1
|
Sivaguru M, Fried G, Sivaguru BS, Sivaguru VA, Lu X, Choi KH, Saif MTA, Lin B, Sadayappan S. Cardiac muscle organization revealed in 3-D by imaging whole-mount mouse hearts using two-photon fluorescence and confocal microscopy. Biotechniques 2015; 59:295-308. [DOI: 10.2144/000114356] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/03/2015] [Indexed: 11/23/2022] Open
Abstract
The ability to image the entire adult mouse heart at high resolution in 3-D would provide enormous advantages in the study of heart disease. However, a technique for imaging nuclear/cellular detail as well as the overall structure of the entire heart in 3-D with minimal effort is lacking. To solve this problem, we modified the benzyl alcohol:benzyl benzoate (BABB) clearing technique by labeling mouse hearts with periodic acid Schiff (PAS) stain. We then imaged the hearts with a combination of two-photon fluorescence microscopy and automated tile-scan imaging/stitching. Utilizing the differential spectral properties of PAS, we could identify muscle and nuclear compartments in the heart. We were also able to visualize the differences between a 3-month-old normal mouse heart and a mouse heart that had undergone heart failure due to the expression of cardiac myosin binding protein-C (cMyBP-C) gene mutation (t/t). Using 2-D and 3-D morphometric analysis, we found that the t/t heart had anomalous ventricular shape, volume, and wall thickness, as well as a disrupted sarcomere pattern. We further validated our approach using decellularized hearts that had been cultured with 3T3 fibroblasts, which were tracked using a nuclear label. We were able to detect the 3T3 cells inside the decellularized intact heart tissue, achieving nuclear/cellular resolution in 3-D. The combination of labeling, clearing, and two-photon microscopy together with tiling eliminates laborious and time-consuming physical sectioning, alignment, and 3-D reconstruction.
Collapse
Affiliation(s)
- Mayandi Sivaguru
- Microscopy and Imaging Core Facility, Carl R. Woese Institute for Genomic Biology
| | - Glenn Fried
- Microscopy and Imaging Core Facility, Carl R. Woese Institute for Genomic Biology
| | | | | | - Xiaochen Lu
- Department of Cell and Developmental Biology and Carl R. Woese Institute for Genomic Biology
| | - Kyung Hwa Choi
- Department of Mechanical Science and Engineering, University of Illinois at Urbana Champaign, Urbana
| | - M Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana Champaign, Urbana
| | - Brian Lin
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Maywood, IL
| | - Sakthivel Sadayappan
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Maywood, IL
| |
Collapse
|
2
|
Effect of the expression of matrix metalloproteases and their tissue inhibitors on survival of patients with resectable colorectal cancer. Dig Dis Sci 2012; 57:2063-71. [PMID: 22488635 DOI: 10.1007/s10620-012-2154-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 03/16/2012] [Indexed: 12/09/2022]
Abstract
BACKGROUND Matrix metalloproteases (MMPs) and their tissue inhibitors (TIMPs) are of crucial importance in the degradation of the stromal connective tissue and basement membrane components. Study of the behavior of these components might help to predict the aggressiveness of tumors. AIMS To evaluate the expression and clinical relevance of MMPs and TIMPs for patients with resectable colorectal carcinoma. METHODS 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. Determinations were performed in cancer specimens from 104 patients with resectable colorectal cancer. The minimum period of follow-up was 12.5 years for patients without recurrence. To identify specific groups of tumors with distinct expression profiles, the data were analyzed by unsupervised hierarchical cluster analysis. RESULTS Expression of MMP-11 by fibroblasts and MMP-13 by tumor cells were associated with poor prognosis. The dendrogram revealed first-order division of tumors into two distinct MMP/TIMP molecular profiles, designated group 1 (n = 50) and group 2 (n = 54). Group 2 was characterized by significantly higher expression of MMP-1, 11, and 13, and TIMP-3. CONCLUSION Our results emphasize the prognostic value of MMP-11 and 13 expression in colorectal cancer.
