51
|
Martin NK, Gaffney EA, Gatenby RA, Maini PK. Tumour-stromal interactions in acid-mediated invasion: a mathematical model. J Theor Biol 2010; 267:461-70. [PMID: 20816684 PMCID: PMC3005191 DOI: 10.1016/j.jtbi.2010.08.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 08/23/2010] [Accepted: 08/25/2010] [Indexed: 10/19/2022]
Abstract
It is well established that the tumour microenvironment can both promote and suppress tumour growth and invasion, however, most mathematical models of invasion view the normal tissue as inhibiting tumour progression via immune modulation or spatial constraint. In particular, the production of acid by tumour cells and the subsequent creation of a low extracellular pH environment has been explored in several 'acid-mediated tumour invasion' models where the acidic environment facilitates normal cell death and permits tumour invasion. In this paper, we extend the acid-invasion model developed by Gatenby and Gawlinski (1996) to include both the competitive and cooperative interactions between tumour and normal cells, by incorporating the influence of extracellular matrix and protease production at the tumour-stroma interface. Our model predicts an optimal level of tumour acidity which produces both cell death and matrix degradation. Additionally, very aggressive tumours prevent protease production and matrix degradation by excessive normal cell destruction, leading to an acellular (but matrix filled) gap between the tumour and normal tissue, a feature seen in encapsulated tumours. These results suggest, counterintuitively, that increasing tumour acidity may, in some cases, prevent tumour invasion.
Collapse
Affiliation(s)
- Natasha K Martin
- Centre for Mathematical Biology, Mathematical Institute, Oxford University, 24-29 St Giles', Oxford OX1 3LB, UK.
| | | | | | | |
Collapse
|
52
|
Fry JL, Toker A. Secreted and membrane-bound isoforms of protease ADAM9 have opposing effects on breast cancer cell migration. Cancer Res 2010; 70:8187-98. [PMID: 20736367 DOI: 10.1158/0008-5472.can-09-4231] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumor cell migration is mediated by cell-autonomous signaling mechanisms as well as paracrine and autocrine factors secreted by activated stromal cells in the tumor microenvironment. Like other members of the ADAM (a disintegrin and metalloproteinase) family, the integrin-binding metalloproteinase ADAM9 modulates cell-cell and cell-matrix interactions as well as ectodomain shedding of cell surface receptors and ligands, thereby modifying intracellular and extracellular signaling. ADAM9 transcripts are alternatively spliced to express a transmembrane protein (ADAM9-L) and a secreted variant (ADAM9-S). In this study, we show that ADAM9-S promotes breast cancer cell migration in a manner requiring its metalloproteinase activity, whereas ADAM9-L suppresses cell migration independent of its metalloproteinase activity. Suppression of migration by ADAM9-L requires a functional disintegrin domain and integrin binding. Expression analysis revealed that both ADAM9 isoforms are expressed in breast cancer cell lines and tissues. Therefore, relative levels of membrane-tethered and secreted variants of ADAM9 are a key determinant in manifestation of aggressive migratory phenotypes associated with breast cancer progression.
Collapse
Affiliation(s)
- Jessica L Fry
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
53
|
Tai CJ, Hsu CH, Shen SC, Lee WR, Jiang MC. Cellular apoptosis susceptibility (CSE1L/CAS) protein in cancer metastasis and chemotherapeutic drug-induced apoptosis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:110. [PMID: 20701792 PMCID: PMC2925819 DOI: 10.1186/1756-9966-29-110] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 08/11/2010] [Indexed: 11/10/2022]
Abstract
The cellular apoptosis susceptibility (CSE1L/CAS) protein is highly expressed in cancer, and its expression is positively correlated with high cancer stage, high cancer grade, and worse outcomes of patients. CSE1L (or CAS) regulates chemotherapeutic drug-induced cancer cell apoptosis and may play important roles in mediating the cytotoxicities of chemotherapeutic drugs against cancer cells in cancer chemotherapy. CSE1L was originally regarded as a proliferation-associated protein and was thought to regulate the proliferation of cancer cells in cancer progression. However, the results of experimental studies showed that enhanced CSE1L expression is unable to increase proliferation of cancer cells and CSE1L regulates invasion and metastasis but not proliferation of cancer cells. Recent studies revealed that CSE1L is a secretory protein, and there is a higher prevalence of secretory CSE1L in the sera of patients with metastatic cancer. Therefore, CSE1L may be a useful serological marker for screening, diagnosis and prognosis, assessment of therapeutic responses, and monitoring for recurrence of cancer. In this paper, we review the expression of CSE1L in cancer and discuss why CSE1L regulates the invasion and metastasis rather than the proliferation of cancer.
Collapse
Affiliation(s)
- Cheng-Jeng Tai
- Section of Hematology-Oncology, Department of Medicine, Taipei Medical University and Hospital, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
54
|
Liu W, Li L, Wang X, Ren L, Wang X, Wang J, Tu Q, Huang X, Wang J. An integrated microfluidic system for studying cell-microenvironmental interactions versatilely and dynamically. LAB ON A CHIP 2010; 10:1717-24. [PMID: 20422110 DOI: 10.1039/c001049a] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
We presented an integrated microfluidic system for dynamical study of cell-microenvironmental interactions. We demonstrated its precisely spatio-temporal control in the flow direction and the multi-site staying of the fluids by groups of monolithic microfabricated valves through digital operation, aside from the regulated communication between two loci based on real-time microenvironment transition. Using this system, a series of functional manipulations, including specific delivery, addressable surface treatment, positional cell loading and co-culture were performed quickly and efficiently for biological applications. Sequentially, we carried out the potential utility of this system in the research of dynamic microenvironmental influence to cells using a patho-physiological interaction during cancer initiation and progression. Our results exhibit the passive role but collaborative response of NIH 3T3 fibroblasts to the soluble signals from hepatocellular carcinoma cells, and also the variable behaviors of carcinoma cells under different environmental stimulation. This system can facilitate the in vitro investigation of cell-microenvironmental interactions occurred in numerous biological and pathogenic processes.
