151
|
Boudreau A, van't Veer LJ, Bissell MJ. An "elite hacker": breast tumors exploit the normal microenvironment program to instruct their progression and biological diversity. Cell Adh Migr 2012; 6:236-48. [PMID: 22863741 DOI: 10.4161/cam.20880] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The year 2011 marked the 40 year anniversary of Richard Nixon signing the National Cancer Act, thus declaring the beginning of the "War on Cancer" in the United States. Whereas we have made tremendous progress toward understanding the genetics of tumors in the past four decades, and in developing enabling technology to dissect the molecular underpinnings of cancer at unprecedented resolution, it is only recently that the important role of the stromal microenvironment has been studied in detail. Cancer is a tissue-specific disease, and it is becoming clear that much of what we know about breast cancer progression parallels the biology of the normal breast differentiation, of which there is still much to learn. In particular, the normal breast and breast tumors share molecular, cellular, systemic and microenvironmental influences necessary for their progression. It is therefore enticing to consider a tumor to be a "rogue hacker"--one who exploits the weaknesses of a normal program for personal benefit. Understanding normal mammary gland biology and its "security vulnerabilities" may thus leave us better equipped to target breast cancer. In this review, we will provide a brief overview of the heterotypic cellular and molecular interactions within the microenvironment of the developing mammary gland that are necessary for functional differentiation, provide evidence suggesting that similar biology--albeit imbalanced and exaggerated--is observed in breast cancer progression particularly during the transition from carcinoma in situ to invasive disease. Lastly we will present evidence suggesting that the multigene signatures currently used to model cancer heterogeneity and clinical outcome largely reflect signaling from a heterogeneous microenvironment-a recurring theme that could potentially be exploited therapeutically.
Collapse
Affiliation(s)
- Aaron Boudreau
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | | | | |
Collapse
|
152
|
|
153
|
Tasnim F, Zink D. Cross talk between primary human renal tubular cells and endothelial cells in cocultures. Am J Physiol Renal Physiol 2012; 302:F1055-62. [DOI: 10.1152/ajprenal.00621.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interactions between renal tubular epithelial cells and adjacent endothelial cells are essential for normal renal functions but also play important roles in renal disease and repair. Here, we investigated cocultures of human primary renal proximal tubular cells (HPTC) and human primary endothelial cells to address the cross talk between these cell types. HPTC showed improved proliferation, marker gene expression, and enzyme activity in cocultures. Also, the long-term maintenance of epithelia formed by HPTC was improved, which was due to the secretion of transforming growth factor-β1 and its antagonist α2-macroglobulin. HPTC induced endothelial cells to secrete increased amounts of these factors, which balanced each other functionally and only displayed in combination the observed positive effects. In addition, in the presence of HPTC endothelial cells expressed increased amounts of hepatocyte growth factor and vascular endothelial growth factor, which have well-characterized effects on renal tubular epithelial cells as well as on endothelial cells. Together, the results showed that HPTC stimulated endothelial cells to express a functionally balanced combination of various factors, which in turn improved the performance of HPTC. The results give new insights into the cross talk between renal epithelial and endothelial cells and suggest that cocultures could be also useful models for the analysis of cellular communication in renal disease and repair. Furthermore, the characterization of defined microenvironments, which positively affect HPTC, will be helpful for improving the performance of this cell type in in vitro applications including in vitro toxicology and kidney tissue engineering.