Collapse
|
3
|
Zhu QC, Qin HL. Progress in understanding the role of epithelial-mesenchymal transition in the pathogenesis of colorectal tumors. Shijie Huaren Xiaohua Zazhi 2012; 20:1949-1956. [DOI: 10.11569/wcjd.v20.i21.1949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition is a well established biological event that plays an important role not only in the normal development of tissues and organs but also in the pathogenesis of many diseases. Increasing evidence has established its presence in the human colon during colorectal carcinogenesis and cancer invasion, chronic inflammation-related fibrosis, and mucosal healing. A large body of evidence supports the role of transforming growth factor-β and its downstream Smad signaling, the phosphatidylinositol 3'-kinase/Akt/mTOR axis, the Ras-mitogen-activated protein kinase/Snail/Slug and FOXC2 pathway, and Hedgehog signaling and microRNAs in epithelial-mesenchymal transition in the development of colorectal cancers. Here we discuss the role of these pathways in the initiation and development of the transition events. A better understanding of their induction and regulation may lead to the identification of pathways and factors that could be potent therapeutic targets.
Collapse
|
4
|
The metastasis-promoting protein S100A4 regulates mammary branching morphogenesis. Dev Biol 2010; 352:181-90. [PMID: 21195708 DOI: 10.1016/j.ydbio.2010.12.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 11/15/2010] [Accepted: 12/17/2010] [Indexed: 12/18/2022]
Abstract
High levels of the S100 calcium binding protein S100A4 also called fibroblast specific protein 1 (FSP1) have been established as an inducer of metastasis and indicator of poor prognosis in breast cancer. The mechanism by which S100A4 leads to increased cancer aggressiveness has yet to be established; moreover, the function of this protein in normal mammary gland biology has not been investigated. To address the role of S100A4 in normal mammary gland, its spatial and temporal expression patterns and possible function in branching morphogenesis were investigated. We show that the protein is expressed mainly in cells of the stromal compartment of adult humans, and during active ductal development, in pregnancy and in involution of mouse mammary gland. In 3D culture models, topical addition of S100A4 induced a significant increase in the TGFα mediated branching phenotype and a concomitant increase in expression of a previously identified branching morphogen, metalloproteinase-3 (MMP-3). These events were found to be dependent on MEK activation. Downregulation of S100A4 using shRNA significantly reduced TGFα induced branching and altered E-cadherin localization. These findings provide evidence that S100A4 is developmentally regulated and that it plays a functional role in mammary gland development, in concert with TGFα by activating MMP-3, and increasing invasion into the fat pad during branching. We suggest that S100A4-mediated effects during branching morphogenesis provide a plausible mechanism for how it may function in breast cancer progression.
Collapse
|
5
|
Karanikolas BDW, Figueiredo ML, Wu L. Polycomb group protein enhancer of zeste 2 is an oncogene that promotes the neoplastic transformation of a benign prostatic epithelial cell line. MOLECULAR CANCER RESEARCH : MCR 2009. [PMID: 19723877 DOI: 10.1158/1541‐7786.mcr‐09‐0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Polycomb group protein enhancer of zeste 2 (EZH2) is a master regulatory protein that plays a critical role in development as part of the polycomb repressive complex 2. Polycomb repressive complex 2 controls numerous cell cycle and regulatory genes through trimethylation of histone 3, which results in chromatin condensation and transcriptional silencing. EZH2 overexpression has been correlated with high incidence of more aggressive, metastatic prostate cancers. Although this correlation means EZH2 could prove valuable as a biomarker in clinical settings, the question remains whether EZH2 is actually responsible for the initiation of these more aggressive tumor types. In this study, EZH2-mediated neoplastic transformation of the normal prostate epithelial cell line benign prostate hyperplasia 1 (BPH1) was confirmed by in vivo tumor growth and in vitro colony formation. Furthermore, EZH2 transformation resulted in increased invasive behavior of BPH1 cells, indicating that EZH2 may be responsible for aggressive behavior in prostate cancers. BPH1 was also transformed with the classic oncogenes myristoylated Akt and activated Ras(V12) to allow phenotype comparisons with the EZH2-transformed cells. This study marks the first demonstration of neoplastic transformation in prostate cells mediated by EZH2 and establishes that EZH2 possesses stronger transforming activity than Akt but weaker activity than activated Ras.