Collapse
Affiliation(s)
- Wenming Liu
- College of Animal Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Chenau J, Michelland S, de Fraipont F, Josserand V, Coll JL, Favrot MC, Seve M. The cell line secretome, a suitable tool for investigating proteins released in vivo by tumors: application to the study of p53-modulated proteins secreted in lung cancer cells. J Proteome Res 2010; 8:4579-91. [PMID: 19639960 DOI: 10.1021/pr900383g] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Malignant processes such as metastasis, invasion, or angiogenesis are tightly dependent on the composition of the extracellular medium, which is itself affected by the release of proteins by the tumor cells. p53, a major tumor suppressor protein very frequently mutated and/or inactivated in cancer cells, is known to modulate the release of proteins by the tumor cells; however, while p53-modulated intracellular proteins have been extensively studied, little is known concerning their extracellular counterparts. Here, we characterized the p53-dependent secretome of a lung tumor model in vitro (H358 human nonsmall cell lung adenocarcinoma cell line with a homozygous deletion of p53) and demonstrate that the modulation of exported proteins can also be detected in vivo in the plasma of tumor-bearing mice. We used a clone of H358, stably transfected with a tetracycline-inducible wild-type p53-expressing vector. With the use of iTRAQ labeling and LC-MALDI-MS/MS analysis, we identified 909 proteins released in vitro by the cells, among which 91 are p53-modulated. Three proteins (GDF-15, FGF-19, and VEGF) were also investigated in H358/TetOn/p53 xenograft mice. The ELISA dosage on total tumor protein extracts confirmed the influence of p53 on the release of these proteins in vivo. Moreover, the GDF-15 concentration was measured in the plasma and its p53-dependent modulation was confirmed. To our knowledge, this is the first report establishing that the in vitro cell line secretome is reliable and reflects the extracellular release of proteins from tumor cells in vivo and could be used to identify putative tumor markers.
Collapse
Affiliation(s)
- Jérôme Chenau
- Université Joseph Fourier-Grenoble 1, INSERM, Institut Albert Bonniot U823, Grenoble, France
| | | | | | | | | | | | | |
Collapse
|
56
|
Chatzinikolaou G, Nikitovic D, Berdiaki A, Zafiropoulos A, Katonis P, Karamanos NK, Tzanakakis GN. Heparin regulates colon cancer cell growth through p38 mitogen-activated protein kinase signalling. Cell Prolif 2009; 43:9-18. [PMID: 19845689 DOI: 10.1111/j.1365-2184.2009.00649.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Heparin acts as an extracellular stimulus capable of activating major cell signalling pathways. Thus, we examined the putative mechanisms utilized by heparin to stimulate HT29, SW1116 and HCT116 colon cancer cell growth. MATERIALS AND METHODS Possible participation of the mitogen-activated protein kinase (MAPK) cascade on heparin-induced HT29, SW1116 and HCT116 colon cancer cell growth was evaluated using specific MAPK cascade inhibitors, Western blot analysis, real-time quantitative PCR and FACS apoptosis analysis. RESULTS Treatment with a highly specific p38 kinase inhibitor, SB203580, significantly (50-70%) inhibited heparin-induced colon cancer cell growth, demonstrating that p38 MAPK signalling is involved in their heparin-induced proliferative response. This was shown to be correlated with increased (up to 3-fold) phosphorylation of 181/182 threonine/tyrosine residues on p38 MAP kinase. Furthermore, heparin inhibited cyclin-dependent kinase inhibitor p21(WAF1/CIP1) and p53 tumour suppressor gene and protein expression up to 2-fold or 1.8-fold, respectively, and stimulated cyclin D1 expression up to 1.8-fold, in these cell lines through a p38-mediated mechanism. On the other hand, treatment with heparin did not appear to affect HT29, SW1116 and HCT116 cell levels of apoptosis. CONCLUSIONS This study demonstrates that an extracellular glycosaminoglycan, heparin, finely modulates expression of genes crucial to cell cycle regulation through specific activation of p38 MAP kinase to stimulate colon cancer cell growth.
Collapse
Affiliation(s)
- G Chatzinikolaou
- Department of Histology, Division of Morphology, School of Medicine, University of Crete, Heraklion, Greece
| | | | | | | | | | | | | |
Collapse
|
57
|
Baglole CJ, Ray DM, Bernstein SH, Feldon SE, Smith TJ, Sime PJ, Phipps RP. More Than Structural Cells, Fibroblasts Create and Orchestrate the Tumor Microenvironment. Immunol Invest 2009; 35:297-325. [PMID: 16916756 DOI: 10.1080/08820130600754960] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The tumor microenvironment comprises many cell types including infiltrating immune cells such as lymphocytes, endothelial cells and a complex stroma consisting mainly of fibroblasts. Fibroblasts are heterogeneous and consist of Thy-1+ and Thy-1- subsets that define different biosynthetic and differentiation potential. They produce mediators linked to carcinogenesis and metastasis, including Cox-2 and PGE2, both of which are also increased in most cancers. This review will highlight the emerging role of the complex fibroblastic stroma in establishing a microenvironment supporting malignant transformation, tumor growth and attenuation of host anti-tumor immune responses.
Collapse
Affiliation(s)
- Carolyn J Baglole
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, and Lymphoma Biology Program, James P. Wilmot Cancer Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
58
|
FOXO3a promotes tumor cell invasion through the induction of matrix metalloproteinases. Mol Cell Biol 2009; 29:4906-17. [PMID: 19564415 DOI: 10.1128/mcb.00077-09] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The role of the Forkhead transcription factor FOXO3a in processes that promote tumor metastasis is poorly defined. Here, we show that depletion of FOXO3a from cancer cells leads to decreased tumor size specifically due to attenuated invasive migration. During tumor progression, an increase in tumor mass is concomitant with serum deprivation prior to tumor angiogenesis. We show that nuclear retention of FOXO3a due to serum starvation results in greatly increased cancer cell invasion. Exploration of the mechanism by which FOXO3a promotes invasive migration revealed that it induces the expression of matrix metalloproteinase 9 (MMP-9) and MMP-13, both of which have been causally linked to the invasion and progression of numerous human solid tumors. Our results link Forkhead transcription factors to a previously unexplored function in cancer progression by promoting extracellular matrix degradation, allowing tumors to invade neighboring tissues and ultimately metastasize to distant organs.