Collapse
Affiliation(s)
- Farah Tasnim
- Institute of Bioengineering and Nanotechnology, The Nanos, Singapore
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, The Nanos, Singapore
| |
Collapse
|
154
|
Tsai SY, Huang YL, Yang WH, Tang CH. Hepatocyte growth factor-induced BMP-2 expression is mediated by c-Met receptor, FAK, JNK, Runx2, and p300 pathways in human osteoblasts. Int Immunopharmacol 2012; 13:156-62. [PMID: 22504529 DOI: 10.1016/j.intimp.2012.03.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 03/16/2012] [Accepted: 03/29/2012] [Indexed: 12/24/2022]
Abstract
Hepatocyte growth factor (HGF) has been demonstrated to stimulate osteoblast proliferation and participated bone remodeling. Bone morphogenetic protein-2 (BMP-2) is a crucial mediator in bone formation during fracture healing. However, the effects of HGF in BMP-2 expression in human osteoblasts are large unknown. Here we found that HGF induced BMP-2 expression in human osteoblasts dose-dependently. HGF-mediated BMP-2 production was attenuated by c-Met inhibitor or siRNA. Pretreatment with FAK inhibitor or JNK inhibitor (SP600125) also blocked the potentiating action of HGF. Stimulation of osteoblasts with HGF enhanced FAK phosphorylation, JNK phosphorylation, and RunX2 translocation from cytosol to the nucleus. HGF-mediated Runx2 binding to BMP-2 promoter was inhibited by c-Met inhibitor, FAK inhibitor, and SP600125. The binding of Runx2 to the BMP-2 promoter, as well as the recruitment of p300 and the enhancement of histones H3 and H4 acetylation on the BMP-2 promoter was enhanced by HGF. Our results suggest that HGF increased BMP-2 production in human osteoblasts via the c-Met receptor/FAK/JNK/Runx2 and p300 signaling pathways.
Collapse
Affiliation(s)
- Shu-Yao Tsai
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | | | | | | |
Collapse
|
155
|
Ozeki M, Tabata Y. Affinity evaluation of gelatin for hepatocyte growth factor of different types to design the release carrier. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 17:139-50. [PMID: 16411604 DOI: 10.1163/156856206774879027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The objective of this study was to investigate the physicochemical interaction of hepatocyte growth factor (HGF) and its variant with 5 amino-acid residues deleted (dHGF) with an acidic gelatin for the design of factors release from the gelatin hydrogel. When the interaction of HGF or dHGF with gelatin-immobilized agarose beads was evaluated by Scatchard binding assay, the dissociation constant of dHGF was higher than that of HGF, although the two proteins had a similar binding ratio. dHGF was released more rapidly from the hydrogel of acidic gelatin than HGF. In vivo release study with 125I-labeled HGF or dHGF in mice subcutis showed that HGF was released from the gelatin hydrogel as a result of hydrogel degradation. In contrast, dHGF was rapidly released by a simple diffusion from the gelatin hydrogel. From electrophoresis experiments, mixing with the acidic gelatin enabled HGF to complex and suppressing the trypsin-digested molecular weight loss, in marked contrast to that of dHGF. In addition, the percentage of HGF recognized by the antibody was reduced by the gelatin complexation, but that of dHGF was not. We conclude that unlike dHGF, HGF has a strong affinity for the acidic gelatin, resulting in the controlled release of HGF accompanied with hydrogel degradation of the release carrier.
Collapse
Affiliation(s)
- Makoto Ozeki
- Institute for Frontier Medical Sciences, Kyoto University, Sakyo-ku, Japan
| | | |
Collapse
|
156
|
Ozeki M, Tabata Y. Interaction of hepatocyte growth factor with gelatin as the carrier material. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 17:163-75. [PMID: 16411606 DOI: 10.1163/156856206774879162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The objective of this study was to physicochemically investigate the interaction between hepatocyte growth factor (HGF) and acidic gelatin compared with that between HGF and basic gelatin or heparin. Gelatin- or heparin-immobilized agarose beads were prepared and HGF interaction with them was evaluated by Scatchard binding assay. The dissociation constant of HGF with the acidic gelatin was about 2-3 orders of magnitude higher than that of heparin. The cell proliferation assay revealed that the proliferation promotion activity of HGF complexed with the acidic gelatin was detected, although it was lower than that of original HGF. The ability of HGF to enhance the cell proliferation was reduced by the trypsin treatment, although the extent of the reduction was significantly suppressed by HGF complexation with acidic gelatin. Electrophoresis experimentally confirmed enhanced resistance to the molecular mass loss of HGF by gelatin complexation. Moreover, the recognized level of an antibody to HGF was reduced by the complexation with the acidic gelatin, indicating that the acidic gelatin is present around HGF molecules. It is possible that the HGF molecule is covered with the acidic gelatin, resulting in protection from enzymatic digestion.