Collapse
Affiliation(s)
- Breanne D W Karanikolas
- Departments of Molecular and Medical Pharmacology and Urology, University of California at Los Angeles, Los Angeles, California 90095-1738, USA
| | | | | |
Collapse
|
6
|
Karanikolas BDW, Figueiredo ML, Wu L. Polycomb group protein enhancer of zeste 2 is an oncogene that promotes the neoplastic transformation of a benign prostatic epithelial cell line. Mol Cancer Res 2009; 7:1456-65. [PMID: 19723877 DOI: 10.1158/1541-7786.mcr-09-0121] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Polycomb group protein enhancer of zeste 2 (EZH2) is a master regulatory protein that plays a critical role in development as part of the polycomb repressive complex 2. Polycomb repressive complex 2 controls numerous cell cycle and regulatory genes through trimethylation of histone 3, which results in chromatin condensation and transcriptional silencing. EZH2 overexpression has been correlated with high incidence of more aggressive, metastatic prostate cancers. Although this correlation means EZH2 could prove valuable as a biomarker in clinical settings, the question remains whether EZH2 is actually responsible for the initiation of these more aggressive tumor types. In this study, EZH2-mediated neoplastic transformation of the normal prostate epithelial cell line benign prostate hyperplasia 1 (BPH1) was confirmed by in vivo tumor growth and in vitro colony formation. Furthermore, EZH2 transformation resulted in increased invasive behavior of BPH1 cells, indicating that EZH2 may be responsible for aggressive behavior in prostate cancers. BPH1 was also transformed with the classic oncogenes myristoylated Akt and activated Ras(V12) to allow phenotype comparisons with the EZH2-transformed cells. This study marks the first demonstration of neoplastic transformation in prostate cells mediated by EZH2 and establishes that EZH2 possesses stronger transforming activity than Akt but weaker activity than activated Ras.
Collapse
Affiliation(s)
- Breanne D W Karanikolas
- Departments of Molecular and Medical Pharmacology and Urology, University of California at Los Angeles, Los Angeles, California 90095-1738, USA
| | | | | |
Collapse
|
7
|
Quaresima B, Romeo F, Faniello MC, Di Sanzo M, Liu CG, Lavecchia A, Taccioli C, Gaudio E, Baudi F, Trapasso F, Croce CM, Cuda G, Costanzo F. BRCA1 5083del19 mutant allele selectively up-regulates periostin expression in vitro and in vivo. Clin Cancer Res 2008; 14:6797-803. [PMID: 18980973 DOI: 10.1158/1078-0432.ccr-07-5208] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of this study was to explore the gene expression pattern produced by the cancer-associated BRCA1 5083del19 founder mutation by using a microarray analysis. Such a mutation, identified in a subset of familial breast cancer patients, involves a deletion at the 3' end of the BRCA1 messenger leading, in the mature protein, to the ablation of the BRCT tandem domain. EXPERIMENTAL DESIGN We generated HeLa cells stably expressing both exogenous wild-type (HeLa/(wt)BRCA1), used as a control, and 5083del19 BRCA1 (HeLa/(5083del19)BRCA1) alleles; gene chips were then used to investigate any changes in the transcription profile induced by the 5083del19 BRCA1 mutant compared with controls. RESULTS Among the genes showing perturbation of their expression, periostin was found to be up-regulated in HeLa/(5083del19)BRCA1 cells to an extent of 72-fold versus HeLa/(pcDNA3.1/empty) and 76-fold versus HeLa/(wt)BRCA1 cells. This finding was validated both in vitro in breast cancer cell lines harboring mutations of BRCA1 and in vivo by immunohistochemistry of breast cancer specimens bearing the 5083del19 BRCA1 mutation as well as by Western blot analysis of sera obtained from patients and healthy carriers of the same mutation. CONCLUSIONS Our results suggest that periostin overexpression, whose product is released from cells in the extracellular fluids, might be a potential marker for early cancer detection in a specific subset of hereditary breast carcinomas triggered by cancer-associated BRCA1 mutations that affect the BRCT tandem domain.