Collapse
|
59
|
Stromal expression of MMP-13 is required for melanoma invasion and metastasis. J Invest Dermatol 2009; 129:2686-93. [PMID: 19516266 DOI: 10.1038/jid.2009.130] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor invasion and metastasis of malignant melanoma have been shown to require proteolytic degradation of the extracellular environment achieved primarily by enzymes of the matrix metalloproteinases (MMP) family. We have earlier shown that increased enzyme activity is localized at the border of tumor cells and the adjacent peritumoral connective tissue, emphasizing the importance of tumor-stroma interactions in the regulation of MMP activity. To confirm the role of stroma-derived MMP-13 in the invasion process, we investigated the invasiveness of melanoma cells upon intradermal injection in mice with complete inactivation of MMP-13. Tumor growth was significantly impaired in mmp-13(-/-) mice and most significant at early time points as compared with wild-type littermates. Moreover, metastasis to various organs was reduced to 17.6 vs 30% in lungs, 2.9 vs 30% in the liver. Strikingly, ablation of MMP-13 completely abrogated formation of metastasis in the heart (0 vs 40%). Notably, decreased tumor growth in mmp-13(-/-) mice was associated with reduced blood vessel density. In addition, decreased blood vessel permeability in the tumors was measured by magnetic resonance imaging of tumor-bearing animals. These data suggest an important role of MMP-13 in tumor growth and an unexpected role in organ-specific metastasis of melanoma cells.
Collapse
|
60
|
Tung MC, Tsai CSS, Tung JN, Tsao TY, Chen HC, Yeh KT, Liao CF, Jiang MC. Higher prevalence of secretory CSE1L/CAS in sera of patients with metastatic cancer. Cancer Epidemiol Biomarkers Prev 2009; 18:1570-7. [PMID: 19383891 DOI: 10.1158/1055-9965.epi-08-0948] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Metastatic markers are highly useful diagnostic and prognostic indicators of cancer metastasis. Herein, we report that secretory CSE1L/CAS, a cellular apoptosis susceptibility protein, is a new marker for metastatic cancer. CAS was colocalized with matrix metalloproteinase-2 in vesicles surrounding the outside of MCF-7 cell membranes, and the COOH-terminal domain of CAS was associated with matrix metalloproteinase-2-containing vesicles. Immunohistochemical staining for CAS was positive in the stroma and gland lumens of human metastatic cancer tissues. CAS was also detected in conditioned medium from B16-F10 melanoma cells and more frequently in the sera of patients with metastatic cancer than in sera from patients with primary cancer. Specifically, the prevalence of serum CAS in serum samples from 146 patients was 58.2% (32 of 55), 32.0% (8 of 25), and 12.1% (8 of 66) for patients with metastatic, invasive, and primary cancers, respectively. Our results suggest that CAS is a secretory protein associated with cancer metastasis, which may have clinical utility in metastatic cancer screening and diagnosis.
Collapse
Affiliation(s)
- Min-Che Tung
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung County 435, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Nieman LT, Jakovljevic M, Sokolov K. Compact beveled fiber optic probe design for enhanced depth discrimination in epithelial tissues. OPTICS EXPRESS 2009; 17:2780-96. [PMID: 19219183 DOI: 10.1364/oe.17.002780] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
We report the development and evaluation of a simple compact probe that incorporates multiple beveled fibers for depth sensitive detection of spectroscopic signals in vivo. We evaluated three probes with bevel angles 35, 40, and 45 degrees for their collection efficiency and depth resolution using a thin highly scattering white substrate and found that a 40 degree bevel provides the best characteristics for depth-resolved spectroscopy. The depth sensitivity of the probe with 40 degree beveled fibers was then evaluated using multilayer phantoms with scattering properties mimicking precancerous tissue and in vivo on normal human oral mucosa. The results demonstrate that the use of multiple beveled fibers has the capability to simultaneously collect scattering spectra from a range of depths within epithelial tissue that has the potential to provide further significant improvement of detection and monitorin of epithelial precancers.
Collapse
Affiliation(s)
- Linda T Nieman
- Department of Biomedical Engineering, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Austin, TX 77030, USA.
| | | | | |
Collapse
|
62
|
Tristram Arscott W, LaBauve AE, May V, Wesley UV. Suppression of neuroblastoma growth by dipeptidyl peptidase IV: relevance of chemokine regulation and caspase activation. Oncogene 2009; 28:479-91. [PMID: 18978811 PMCID: PMC2633428 DOI: 10.1038/onc.2008.402] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 09/10/2008] [Accepted: 09/28/2008] [Indexed: 02/07/2023]
Abstract
Imbalanced protease expression and activities may contribute to the development of cancers, including neuroblastoma (NB). NB is a fatal childhood cancer of the sympathetic nervous system that frequently overexpresses mitogenic peptides, chemokines and their receptors. Dipeptidyl peptidase IV (DPPIV), a cell surface serine protease, inactivates or degrades some of these bioactive peptides and chemokines, thereby regulating cell proliferation and survival. Our studies show that DPPIV is expressed in normal neural crest-derived structures, including superior cervical and dorsal root ganglion cells, sciatic nerve, and in adrenal glands, but its expression is greatly decreased or lost in cells derived from NB, their malignant counterpart. Restoration of DPPIV expression in NB cells led to their differentiation in association with increased expression of the neural marker MAP2 and decreased expression of chemokines, including stromal-derived factor 1 (SDF1) and its receptor CXCR4. Furthermore, DPPIV promoted apoptosis, and inhibited SDF1-mediated in vitro cell migration and angiogenic potential. These changes were accompanied by caspase activation and decreased levels of phospho-Akt and MMP9 activity, which are downstream effectors of SDF1-CXCR4 signaling. Importantly, DPPIV suppressed the tumorigenic potential of NB cells in a xenotransplantation mouse model. These data support a potential role for DPPIV in inhibiting NB growth and progression.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/genetics
- Apoptosis/genetics
- Caspases/genetics
- Caspases/metabolism
- Cell Differentiation/genetics
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation
- Cell Survival/genetics
- Chemokine CXCL12/biosynthesis
- Chemokine CXCL12/genetics
- Dipeptidyl Peptidase 4/biosynthesis
- Dipeptidyl Peptidase 4/genetics
- Enzyme Activation/genetics
- Gene Expression Regulation, Neoplastic/genetics
- Growth Substances/biosynthesis
- Growth Substances/genetics
- Matrix Metalloproteinase 9/biosynthesis