Collapse
Affiliation(s)
- Makoto Ozeki
- Institute of Frontier Medical Sciences, Kyoto University, Shogoin, Sakyo-ku, Japan
| | | |
Collapse
|
157
|
Wang W, Li Q, Takeuchi S, Yamada T, Koizumi H, Nakamura T, Matsumoto K, Mukaida N, Nishioka Y, Sone S, Nakagawa T, Uenaka T, Yano S. Met kinase inhibitor E7050 reverses three different mechanisms of hepatocyte growth factor-induced tyrosine kinase inhibitor resistance in EGFR mutant lung cancer. Clin Cancer Res 2012; 18:1663-1671. [PMID: 22317763 DOI: 10.1158/1078-0432.ccr-11-1171] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Hepatocyte growth factor (HGF) induces resistance to reversible and irreversible epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) in EGFR mutant lung cancer cells by activating Met and the downstream phosphoinositide 3-kinase (PI3K)/Akt pathway. Moreover, continuous exposure to HGF accelerates the emergence of EGFR-TKI-resistant clones. We assayed whether a new Met kinase inhibitor, E7050, which is currently being evaluated in clinical trials, could overcome these three mechanisms of resistance to EGFR-TKIs. EXPERIMENTAL DESIGN The effects of E7050 on HGF-induced resistance to reversible (gefitinib), irreversible (BIBW2992), and mutant-selective (WZ4002) EGFR-TKIs were determined using the EGFR mutant human lung cancer cell lines PC-9 and HCC827 with an exon 19 deletion and H1975 with an T790M secondary mutation. PC-9 cells were mixed with HGF-producing fibroblasts, MRC-5 cells, and subcutaneously inoculated into severe combined immunodeficient mice, and the therapeutic effects of E7050 plus gefitinib were assayed. RESULTS E7050 circumvented resistance to all of the reversible, irreversible, and mutant-selective EGFR-TKIs induced by exogenous and/or endogenous HGF in EGFR mutant lung cancer cell lines, by blocking the Met/Gab1/PI3K/Akt pathway in vitro. E7050 also prevented the emergence of gefitinib-resistant HCC827 cells induced by continuous exposure to HGF. In the in vivo model, E7050 plus gefitinib resulted in marked regression of tumor growth associated with inhibition of Akt phosphorylation in cancer cells. CONCLUSIONS A new Met kinase inhibitor, E7050, reverses the three HGF-induced mechanisms of gefitinib resistance, suggesting that E7050 may overcome HGF-induced resistance to gefitinib and next-generation EGFR-TKIs.
Collapse
Affiliation(s)
- Wei Wang
- Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
CNK3 and IPCEF1 produce a single protein that is required for HGF dependent Arf6 activation and migration. Exp Cell Res 2012; 318:228-37. [DOI: 10.1016/j.yexcr.2011.10.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/13/2011] [Accepted: 10/31/2011] [Indexed: 11/20/2022]
|
159
|
Abstract
Uncontrolled cell survival, growth, angiogenesis and metastasis are essential hallmarks of cancer. Genetic and biochemical data have demonstrated that the growth and motility factor hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, the tyrosine kinase MET, have a causal role in all of these processes, thus providing a strong rationale for targeting these molecules in cancer. Parallel progress in understanding the structure and function of HGF/SF, MET and associated signalling components has led to the successful development of blocking antibodies and a large number of small-molecule MET kinase inhibitors. In this Review, we discuss these advances, as well as results from recent clinical studies that demonstrate that inhibiting MET signalling in several types of solid human tumours has major therapeutic value.
Collapse
Affiliation(s)
- Ermanno Gherardi
- Medical Research Council (MRC) Centre, Hills Road, Cambridge CB2 2QH, UK.
| | | | | | | |
Collapse
|
160
|
Wang CC, Jamal L, Janes KA. Normal morphogenesis of epithelial tissues and progression of epithelial tumors. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2012; 4:51-78. [PMID: 21898857 PMCID: PMC3242861 DOI: 10.1002/wsbm.159] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epithelial cells organize into various tissue architectures that largely maintain their structure throughout the life of an organism. For decades, the morphogenesis of epithelial tissues has fascinated scientists at the interface of cell, developmental, and molecular biology. Systems biology offers ways to combine knowledge from these disciplines by building integrative models that are quantitative and predictive. Can such models be useful for gaining a deeper understanding of epithelial morphogenesis? Here, we take inventory of some recurring themes in epithelial morphogenesis that systems approaches could strive to capture. Predictive understanding of morphogenesis at the systems level would prove especially valuable for diseases such as cancer, where epithelial tissue architecture is profoundly disrupted.