Collapse
Affiliation(s)
- Barbara Quaresima
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Ingber DE. Can cancer be reversed by engineering the tumor microenvironment? Semin Cancer Biol 2008; 18:356-64. [PMID: 18472275 DOI: 10.1016/j.semcancer.2008.03.016] [Citation(s) in RCA: 196] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 03/20/2008] [Accepted: 03/25/2008] [Indexed: 01/08/2023]
Abstract
To advance cancer research in a transformative way, we must redefine the problem. Although epithelial cancers, such as breast cancer, may be caused by random somatic gene mutations, the reality is that this is only one of many ways to induce tumor formation. Cancers also can be produced in experimental systems in vitro and in vivo, for example, by inducing sustained alterations of extracellular matrix (ECM) structure. Moreover, certain epithelial cancers can be induced to 'reboot' and regenerate normal tissue morphology when combined with embryonic mesenchyme or exogenous ECM scaffolds that are produced through epithelial-stromal interactions. At the same time, work in the field of Mechanical Biology has revealed that many cell behaviors critical for cancer formation (e.g., growth, differentiation, motility, apoptosis) can be controlled by physical interactions between cells and their ECM adhesions that alter the mechanical force balance in the ECM, cell and cytoskeleton. Epithelial tumor progression also can be induced in vitro by changing ECM mechanics or altering cytoskeletal tension generation through manipulation of the Rho GTPase signaling pathway. Mechanical interactions between capillary cells and ECM that are mediated by Rho signaling similarly mediate control of capillary cell growth and angiogenesis, which are equally critical for cancer progression and metastasis. These findings question basic assumptions in the cancer field, and raise the intriguing possibility that cancer may be a reversible disease that results from progressive deregulation of tissue architecture, which leads to physical changes in cells and altered mechanical signaling. This perspective raises the possibility of developing a tissue engineering approach to cancer therapy in which biologically inspired materials that mimic the embryonic microenvironment are used to induce cancers to revert into normal tissues.
Collapse
Affiliation(s)
- Donald E Ingber
- Vascular Biology Program, Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
9
|
Abstract
AIM: To investigate the expression and localization of periostin in gastric cancer and its clinical relevance.
METHODS: Reverse transcriptase polymerase chain reaction was used to measure periostin mRNA expression. Western blotting was carried out to detect periostin protein expression. Immunohistochemistry was performed to localize and quantify the expression of periostin in benign gastric diseases and gastric cancer, and immunostaining results were correlated with gastric cancer pathological stages.
RESULTS: Periostin expression was low at both mRNA and protein levels in normal gastric tissues, but was overexpressed in gastric cancer tissues. Immunohistochemical staining revealed that periostin was overexpressed in primary gastric cancer, as well as in metastatic lymph nodes, but only faint staining was found in benign gastric ulcers. By quantitative analysis of the immunostaining results, periostin expression was increased 2.5-4-fold in gastric cancer, compared to that in benign gastric disease, and there was a trend toward increasing periostin expression with tumor stage.
CONCLUSION: This observation demonstrated that periostin was overexpressed in gastric cancer and lymph node metastasis, which suggests that periostin plays an important role in the progression and metastasis of gastric cancer.
Collapse
Affiliation(s)
- Jun-Sheng Li
- Department of General Surgery, Affiliated Zhong-Da Hospital, Southeast University, Nanjing 210009, Jiangsu Province, China.