- Matrix Metalloproteinase 9/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microtubule-Associated Proteins/biosynthesis
- Microtubule-Associated Proteins/genetics
- Neoplasm Transplantation
- Neural Crest/enzymology
- Neural Crest/metabolism
- Neuroblastoma/enzymology
- Neuroblastoma/genetics
- Neuroblastoma/pathology
- Proto-Oncogene Proteins c-akt
- Rats
- Rats, Sprague-Dawley
- Receptors, CXCR4/biosynthesis
- Receptors, CXCR4/genetics
- Sympathetic Nervous System/enzymology
- Sympathetic Nervous System/pathology
- Transplantation, Heterologous
Collapse
Affiliation(s)
- W. Tristram Arscott
- Department of Microbiology and Molecular Genetics, Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| | - Annette E. LaBauve
- Department of Microbiology and Molecular Genetics, Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| | - Victor May
- Department of Anatomy and Neurobiology, Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| | - Umadevi V. Wesley
- Department of Microbiology and Molecular Genetics, Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| |
Collapse
|
63
|
Lo PC, Chen J, Stefflova K, Warren MS, Navab R, Bandarchi B, Mullins S, Tsao M, Cheng JD, Zheng G. Photodynamic molecular beacon triggered by fibroblast activation protein on cancer-associated fibroblasts for diagnosis and treatment of epithelial cancers. J Med Chem 2009; 52:358-68. [PMID: 19093877 PMCID: PMC2773291 DOI: 10.1021/jm801052f] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fibroblast activation protein (FAP) is a cell-surface serine protease highly expressed on cancer-associated fibroblasts of human epithelial carcinomas but not on normal fibroblasts, normal tissues, and cancer cells. We report herein a novel FAP-triggered photodynamic molecular beacon (FAP-PPB) comprising a fluorescent photosensitizer and a black hole quencher 3 linked by a peptide sequence (TSGPNQEQK) specific to FAP. FAP-PPB was effectively cleaved by both human FAP and murine FAP. By use of the HEK293 transfected cells (HEK-mFAP, FAP(+); HEK-vector, FAP(-)), systematic in vitro and in vivo experiments validated the FAP-specific activation of FAP-PPB in cancer cells and mouse xenografts, respectively. FAP-PPB was cleaved by FAP, allowing fluorescence restoration in FAP-expressing cells while leaving non-expressing FAP cells undetectable. Moreover, FAP-PPB showed FAP-specific photocytotoxicity toward HEK-mFAP cells whereas it was non-cytotoxic toward HEK-Vector cells. This study suggests that the FAP-PPB is a potentially useful tool for epithelial cancer detection and treatment.
Collapse
Affiliation(s)
- Pui-Chi Lo
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Division of Biophysics and Bioimaging, Ontario Cancer Institute, Toronto, ON, M5G 1L7, Canada
| | - Juan Chen
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Division of Biophysics and Bioimaging, Ontario Cancer Institute, Toronto, ON, M5G 1L7, Canada
| | - Klara Stefflova
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael S. Warren
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roya Navab
- Division of Applied Molecular Oncology, Ontario Cancer Institute, Toronto, ON, M5G 1L7, Canada
| | - Bizhan Bandarchi
- Division of Applied Molecular Oncology, Ontario Cancer Institute, Toronto, ON, M5G 1L7, Canada
| | - Stefanie Mullins
- Fox Chase Cancer Center, Department of Medical Oncology, Philadelphia, Pennsylvania 19111, USA
| | - Ming Tsao
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Division of Applied Molecular Oncology, Ontario Cancer Institute, Toronto, ON, M5G 1L7, Canada
| | - Jonathan D. Cheng
- Fox Chase Cancer Center, Department of Medical Oncology, Philadelphia, Pennsylvania 19111, USA
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Division of Biophysics and Bioimaging, Ontario Cancer Institute, Toronto, ON, M5G 1L7, Canada
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
64
|
Organotypic modelling as a means of investigating epithelial-stromal interactions during tumourigenesis. FIBROGENESIS & TISSUE REPAIR 2008; 1:8. [PMID: 19077226 PMCID: PMC2614933 DOI: 10.1186/1755-1536-1-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 12/11/2008] [Indexed: 11/10/2022]
Abstract
The advent of co-culture approaches has allowed researchers to more accurately model the behaviour of epithelial cells in cell culture studies. The initial work on epidermal modelling allowed the development of reconstituted epidermis, growing keratinocytes on top of fibroblasts seeded in a collagen gel at an air-liquid interface to generate terminally differentiated 'skin equivalents'. In addition to developing ex vivo skin sheets for the treatment of burns victims, such cultures have also been used as a means of investigating both the development and repair of the epidermis, in more relevant conditions than simple two-dimensional culture, but without the use of animals. More recently, by varying the cell types used and adjusting the composition of the matrix components, this physiological system can be adapted to allow the study of interactions between tumour cells and their surrounding stroma, particularly with regards to how such interactions regulate invasion. Here we provide a summary of the major themes involved in tumour progression and consider the evolution of the approaches used to study cancer cell behaviour. Finally, we review how organotypic models have facilitated the study of several key pathways in cancer development and invasion, and speculate on the exciting future roles for these models in cancer research.
Collapse
|
65
|
Wu YM, Tang J, Zhao P, Chen ZN, Jiang JL. Enhanced expression of Hab18g/CD147 and activation of integrin pathway in HCC cells under 3-D co-culture conditions. Cell Biol Int 2008; 33:199-206. [PMID: 19059491 DOI: 10.1016/j.cellbi.2008.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 06/04/2008] [Accepted: 11/12/2008] [Indexed: 11/30/2022]
Abstract
CD147 is reported to be correlated with the malignancy of some cancers, and its overexpression affects the progression of tumor. In the present study, we investigated the function of HAb18G/CD147, a member of CD147 family, on hepatocellular carcinoma (HCC) adhesion, invasion and metastasis in 3-dimensional (3-D) cell co-culture model. The results showed that the extracellular microenvironment could determine the cellular phenotypes and then affected the cellular functions. The expressions of HAb18G/CD147 in HCC cells and fibroblasts were both obviously elevated in 3-D co-culture model. The overexpression of HAb18G/CD147 increased MMPs' (MMP-2 and MMP-9) production (P < 0.01), and was obviously accompanied with enhanced expressions of paxillin, FAK and p-FAK in 3-D cell co-culture model. All the results suggest that HAb18G/CD147 plays an important role in HCC adhesion, invasion and metastasis mainly via modulating synthesis of MMPs and activating integrin signal pathways in fibroblasts and tumor cells themselves under the 3-D co-culture conditions.