Collapse
Affiliation(s)
- Chun-Chao Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Leen Jamal
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
161
|
Izumida Y, Aoki T, Yasuda D, Koizumi T, Suganuma C, Saito K, Murai N, Shimizu Y, Hayashi K, Odaira M, Kusano T, Kushima M, Kudano M. Hepatocyte growth factor is constitutively produced by donor-derived bone marrow cells and promotes regeneration of pancreatic beta-cells. Biochem Biophys Res Commun 2011; 333:273-82. [PMID: 15950193 DOI: 10.1016/j.bbrc.2005.05.100] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 05/17/2005] [Indexed: 01/29/2023]
Abstract
Recent studies have demonstrated that the transplantation of bone marrow cells following diabetes induced by streptozotocin can support the recovery of pancreatic b-cell mass and a partial reversal of hyperglycemia. To address this issue, we examined whether the c-Met/hepatocyte growth factor (HGF) signaling pathway was involved in the recovery of b-cell injury after bone marrow transplantation (BMT). In this model, donor-derived bone marrow cells were positive for HGF immunoreactivity in the recipient spleen, liver, lung, and pancreas as well as in the host hepatocytes. Indeed, plasma HGF levels were maintained at a high value.The frequency of c-Met expression and its proliferative activity and differentiative response in the pancreatic ductal cells in the BMT group were greater than those in the PBS-treated group, resulting in an elevated number of endogenous insulin-producing cells. The induction of the c-Met/HGF signaling pathway following BMT promotes pancreatic regeneration in diabetic rats.
Collapse
Affiliation(s)
- Yoshihiko Izumida
- Department of General and Gastrointestinal Surgery, Showa University, School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Talbot NC, Sparks WO, Powell AM, Kahl S, Caperna TJ. Quantitative and semiquantitative immunoassay of growth factors and cytokines in the conditioned medium of STO and CF-1 mouse feeder cells. In Vitro Cell Dev Biol Anim 2011; 48:1-11. [PMID: 22179674 DOI: 10.1007/s11626-011-9467-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 10/31/2011] [Indexed: 01/14/2023]
Abstract
Feeder cells of irradiated mouse fibroblasts are commonly used for, and are generally necessary for, the in vitro maintenance and growth of many fastidious cell types, particularly embryonic stem cells or induced pluripotent stem cells. Quantitative and semiquantitative immunoassays of conditioned media were performed to identify some of the soluble cytokines, chemokines, protein hormones, and cell matrix/adhesion molecules that are elaborated from two commonly used feeder cells, STO and CF-1. Among those quantitatively assayed, the most abundant cytokine proteins expressed by the feeder cells were activin A, hepatocyte growth factor (HGF), insulin-like growth factor 1, insulin-like growth factor 2, insulin-like growth factor binding protein (IGFBP)-6, macrophage colony-stimulating factor (a.k.a. CSF-1), and pigment epithelium-derived factor (a.k.a. serine protease inhibitor, clade F, member 1). CF-1 cells expressed ten times more activin A than STO cells and also produced larger amounts of interleukin-6 and IGFBP-2, IGFBP-3, IGFBP-4, and IGFBP-5. Conversely, STO cell produced almost ten times more HGF and five times more stem cell factor (a.k.a. c-kit ligand) than CF-1 cells. Assayed semiquantitatively, relatively large amounts of chemokines were produced by both feeder cells including fractalkine (CX3CL1), interferon-inducible protein 10 (a.k.a. CXCL10 and cytokine-responsive gene-2, CRG-2), monocyte chemotactic protein (MCP)-1 (a.k.a. CCL2 and junctional epithelium chemokine (JE), MCP-5/CCL12), keratinocyte-derived chemokine (a.k.a. CXCL1 and growth-related oncogene alpha, GROα), nephroblastoma overexpressed gene (CCN3, IGFBP-9), stromal cell-derived factor 1 (CXCL12), and serpin E1 (PAI-1). In contrast to one another, STO produced more CXCL16 than CF-1 cells, and CF-1 cell produced more MCP-5 (CCL12), macrophage inflammatory protein (MIP)-1α (CCL3), MIP-1β (CCL4), pentraxin-3 (TSG-14), and platelet factor-4 (CXCL4) than STO cells. Soluble adhesion molecule, sICAM (ICAM-1, CD54), was expressed by CF-1 cells, but not STO cells, and similarly, the cell matrix-associated molecules endocan (endothelial cell-specific molecule 1), endostatin (collagen XVIII), and matrix metalloproteinase 3 were expressed more by CF-1 cells. Tissue inhibitor of metalloproteinases 1 was robustly expressed by both feeder cells. Other proteins primarily detected from CF-1 cells included retinol-binding protein 4 and FGF21, while STO cells secreted more interferon gamma. Both feeder cells produced no or low amounts of LIF, tumor necrosis factor alpha, vascular endothelial growth factor (VEGF), VEGF-B, prolactin, various interleukins, fibroblast growth factor (FGF)-1, FGF-2, FGF-7, EGF, HB-EGF, and amphiregulin. The results may explain some of the cell growth and maintenance responses by various types of cells co-cultured on STO or CF-1 feeder cells.
