminin-332. The positivity was significantly correlated with pTNM stage and poor prognosis. Inactivation of the PI3K pathway by laminin-332 blocking antibody suppressed the invasiveness of TE8 cell line, which secreted laminin-332 at high level and had high PI3K activity. The addition of the purified laminin-332 activated the PI3K pathway and increased the invasiveness of TE11 cell line, which secreted laminin-332 at lower level and had low PI3K activity. The deactivation of PI3K pathway using the PI3K inhibitor decreased the invasiveness of ESCCs and the secretion of laminin-332 in vitro. The expression of laminin-332 was one of the prognostic factors of ESCC. Laminin-332 could provide the autocrine positive-feedback loop through PI3K activation, contributing the invasive ability. Therefore, the inhibitor of PI3K pathway might be useful as the anticancer therapies for ESCC.
Collapse
Affiliation(s)
- Y Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
- Department of Surgical Pathology, Kumamoto University Hospital, 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - K-i Iyama
- Department of Surgical Pathology, Kumamoto University Hospital, 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - K Hirashima
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - Y Nagai
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - N Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - N Hayashi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - N Miyanari
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| | - H Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto City, Kumamoto 860-8556, Japan
| |
Collapse
|
149
|
Cretu A, Brooks PC. Impact of the non-cellular tumor microenvironment on metastasis: potential therapeutic and imaging opportunities. J Cell Physiol 2008; 213:391-402. [PMID: 17657728 DOI: 10.1002/jcp.21222] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Evidence is accumulating that the malignant phenotype of a given tumor is dependent not only on the intrinsic characteristics of tumor cells, but also on the cooperative interactions of non-neoplastic cells, soluble secreted factors and the non-cellular solid-state ECM network that comprise the tumor microenvironment. Given the ability of the tumor microenvironment to regulate the cellular phenotype, recent efforts have focused on understanding the molecular mechanisms by which cells sense, assimilate, interpret, and ultimately respond to their immediate surroundings. Exciting new studies are beginning to unravel the complex interactions between the numerous cell types and regulatory factors within the tumor microenvironment that function cooperatively to control tumor cell invasion and metastasis. Here, we will focus on studies concerning a common theme, which is the central importance of the non-cellular solid-state compartment as a master regulator of the malignant phenotype. We will highlight the non-cellular solid-state compartment as a relatively untapped source of therapeutic and imaging targets and how cellular interactions with these targets may regulate tumor metastasis.
Collapse
Affiliation(s)
- Alexandra Cretu
- Department of Radiation Oncology, NYU Cancer Institute, New York University School of Medicine, New York, New York 10016, USA.
| | | |
Collapse
|
150
|
Nangia-Makker P, Raz T, Tait L, Hogan V, Fridman R, Raz A. Galectin-3 cleavage: a novel surrogate marker for matrix metalloproteinase activity in growing breast cancers. Cancer Res 2008; 67:11760-8. [PMID: 18089806 DOI: 10.1158/0008-5472.can-07-3233] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Failed therapies directed against matrix metalloproteinases (MMP) in cancer patients may be attributed, in part, to lack of diagnostic tools to differentiate between pro-MMPs and active MMPs, which indicate whether a treatment is efficacious or not. Because galectin-3 is cleavable in vitro by MMPs, we have developed differential antibodies recognizing its cleaved and noncleaved forms and tested their clinical utilization as a surrogate diagnostic marker for the presence of active MMPs in growing breast cancers. Wild-type and cleavage-resistant galectin-3 were constructed and expressed in galectin-3-null human breast carcinoma cells (BT-549). Tumorigenic and angiogenic potential of the clones was studied by injections into nude mice. MMP-2, MMP-9, full-length, and cleaved galectin-3 were localized in the xenografts by immunohistochemical analysis of paraffin-embedded sections using specific antibodies. Activities of MMP-2/9 were corroborated by in situ zymography on frozen tissue sections. Galectin-3 cleavage was shown in vivo by differential antibody staining and colocalized with predicted active MMPs both in mouse xenografts and human breast cancer specimens. In situ zymography validated these results. In addition, BT-549 cells harboring noncleavable galectin-3 showed reduced tumor growth and angiogenesis compared with the wild-type. We conclude that galectin-3 cleavage is an active process during tumor progression and could be used as a simple, rapid, and reliable surrogate marker for the activities of MMPs in growing breast cancers.
Collapse
Affiliation(s)
- Pratima Nangia-Makker
- Tumor Progression and Metastasis, Karmanos Cancer Institute, Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | |
Collapse
|