Collapse
Affiliation(s)
- Ya-Mei Wu
- Cell Engineering Research Centre & Department of Cell Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, 17 West Changle Road, Xi'an 710032, People's Republic of China
| | | | | | | | | |
Collapse
|
66
|
Zhuo S, Chen J, Yu B, Jiang X, Luo T, Liu Q, Chen R, Xie S. Nonlinear optical microscopy of the bronchus. JOURNAL OF BIOMEDICAL OPTICS 2008; 13:054024. [PMID: 19021404 DOI: 10.1117/1.2982534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Because of frequent exposure to carcinogens, the bronchus is prone to early pathologic alterations. The assessment of these early changes is of key significance in physiological studies and disease diagnosis of the bronchus. We utilize nonlinear optical microscopy (NLOM) to image mouse bronchial tissue based on intrinsic nonlinear optical contrast. Our results show that NLOM is effective for imaging the bronchial intact microstructural components, providing quantitative information about the biomorphology and biochemistry of tissue. Our findings also display that NLOM can provide a two-photon ratiometric redox fluorometry, based on mitochondrial signals and reduced pyridine nucleotide (NADH and NADPH) and oxidized flavoproteins (Fp) signals, to assess the metabolic state of the epithelial cells and chondrocytes. It was found that NLOM can offer a sensitive tool, based on the second-harmonic signal depth-dependent decay, to obtain quantitative information on the optical property of the stroma associated with normal and diseased tissue states. Our results suggest that with the advent of the clinical portability of typical nonlinear optical endoscopy, the NLOM technique has the potential to be applied in vivo to the clinical diagnosis and monitoring of bronchial disease.
Collapse
Affiliation(s)
- Shuangmu Zhuo
- Fujian Normal University, Institute of Laser and Optoelectronics Technology, Fujian Provincial Key Laboratory for Photonics Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fuzhou 350007, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Daly AJ, McIlreavey L, Irwin CR. Regulation of HGF and SDF-1 expression by oral fibroblasts – Implications for invasion of oral cancer. Oral Oncol 2008; 44:646-51. [DOI: 10.1016/j.oraloncology.2007.08.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 07/30/2007] [Accepted: 08/20/2007] [Indexed: 01/02/2023]
|
68
|
Cathepsin B mediates the pH-dependent proinvasive activity of tumor-shed microvesicles. Neoplasia 2008; 10:481-8. [PMID: 18472965 DOI: 10.1593/neo.08178] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 02/14/2008] [Accepted: 02/15/2008] [Indexed: 12/11/2022] Open
Abstract
Vesicles shed by cancer cells are known to mediate several tumor-host interactions. Tumor microenvironment may, in turn, influence the release and the activity of tumor-shed microvesicles. In this study, we investigated the molecular mediators of the pH-dependent proinvasive activity of tumor-shed vesicles. Gelatinase zymography showed increased microvesicle activity of matrix metalloproteinases 9 and 2 as a result of acid exposure (pH 5.6) compared to pH 7.4. Thus, we reasoned that the cysteine protease cathepsin B might play a role in mediating the pH-dependent activation of gelatinases. Cathepsin B expression in tumor-shed microvesicles was confirmed by Western blot analysis and zymography. The activity of vesicle-associated cathepsin B measured using Z-Arg-Arg-pNA as substrate was significantly increased at acidic pH values. Inhibition of protease activity by the cysteine protease inhibitor, E-64, and treatment of ovarian cancer cells with small interfering RNA against cathepsin B suppressed the ability of tumor-shed microvesicles to stimulate both gelatinase activation and the invasiveness of endothelial cells observed at low pH values. We conclude that microvesicle shedding is a major secretory pathway for cathepsin B release from tumor cells. Hence, the acidic microenvironment found in most solid tumors may contribute to cathepsin B-mediated proinvasive capabilities of tumor-shed vesicles.
Collapse
|
69
|
Zeng Y, Lee TS, Yu P, Low HT. Numerical Simulation of Mass Transport in a Microchannel Bioreactor With Cell Micropatterning. J Biomech Eng 2008; 130:031018. [DOI: 10.1115/1.2913231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Micropatterning of two different cell types based on surface modification allows spatial control over two distinct cell subpopulations. This study considers a micropatterned coculture system, which has release and absorption parts alternately arranged at the base, and each part has a single cell type. A micropattern unit was defined and within each unit, there are one release part and one absorption part. The cells in the absorption parts consume species, which are secreted by the cells in the release parts. The species concentrations at the micropatterned cell base were computed from a three-dimensional numerical flow model incorporating mass transport. Different combined parameters were developed for the release and absorption parts to make the data collapse in each part. Combination of the collapse data in the release and absorption parts can be used to predict the concentration distribution through the whole channel. The correlated results were applied to predict the critical length ratio of the release and absorption parts for an actual micropatterned system (Bhatia et al., 1999, “Effect of Cell-Cell Interactions in Preservation of Cellular Phenotype: Co-Cultivation of Hepatocytes and Nonparenchymal Cell,” FASEB J. 13, pp. 1883–1900) to avoid species insufficiency based on basic fibroblast growth factor (bFGF). The mass transfer effectiveness was found to be higher with more numbers of micropattern units. The optimal condition for micropatterned coculture bioreactors is achieved by having the product of the length ratio and the reaction ratio equal to 1. This condition was used to optimize the mass transfer in the micropatterned system (Bhatia et al., 1999, “Effect of Cell-Cell Interactions in Preservation of Cellular henotype: Co-Cultivation of Hepatocytes and Nonparenchymal Cell,” FASEB J. 13, pp. 1883–1900) based on bFGF.