Collapse
Affiliation(s)
- Neil C Talbot
- USDA, ARS, Animal and Natural Resources Institute, Animal Biosciences and Biotechnology Laboratory, Beltsville, MD 20705, USA.
| | | | | | | | | |
Collapse
|
163
|
Tsukahara Y, Tanaka M, Miyajima A. TROP2 expressed in the trunk of the ureteric duct regulates branching morphogenesis during kidney development. PLoS One 2011; 6:e28607. [PMID: 22194864 PMCID: PMC3237457 DOI: 10.1371/journal.pone.0028607] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/11/2011] [Indexed: 11/18/2022] Open
Abstract
TROP2, a cell surface protein structurally related to EpCAM, is expressed in various carcinomas, though its function remains largely unknown. We examined the expression of TROP2 and EpCAM in fetal mouse tissues, and found distinct patterns in the ureteric bud of the fetal kidney, which forms a tree-like structure. The tip cells in the ureteric bud proliferate to form branches, whereas the trunk cells differentiate to form a polarized ductal structure. EpCAM was expressed throughout the ureteric bud, whereas TROP2 expression was strongest at the trunk but diminished towards the tips, indicating the distinct cell populations in the ureteric bud. The cells highly expressing TROP2 (TROP2(high)) were negative for Ki67, a proliferating cell marker, and TROP2 and collagen-I were co-localized to the basal membrane of the trunk cells. TROP2(high) cells isolated from the fetal kidney failed to attach and spread on collagen-coated plates. Using MDCK cells, a well-established model for studying the branching morphogenesis of the ureteric bud, TROP2 was shown to inhibit cell spreading and motility on collagen-coated plates, and also branching in collagen-gel cultures, which mimic the ureteric bud's microenvironment. These results together suggest that TROP2 modulates the interaction between the cells and matrix and regulates the formation of the ureteric duct by suppressing branching from the trunk during kidney development.
Collapse
Affiliation(s)
- Yuko Tsukahara
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Minoru Tanaka
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (MT); (AM)
| | - Atsushi Miyajima
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (MT); (AM)
| |
Collapse
|
164
|
Gao J, Inagaki Y, Song P, Qu X, Kokudo N, Tang W. Targeting c-Met as a promising strategy for the treatment of hepatocellular carcinoma. Pharmacol Res 2011; 65:23-30. [PMID: 22138044 DOI: 10.1016/j.phrs.2011.11.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 11/15/2011] [Accepted: 11/16/2011] [Indexed: 02/05/2023]
Abstract
Hepatocellular carcinoma (HCC) is a severe condition that is found worldwide. Liver transplantation, surgical resection, and local-regional therapy such as transarterial chemoembolization have made great progress and play a dominant role in HCC management. However, the high frequency of tumor recurrence and/or metastasis after those treatments acquires systematic drug intervention. The approval of sorafenib, an agent that targets receptor tyrosine kinases (RTKs), as the first effective drug for systemic treatment of HCC represents a milestone in treatment of this disease. As a typical member of the RTK family, c-Met represents an intriguing target for cancer therapy. However, the role of the c-Met signal transduction pathway is less unambiguous in HCC pathology, giving rise to concerns about the feasibility of utilizing c-Met targeting approaches for HCC treatment. Recently, studies on des-γ-carboxy prothrombin, an abnormal cytokine secreted by HCC cells, by the current authors and other researchers have highlighted the critical role of c-Met signaling in HCC progression. This review takes a second look at the c-Met signal transduction pathway and discusses the possibility of targeting c-Met as a therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Jianjun Gao
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | | | | | | | | | | |
Collapse
|
165
|