| | | | | | | |
Collapse
|
10
|
González LO, Pidal I, Junquera S, Corte MD, Vázquez J, Rodríguez JC, Lamelas ML, Merino AM, García-Muñiz JL, Vizoso FJ. Overexpression of matrix metalloproteinases and their inhibitors in mononuclear inflammatory cells in breast cancer correlates with metastasis-relapse. Br J Cancer 2007; 97:957-63. [PMID: 17848954 PMCID: PMC2360420 DOI: 10.1038/sj.bjc.6603963] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
An immunohistochemical study was performed using tissue microarrays and specific antibodies against matrix metalloproteinase (MMP)-1, -2, -7, -9, -11, -13 and –14, tissular inhibitors of metalloproteinase (TIMP)-1, -2 and -3. More than 2600 determinations on cancer specimens from 131 patients with primary ductal invasive tumours of the breast were performed. To identify specific groups of tumours with distinct expression profiles the data were analysed by unsupervised hierarchical cluster analysis by each cellular type. We did not find well-defined cluster of cases for tumour cells or fibroblastic cells. However, for mononuclear inflammatory cells the dendogram shows a first-order division of the tumours into two distinct MMP/TIMP molecular profiles, designated group 1 (n=89) and group 2 (n=42). Matrix metalloproteinase-7, -9, -11, -13 and -14, and TIMP-1 and -2, were identified as showing significant high expression in group 2 compared with group 1. Multivariate analysis demonstrated that clustering for mononuclear inflammatory cells was the most potent independent factor associated with distant relapse-free survival (group 2: 5.6 (3.5–9.6), P<0.001). We identify a phenotype of mononuclear inflammatory cells infiltrating tumours, which is associated with the development of distant metastasis. Therefore, this finding suggests that these host inflammatory cells could be a possible target for inhibition of metastasis.
Collapse
Affiliation(s)
- L O González
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
- Servicio de Anatomía Patológica, Hospital de Jove, Gijón, Spain
| | - I Pidal
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
| | - S Junquera
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
| | - M D Corte
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
| | - J Vázquez
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
| | - J C Rodríguez
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
- Servicio de Cirugía General, Hospital de Jove, Gijón, Spain
| | - M L Lamelas
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
- Servicio de Ginecología, Hospital de Jove, Gijón, Spain
| | - A M Merino
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
- Servicio de Anatomía Patológica, Hospital de Cabueñes, Gijón, Spain
| | - J L García-Muñiz
- Servicio de Cirugía General, Hospital Universitario Central de Asturias, Asturias, Oviedo, Spain
| | - F J Vizoso
- Unidad de Investigación, Hospital de Jove, Gijón, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Asturias, Oviedo, Spain
- Servicio de Cirugía General, Hospital de Jove, Gijón, Spain
- Servicio de Cirugía General, Hospital de Jove, Avda. Eduardo Castro s/n, Asturias 33290, Gijón, Spain. E-mail:
| |
Collapse
|
11
|
Kokkinos MI, Wafai R, Wong MK, Newgreen DF, Thompson EW, Waltham M. Vimentin and epithelial-mesenchymal transition in human breast cancer--observations in vitro and in vivo. Cells Tissues Organs 2007; 185:191-203. [PMID: 17587825 DOI: 10.1159/000101320] [Citation(s) in RCA: 281] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Breast cancer is a highly prevalent disease among women worldwide. While the expression of certain proteins within these tumours is used for prognosis and selection of therapies, there is a continuing need for additional markers to be identified. A considerable amount of current literature, based predominantly on cell culture systems, suggests that a major mechanism responsible for the progression of breast cancer is due to tumour cells losing their epithelial features and gaining mesenchymal properties. These events are proposed to be very similar to the epithelial-mesenchymal transition (EMT) process that has been well characterised in embryonic development. For the developmental and putative cancer EMT, the cell intermediate filament status changes from a keratin-rich network which connects to adherens junctions and hemidesmosomes, to a vimentin-rich network connecting to focal adhesions. This review summarises observations of vimentin expression in breast cancer model systems, and discusses the potential role of EMT in human breast cancer progression, and the prognostic usefulness of vimentin expression.