Collapse
Affiliation(s)
- Yan Zeng
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Thong-See Lee
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Peng Yu
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Hong-Tong Low
- Division of Bioengineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
70
|
Pavlova I, Williams M, El-Naggar A, Richards-Kortum R, Gillenwater A. Understanding the biological basis of autofluorescence imaging for oral cancer detection: high-resolution fluorescence microscopy in viable tissue. Clin Cancer Res 2008; 14:2396-404. [PMID: 18413830 PMCID: PMC2773159 DOI: 10.1158/1078-0432.ccr-07-1609] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE Autofluorescence imaging is increasingly used to noninvasively identify neoplastic oral cavity lesions. Improving the diagnostic accuracy of these techniques requires a better understanding of the biological basis for optical changes associated with neoplastic transformation in oral tissue. EXPERIMENTAL DESIGN A total of 49 oral biopsies were considered in this study. The autofluorescence patterns of viable normal, benign, and neoplastic oral tissue were imaged using high-resolution confocal fluorescence microscopy. RESULTS The autofluorescence properties of oral tissue vary significantly based on anatomic site and pathologic diagnosis. In normal oral tissue, most of the epithelial autofluorescence originates from the cytoplasm of cells in the basal and intermediate regions, whereas structural fibers are responsible for most of the stromal fluorescence. A strongly fluorescent superficial layer was observed in tissues from the palate and the gingiva, which contrasts with the weakly fluorescent superficial layer found in other oral sites. Upon UV excitation, benign inflammation shows decreased epithelial fluorescence, whereas dysplasia displays increased epithelial fluorescence compared with normal oral tissue. Stromal fluorescence in both benign inflammation and dysplasia drops significantly at UV and 488 nm excitation. CONCLUSION Imaging oral lesions with optical devices/probes that sample mostly stromal fluorescence may result in a similar loss of fluorescence intensity and may fail to distinguish benign from precancerous lesions. Improved diagnostic accuracy may be achieved by designing optical probes/devices that distinguish epithelial fluorescence from stromal fluorescence and by using excitation wavelengths in the UV range.
Collapse
Affiliation(s)
- Ina Pavlova
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | | | - Adel El-Naggar
- Pathology, The University of Texas M. D. Anderson Cancer Center
| | | | - Ann Gillenwater
- Head and Neck Surgery, The University of Texas M. D. Anderson Cancer Center
| |
Collapse
|
71
|
Abstract
The preferential proliferation of cancer cells in the bone microenvironment is poorly characterised. Expression pattern of bone marrow and other organ microenvironment in contact with osteolytic (Walker W256) and osteoblastic (MatLyLu MLL) metastases were investigated. Fisher and Copenhagen rats received, respectively, W256 and MLL cells injection. Bone and soft tissues were analysed by immunochemistry for DKK1, cathepsin K, RANKL, MCSF or IL6 expression. Tartrate-resistant acid phosphatase (TRAcP)-positive cells were detected by a histoenzymatic technique. In bone, expressions of MCSF and DKK1 were shown in stromal cells of the bone marrow, in contact with metastatic foci of both tumours. Many stromal cells were found RANKL positive in the vicinity of the tumours. Cells expressing cathepsin K and multinucleated TRAcP+ cells were found in direct contact with trabeculae but also in bone marrow spaces near metastatic cells. In extraosseous tumours, cells in contact with malignant cells did not expressed DKK1, MCSF, cathepsin K and IL6. Some RANKL+ cells were found in the periphery of subcutaneous tumours but may represent Langerhans cells. Abnormal presence of TRAcP+ cells was never observed in the vicinity of malignant cells. Interaction between stromal and cancer cells induces the expression on the formers of characteristics leading to osteoclastogenesis only in the bone microenvironment.
Collapse
|
72
|
Chabottaux V, Noel A. Breast cancer progression: insights into multifaceted matrix metalloproteinases. Clin Exp Metastasis 2007; 24:647-56. [PMID: 17968664 DOI: 10.1007/s10585-007-9113-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 10/10/2007] [Indexed: 12/14/2022]
Abstract
The restricted view of matrix metalloproteinases (MMPs) as simple destroyers of extracellular matrix components has largely ignored their substantial contribution in many aspects of cancer development and metastatic dissemination. Over the last few years, the relevance of MMPs in the processing of a large array of extracellular and cell surface-associated proteins has grown considerably. Our knowledge about the complex functions of MMPs and how their contribution may differ throughout cancer progression is rapidly expanding. These new findings provide several explanations for the lack of success of MMP inhibition in clinical trials. A complete understanding of MMP biology is needed before considering them, their substrates or their products as therapeutic targets. In this review, we explore the different faces of MMP implication in breast cancer progression by considering both clinical and fundamental aspects.
Collapse
Affiliation(s)
- Vincent Chabottaux
- Laboratory of Tumor and Developmental Biology, Center for Experimental Cancer Research (CRCE), Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA-Research), University of Liege, Tour de Pathologie (B23), Sart-Tilman, Liege, 4000, Belgium
| | | |
Collapse
|
73
|
Smith RA, Lea RA, Weinstein SR, Griffiths LR. Progesterone, glucocorticoid, but not estrogen receptor mRNA is altered in breast cancer stroma. Cancer Lett 2007; 255:77-84. [PMID: 17512111 DOI: 10.1016/j.canlet.2007.03.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 03/26/2007] [Accepted: 03/26/2007] [Indexed: 01/16/2023]
Abstract
Our laboratory has previously found that anti-mitogenic nuclear receptor mRNA is elevated in late stage tumours and this study was performed to scrutinize the possibility of cancer-stroma crosstalk using hormone signaling in these tissues. RNA levels in stromal tissue were examined for the estrogen alpha, estrogen beta, androgen, progesterone and glucocorticoid nuclear receptors by a semi-quantitative PCR. Significant differences in expression between the cancer stroma and control tissue were seen, analyzing for both cancer grade and estrogen receptor status. Stroma and control tissue were significantly different for the progesterone and glucocorticoid nuclear receptors (p=5.908 x 10(-7) and 2.761 x 10(-5), respectively). Glucocorticoid receptor also showed a significant increase to mRNA levels in the stroma of estrogen receptor negative tumours (p=5.85 x 10(-5)). By contrast, the estrogen receptors alpha and beta, those most closely associated with breast tissue growth, showed no significant change in mRNA (p=0.372 and 0.655, respectively). Androgen receptor mRNA also remained unaffected (p=0.174).