Hwang CI, Choi J, Zhou Z, Flesken-Nikitin A, Tarakhovsky A, Nikitin AY. MET-dependent cancer invasion may be preprogrammed by early alterations of p53-regulated feedforward loop and triggered by stromal cell-derived HGF. Cell Cycle 2011; 10:3834-40. [PMID: 22071625 DOI: 10.4161/cc.10.22.18294] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MET, a receptor protein tyrosine kinase activated by hepatocyte growth factor (HGF), is a crucial determinant of metastatic progression. Recently, we have identified p53 as an important regulator of MET-dependent cell motility and invasion. This regulation occurs via feedforward loop suppressing MET expression by miR-34-dependent and -independent mechanisms. Here, by using Dicer conditional knockout, we provide further evidence for microRNA-independent MET regulation by p53. Furthermore, we show that while MET levels increase immediately after p53 inactivation, mutant cells do not contain active phosphorylated MET and remain non-invasive for a long latency period at contrary to cell culture observations. Evaluation of mouse models of ovarian and prostate carcinogenesis indicates that formation of desmoplastic stroma, associated production of HGF by stromal cells and coinciding MET phosphorylation precede cancer invasion. Thus, initiation mutation of p53 is sufficient for preprogramming motile and invasive properties of epithelial cells, but the stromal reaction may represent a critical step for their manifestation during cancer progression.
Collapse
Affiliation(s)
- Chang-Il Hwang
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | | | | | | | | | | |
Collapse
|
166
|
Sattler M, Reddy MM, Hasina R, Gangadhar T, Salgia R. The role of the c-Met pathway in lung cancer and the potential for targeted therapy. Ther Adv Med Oncol 2011; 3:171-84. [PMID: 21904579 DOI: 10.1177/1758834011408636] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hepatocyte growth factor receptor (HGFR), the product of the MET gene, plays an important role in normal cellular function and oncogenesis. In cancer, HGFR has been implicated in cellular proliferation, cell survival, invasion, cell motility, metastasis and angiogenesis. Activation of HGFR can occur through binding to its ligand, hepatocyte growth factor (HGF), overexpression/amplification, mutation, and/or decreased degradation. Amplification of HGFR can occur de novo or in resistance to therapy. Mutations of HGFR have been described in the tyrosine kinase domain, juxtamembrane domain, or semaphorin domain in a number of tumors. These mutations appear to have gain of function, and also reflect differential sensitivity to therapeutic inhibition. There have been various drugs developed to target HGFR, including antibodies to HGFR/HGF, small-molecule inhibitors against the tyrosine kinase domain of HGFR and downstream targets. Different HGFR inhibitors are currently in clinical trials in lung cancer and a number of solid tumors. Several phase I trials have already been completed, and two specific trials have been reported combining HGFR with epidermal growth factor receptor (EGFR) inhibition in non-small cell lung cancer. In particular, trials involving MetMAb and ARQ197 (tivantinib) have gained interest. Ultimately, as individualized therapies become a reality for cancers, HGFR will be an important molecular target.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, and Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
167
|
The Dual Role of Epithelial-to-Mesenchymal Transition in Chronic Allograft Injury in Pediatric Renal Transplantation. Transplantation 2011; 92:787-95. [DOI: 10.1097/tp.0b013e31822d092c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
168
|
Lau PCP, Chan ATC. Novel therapeutic target for head and neck squamous cell carcinoma: HGF-MET signaling pathway. Anticancer Drugs 2011; 22:665-73. [PMID: 21709616 DOI: 10.1097/cad.0b013e328341879d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) represents a devastating type of malignancy characterized by its high incidence of regional and distant metastases at the time of diagnosis. Vital physiological functions in the upper aerodigestive tract are often impaired as a result of the disease and treatment for the disease, giving rise to severe morbidity in patients suffering from this type of cancer. It is crucial to delineate the aberrant growth signaling pathways in HNSCC cells and develop specific target therapies for the disease to improve the treatment outcome. Although the epidermal growth factor receptor pathway has been extensively studied in HNSCC and anti-epidermal growth factor receptor therapy has already shown promise in treating HNSCC in phase III clinical trials, the signaling pathway that accounts for the highly invasive phenotype of HNSCC needs to be defined and also therapeutically targeted. The hepatocyte growth factor-MET signaling pathway has been studied extensively over the past two decades and it is now clear that it plays an important role in mediating invasive growth of many types of cancer. Here, we review comprehensively the evidence on hepatocyte growth factor-MET cascade being a key in the signaling pathway in mediating invasive growth of HNSCC and the potential of this signaling pathway to be a therapeutic target for the treatment of HNSCC.