Collapse
Affiliation(s)
- Maria I Kokkinos
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, Australia.
| | | | | | | | | | | |
Collapse
|
12
|
Haupt LM, Thompson EW, Trezise AEO, Irving RE, Irving MG, Griffiths LR. In vitro and in vivo MMP gene expression localisation by In Situ-RT-PCR in cell culture and paraffin embedded human breast cancer cell line xenografts. BMC Cancer 2006; 6:18. [PMID: 16430785 PMCID: PMC1397851 DOI: 10.1186/1471-2407-6-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Accepted: 01/24/2006] [Indexed: 01/16/2023] Open
Abstract
Background Members of the matrix metalloproteinase (MMP) family of proteases are required for the degradation of the basement membrane and extracellular matrix in both normal and pathological conditions. In vitro, MT1-MMP (MMP-14, membrane type-1-MMP) expression is higher in more invasive human breast cancer (HBC) cell lines, whilst in vivo its expression has been associated with the stroma surrounding breast tumours. MMP-1 (interstitial collagenase) has been associated with MDA-MB-231 invasion in vitro, while MMP-3 (stromelysin-1) has been localised around invasive cells of breast tumours in vivo. As MMPs are not stored intracellularly, the ability to localise their expression to their cells of origin is difficult. Methods We utilised the unique in situ-reverse transcription-polymerase chain reaction (IS-RT-PCR) methodology to localise the in vitro and in vivo gene expression of MT1-MMP, MMP-1 and MMP-3 in human breast cancer. In vitro, MMP induction was examined in the MDA-MB-231 and MCF-7 HBC cell lines following exposure to Concanavalin A (Con A). In vivo, we examined their expression in archival paraffin embedded xenografts derived from a range of HBC cell lines of varied invasive and metastatic potential. Mouse xenografts are heterogenous, containing neoplastic human parenchyma with mouse stroma and vasculature and provide a reproducible in vivo model system correlated to the human disease state. Results In vitro, exposure to Con A increased MT1-MMP gene expression in MDA-MB-231 cells and decreased MT1-MMP gene expression in MCF-7 cells. MMP-1 and MMP-3 gene expression remained unchanged in both cell lines. In vivo, stromal cells recruited into each xenograft demonstrated differences in localised levels of MMP gene expression. Specifically, MDA-MB-231, MDA-MB-435 and Hs578T HBC cell lines are able to influence MMP gene expression in the surrounding stroma. Conclusion We have demonstrated the applicability and sensitivity of IS-RT-PCR for the examination of MMP gene expression both in vitro and in vivo. Induction of MMP gene expression in both the epithelial tumour cells and surrounding stromal cells is associated with increased metastatic potential. Our data demonstrate the contribution of the stroma to epithelial MMP gene expression, and highlight the complexity of the role of MMPs in the stromal-epithelial interactions within breast carcinoma.
Collapse
Affiliation(s)
- Larisa M Haupt
- Genomics Research Centre, Griffith University Gold Coast, School of Medical Science, Griffith University, Queensland, 4217,Australia
- IMCB, Biopolis, Singapore
| | - Erik W Thompson
- VBCRC Invasion and Metastasis Unit, St. Vincent's Institute of Medical Research and University of Melbourne, Department of Surgery, Melbourne, Victoria, 3065, Australia
| | - Ann EO Trezise
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Rachel E Irving
- Genomics Research Centre, Griffith University Gold Coast, School of Medical Science, Griffith University, Queensland, 4217,Australia
| | - Michael G Irving
- Institute of Health Sciences, Bond University, Queensland, 4229, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Griffith University Gold Coast, School of Medical Science, Griffith University, Queensland, 4217,Australia
| |
Collapse
|
13
|
Shao R, Bao S, Bai X, Blanchette C, Anderson RM, Dang T, Gishizky ML, Marks JR, Wang XF. Acquired expression of periostin by human breast cancers promotes tumor angiogenesis through up-regulation of vascular endothelial growth factor receptor 2 expression. Mol Cell Biol 2004; 24:3992-4003. [PMID: 15082792 PMCID: PMC387763 DOI: 10.1128/mcb.24.9.3992-4003.2004] [Citation(s) in RCA: 257] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The late stages of human breast cancer development are poorly understood complex processes associated with the expression of genes by cancers that promote specific tumorigenic activities, such as angiogenesis. Here, we describe the identification of periostin as a mesenchyme-specific gene whose acquired expression by human breast cancers leads to a significant enhancement in tumor progression and angiogenesis. Undetectable in normal human breast tissues, periostin was found to be overexpressed by the vast majority of human primary breast cancers examined. Tumor cell lines engineered to overexpress periostin showed a phenotype of accelerated growth and angiogenesis as xenografts in immunocompromised animals. The underlying mechanism of periostin-mediated induction of angiogenesis was found to derive in part from the up-regulation of the vascular endothelial growth factor receptor Flk-1/KDR by endothelial cells through an integrin alpha(v)beta(3)-focal adhesion kinase-mediated signaling pathway. These findings demonstrate the presence of a novel mechanism by which tumor angiogenesis is acquired with the expression of a mesenchyme-specific gene as a crucial step in late stages of tumorigenesis.