Collapse
Affiliation(s)
- Robert A Smith
- Genomics Research Centre and Wesley Research Institute, School of Health Science, Griffith University Gold Coast, QLD, Australia
| | | | | | | |
Collapse
|
74
|
Arifler D, Pavlova I, Gillenwater A, Richards-Kortum R. Light scattering from collagen fiber networks: micro-optical properties of normal and neoplastic stroma. Biophys J 2007; 92:3260-74. [PMID: 17307834 PMCID: PMC1852360 DOI: 10.1529/biophysj.106.089839] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Accepted: 01/17/2007] [Indexed: 11/18/2022] Open
Abstract
Development of epithelial precancer and cancer leads to well-documented molecular and structural changes in the epithelium. Recently, it has been recognized that stromal biology is also altered significantly with preinvasive disease. We used the finite-difference time-domain method, a popular technique in computational electromagnetics, to model light scattering from heterogeneous collagen fiber networks and to analyze how neoplastic changes alter stromal scattering properties. Three-dimensional optical images from the stroma of fresh normal and neoplastic oral-cavity biopsies were acquired using fluorescence confocal microscopy. These optical sections were then processed to create realistic three-dimensional collagen networks as model input. Image analysis revealed that the volume fraction of collagen fibers in the stroma decreases with precancer and cancer progression, and fibers tend to be shorter and more disconnected in neoplastic stroma. The finite-difference time-domain modeling results showed that neoplastic fiber networks have smaller scattering cross sections compared to normal networks. Computed scattering-phase functions indicate that high-angle scattering probabilities tend to be higher for neoplastic networks. These results provide valuable insight into the micro-optical properties of normal and neoplastic stroma. Characterization of optical signals obtained from epithelial tissues can aid in development of optical spectroscopic and imaging techniques for noninvasive monitoring of early neoplastic changes.
Collapse
Affiliation(s)
- Dizem Arifler
- Department of Physics, Eastern Mediterranean University, Famagusta, Cyprus
| | | | | | | |
Collapse
|
75
|
Abstract
The development and function of living tissues depends largely on interactions between cells that can vary in both time and space; however, temporal control of cell-cell interaction is experimentally challenging. By using a micromachined silicon substrate with moving parts, we demonstrate the dynamic regulation of cell-cell interactions via direct manipulation of adherent cells with micrometer-scale precision. We thereby achieve mechanical control of both tissue composition and spatial organization. As a case study, we demonstrate the utility of this tool in deconstructing the dynamics of intercellular communication between hepatocytes and supportive stromal cells in coculture. Our findings indicate that the maintenance of the hepatocellular phenotype by stroma requires direct contact for a limited time ( approximately hours) followed by a sustained soluble signal that has an effective range of <400 microm. This platform enables investigation of dynamic cell-cell interaction in a multitude of applications, spanning embryogenesis, homeostasis, and pathogenic processes.
Collapse
Affiliation(s)
- Elliot E. Hui
- *Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology/Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139; and
| | - Sangeeta N. Bhatia
- *Department of Bioengineering, University of California at San Diego, La Jolla, CA 92093
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology/Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139; and
- Division of Medicine, Brigham and Women's Hospital, Boston, MA 02115
| |
Collapse
|
76
|
Salvi A, Arici B, Alghisi A, Barlati S, De Petro G. RNA interference against urokinase in hepatocellular carcinoma xenografts in nude mice. Tumour Biol 2006; 28:16-26. [PMID: 17159381 DOI: 10.1159/000097699] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Accepted: 05/16/2006] [Indexed: 11/19/2022] Open
Abstract
The serine protease urokinase-type plasminogen activator (u-PA) is overexpressed in hepatocellular carcinoma (HCC) and its expression level is inversely correlated with the patients' survival. The purpose of this study was to examine the effects of vector-based RNA interference (RNAi) of u-PA on the growth of human HCC xenografts in nude mice in order to investigate the role of u-PA in human HCC. Our results showed that the subcutaneous injection of small interfering RNAs (siRNA) u-PA SKHep1C3 stable transfected cells (pS siRNA u-PA) led to a growth delay in xenograft development, compared to those generated from empty vector; the molecular characterization of nodules (carried out by PCR, RT-PCR and immunohistochemical analysis) revealed the presence of plasmid DNA, the u-PA gene expression knockdown, at both mRNA and protein levels, giving evidence of a long-term and target-specific inhibition by vector-based RNAi 11 weeks after cell inoculation. We further studied the effects of u-PA down modulation on extracellular matrix (ECM) proteins evaluating the expression and organization of fibronectin (FN; one of the main ECM proteins). Immunohistochemical and immunofluorescence analysis of FN revealed FN fibrils in pS siRNA u-PA xenografts and in pS siRNA u-PA cells, thus identifying the FN fibril organization as a downstream effect of u-PA knockdown in this system.
Collapse
Affiliation(s)
- A Salvi
- Biology and Genetics Division, Department of Biomedical Sciences and Biotechnology, IDET Centre of Excellence, University of Brescia, Brescia, Italy
| | | | | | | | | |
Collapse
|
77
|
Khwaja FW, Svoboda P, Reed M, Pohl J, Pyrzynska B, Van Meir EG. Proteomic identification of the wt-p53-regulated tumor cell secretome. Oncogene 2006; 25:7650-61. [PMID: 17043663 DOI: 10.1038/sj.onc.1209969] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumor-stroma interactions play a major role in tumor development, maintenance and progression. Yet little is known on how the genetic alterations that underlie cell transformation elicit cell extrinsic changes modulating heterotypic cell interactions. We hypothesized that these events involve a modification in the complement of secreted proteins by the cell, acting as mediators of intercellular communication. To test this hypothesis, we examined the role of wt-p53, a major tumor suppressor, on the tumor microenvironment through its regulation of secreted factors. Using a combination of 2-DE and cICAT proteomic techniques, we found a total of 111 secreted proteins, 39 of which showed enhanced and 21 inhibited secretion in response to wt-p53 expression. The majority of these were not direct targets of p53 transcription factor activity, suggesting a novel role for wt-p53 in the control of intracellular protein trafficking and/or secreted protein stability. Evidence for p53-controlled post-translational modifications on nine secreted proteins was also found. These findings will enhance our understanding of wt-p53 modulated interactions of the tumor with its environment.