Collapse
Affiliation(s)
- Rong Shao
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Carcinogenesis - the process of cancer formation - is commonly discussed in terms of genetic alterations that lead to deregulation of cell growth. Recently, there has been a resurgence of interest in epigenetic factors and, in particular, the role of the stromal microenvironment and angiogenesis in tumor formation. In this article, cancer is presented as a disease of the developmental processes that govern how cells organize into tissues and tissues into organs. This histogenetic perspective raises the possibility that epithelial-mesenchymal interactions and the extracellular matrix (basement membrane) that is deposited through these interactions may actively contribute to the carcinogenic process. Experimental work is reviewed that confirms that extracellular matrix plays a key role in normal histodifferentiation during both epitheliogenesis and angiogenesis, and that epigenetic deregulation of cell-matrix interactions may actively promote tumor initiation and progression. The contributions of integrins, cytoskeleton, tensegrity and local variations in extracellular matrix mechanics to these processes are discussed, as are the implications of this work for future studies on cancer formation.
Collapse
Affiliation(s)
- Donald E Ingber
- Department of Pathology and Surgery, Harvard Medical School and Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Vécsey-Semjén B, Becker KF, Sinski A, Blennow E, Vietor I, Zatloukal K, Beug H, Wagner E, Huber LA. Novel colon cancer cell lines leading to better understanding of the diversity of respective primary cancers. Oncogene 2002; 21:4646-62. [PMID: 12096341 DOI: 10.1038/sj.onc.1205577] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2001] [Revised: 03/05/2002] [Accepted: 04/15/2002] [Indexed: 11/09/2022]
Abstract
A major obstacle to obtaining more detailed insights into the diversity of phenotypic and molecular changes occurring in colon cancer cells is the lack of low-passage colon cancer cell lines, which would still closely reflect the phenotype of the colon cancer cells in vivo. Here, we characterize eight novel, low passage number human colon carcinoma cell lines, originating from colorectal cancers extensively characterized in the clinics. All cell lines closely resemble the original tumors with respect to phenotype, markers and detectable genetic changes. Cell morphology and marker expression is highly variable, ranging from fully polarized cells correctly expressing all basolateral epithelial markers, to cells with mesenchymal characteristics and a complete loss of polarity due to delocalization or loss of junction complex proteins. The alterations in phenotype and epithelial marker expression correspond to changes already detectable in the primary tumor in vivo. Seven of the cell lines show chromosomal instability, while one cell line is characterized by microsatellite instability. p53 associated with K-ras mutations were detected in three cell lines. Hitherto non-described E-cadherin mutations were found at both alleles in one cell line whereas in another cell line the E-cadherin protein was down-regulated. A stabilizing beta-catenin mutation (S45F) appears in the same cell line that carried the mutated E-cadherin gene. Six cell lines carried APC mutations, which in five of the lines led to an activated beta-catenin/Tcf/LEF signaling pathway. In accordance with beta-catenin/Tcf/LEF activation, the cell lines show increased migration and invasiveness. Our results show that the characterized, low-passage cell lines mirror the diversity of the individual tumors from which they were derived. Through molecular analyses of these cell lines we demonstrate that tumorgenicity events are much more diverse in human colon cancer than expected, despite the common origin of the tumors from a small patient group with similar tumor grading and clinical prognosis.
Collapse
|