Collapse
Affiliation(s)
- F W Khwaja
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
78
|
Cat B, Stuhlmann D, Steinbrenner H, Alili L, Holtkötter O, Sies H, Brenneisen P. Enhancement of tumor invasion depends on transdifferentiation of skin fibroblasts mediated by reactive oxygen species. J Cell Sci 2006; 119:2727-38. [PMID: 16757516 DOI: 10.1242/jcs.03011] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myofibroblasts, pivotal for tumor progression, populate the microecosystem of reactive stroma. Using an in vitro tumor-stroma model of skin carcinogenesis, we report here that tumor-cell-derived transforming growth factor β1 (TGFβ1) initiates reactive oxygen species-dependent expression of α-smooth muscle actin, a biomarker for myofibroblastic cells belonging to a group of late-responsive genes. Moreover, protein kinase C (PKC) is involved in lipid hydroperoxide-triggered molecular events underlying transdifferentiation of fibroblasts to myofibroblasts (mesenchymal-mesenchymal transition, MMT). In contrast to fibroblasts, myofibroblasts secrete large amounts of hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6), resulting in a significant increase in the invasive capacity of tumor cells. The thiol N-acetyl-L-cysteine, the micronutrient selenite as well as selenoprotein P and the lipid peroxidation inhibitors α-tocopherol and butylated hydroxytoluene significantly lower both the number of TGFβ1-initiated myofibroblasts and the secretion of HGF, VEGF and IL-6, correlating with a diminished invasive capacity of tumor cells. This novel concept of stromal therapy, namely the protection of stromal cells against the dominating influence of tumor cells in tumor-stroma interaction by antioxidants and micronutrients, may form the basis for prevention of MMT in strategies for chemoprevention of tumor invasion.
Collapse
Affiliation(s)
- Bahar Cat
- Institute for Biochemistry and Molecular Biology I, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
79
|
Babbin BA, Lee WY, Parkos CA, Winfree LM, Akyildiz A, Perretti M, Nusrat A. Annexin I regulates SKCO-15 cell invasion by signaling through formyl peptide receptors. J Biol Chem 2006; 281:19588-99. [PMID: 16675446 DOI: 10.1074/jbc.m513025200] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Annexin 1 (AnxA1) is a multifunctional phospholipid-binding protein associated with the development of metastasis in some invasive epithelial malignancies. However, the role of AnxA1 in the migration/invasion of epithelial cells is not known. In this study, experiments were performed to investigate the role of AnxA1 in the invasion of a model epithelial cell line, SKCO-15, derived from colorectal adenocarcinoma. Small interfering RNA-mediated knockdown of AnxA1 expression resulted in a significant reduction in invasion through Matrigel-coated filters. Localization studies revealed a translocation of AnxA1 to the cell surface upon the induction of cell migration, and functional inhibition of cell surface AnxA1 using antiserum (LCO1) significantly reduced cell invasion. Conversely, SKCO-15 cell invasion was increased by approximately 2-fold in the presence of recombinant full-length AnxA1 and the AnxA1 N-terminal-derived peptide mimetic, Ac2-26. Because extracellular AnxA1 has been shown to regulate leukocyte migratory events through interactions with n-formyl peptide receptors (nFPRs), we examined the expression of FPR-1, FPRL-1, and FPRL-2 in SKCO-15 cells by reverse transcriptase-PCR and identified expression of all three receptors in this cell line. Treatment of SKCO-15 cells with AnxA1, Ac2-26, and the classical nFPR agonist, formylmethionylleucylphenylalanine, induced intracellular calcium release consistent with nFPR activation. Furthermore, the nFPR antagonist, Boc2, abrogated the AnxA1 and Ac2-26-induced intracellular calcium release and increase in SKCO-15 cell invasion. Together, these results support an autocrine/paracrine role for membrane AnxA1 in stimulating SKCO-15 cell migration through nFPR activation. The findings in this study suggest that activation of nFPRs stimulates epithelial cell motility important in the development of metastasis as well as wound healing.
Collapse
Affiliation(s)
- Brian A Babbin
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
The search for an MMP inhibitor with anticancer efficacy is a nearly three-decade endeavor. This inhibitor is yet to be found. The reasons for this failure include shortcomings in the chemistry of these compounds (including broad MMP sub-type selectivity, metabolic lability, and toxicity) as well as the emerging, and arguably extraordinary, complexity of MMP cell (and cancer) biology. Together these suggest that the successful anticancer inhibitor must possess MMP selectivity against the MMP subtype whose involvement is critical, yet highly temporally (with respect to metastatic progression) and mechanistically (with respect to matrix degradation) regulated. This review summarizes the progression of chemical structure and mechanistic thinking toward these objectives, with emphasis on the disappointment, the perseverance, and the resilient optimism that such an inhibitor is there to be discovered.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556-5670, USA
| | | |
Collapse
|
81
|
Anderegg U, Breitschwerdt K, Köhler MJ, Sticherling M, Haustein UF, Simon JC, Saalbach A. MEL4B3, a novel mRNA is induced in skin tumors and regulated by TGF-beta and pro-inflammatory cytokines. Exp Dermatol 2005; 14:709-18. [PMID: 16098131 DOI: 10.1111/j.0906-6705.2005.00349.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tumor-stroma interactions play a decisive role in the growth and metastasis of solid tumors, and involve signalling either by soluble mediators or direct cell-cell interaction. Here, we report the isolation and characterisation of a novel cDNA (MEL4B3), which is induced in cultured dermal fibroblasts exposed to supernatants of melanoma cell lines. MEL4B3 shares high homology with two predicted cDNA sequences for which no activity has so far been described. In situ hybridisation revealed the expression of MEL4B3 in malignant melanoma increasing with tumor depth; in basal cell carcinoma and in squamous cell carcinoma. MEL4B3 was barely detectable in normal skin or non-malignant melanocytic naevi. Furthermore, MEL4B3 was expressed at high level in the epidermis of psoriatic skin. In vitro, the expression of MEL4B3 was found to be induced by the exposure of human dermal fibroblasts to melanoma cell culture supernatants or to transforming growth factor-beta, interleukin-1 and tumor necrosis factor-alpha. The expression MEL4B3 therefore reflects closely cell activation occurring during tumor growth, metastasis and inflammation.
Collapse
Affiliation(s)
- Ulf Anderegg
- Saxon Academy of Science in Leipzig, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | |
Collapse
|