Collapse
Affiliation(s)
- Patrick Chi-pan Lau
- State Key Laboratory of Oncology, South China, Sir YK Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute, The Chinese University of Hong Kong, Hong Kong SAR.
| | | |
Collapse
|
169
|
Mahjour SB, Ghaffarpasand F, Wang H. Hair follicle regeneration in skin grafts: current concepts and future perspectives. TISSUE ENGINEERING PART B-REVIEWS 2011; 18:15-23. [PMID: 21883016 DOI: 10.1089/ten.teb.2011.0064] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The repair and management of full-thickness skin defects resulting from burns and chronic wounds remain a significant unmet clinical challenge. For those skin defects exceeding 50%-60% of total body surface area, it is impractical to treat with autologous skin transplants because of the shortage of donor sites. The possibility of using tissue-engineered skin grafts for full-thickness wound repair is a promising approach. The primary goal of tissue-engineered skin grafts is to restore lost barrier function, but regeneration of appendages, such as hair follicles, has to be yet achieved. The successful regeneration of hair follicles in immunodeficient mice suggests that creating human hair follicles in tissue-engineered skin grafts is feasible. However, many limitations still need to be explored, particularly enriching isolated cells with trichogenic capacity, maintaining this ability during processing, and providing the cells with proper environmental cues. Current advances in hair follicle regeneration, in vitro and in vivo, are concisely summarized in this report, and key requirements to bioengineer a hair follicle are proposed, with emphasis on a three-dimensional approach.
Collapse
Affiliation(s)
- Seyed Babak Mahjour
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, USA
| | | | | |
Collapse
|
170
|
Abstract
The mammary gland undergoes a spectacular series of changes as it develops, and maintains a remarkable capacity to remodel and regenerate for several decades. Mammary morphogenesis has been investigated for over 100 years, motivated by the dairy industry and cancer biologists. Over the past decade, the gland has emerged as a major model system in its own right for understanding the cell biology of tissue morphogenesis. Multiple signalling pathways from several cell types are orchestrated together with mechanical cues and cell rearrangements to establish the pattern of the mammary gland. The integrated mechanical and molecular pathways that control mammary morphogenesis have implications for the developmental regulation of other epithelial organs.
Collapse
|
171
|
Miya M, Maeshima A, Mishima K, Sakurai N, Ikeuchi H, Kuroiwa T, Hiromura K, Yokoo H, Nojima Y. Enhancement of in vitro human tubulogenesis by endothelial cell-derived factors: implications for in vivo tubular regeneration after injury. Am J Physiol Renal Physiol 2011; 301:F387-95. [DOI: 10.1152/ajprenal.00619.2010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Renal proximal tubular epithelium can regenerate after various insults. To examine whether the tubular repair process is regulated by surrounding peritubular capillaries, we established an in vitro human tubulogenesis model that mimics in vivo tubular regeneration after injury. In this model, HGF, a potent renotropic factor, dose dependently induced tubular structures in human renal proximal tubular epithelial cells cultured in gels. Consistent with regenerating tubular cells after injury, HGF-induced tubular structures expressed a developmental gene, Pax-2, and a mesenchymal marker, vimentin, and formed a lumen with aquaporin-1 expression. Electron microscopic analysis showed the presence of microvilli on the apical site of the lumen, suggesting that these structures are morphologically equivalent to renal tubules in vivo. When cocultured with human umbilical vein endothelial cells (HUVEC), HGF-induced tubular formation was significantly enhanced. This could not be reproduced by the addition of VEGF, basic FGF, or PDGF. Protein array revealed that HUVEC produced various matrix metalloproteinases (MMPs). The stimulatory effects of coculture with HUVEC or HUVEC-derived conditional medium were almost completely abolished by addition of the tissue inhibitor of metalloproteinase (TIMP)-1 or TIMP-2. These data suggest that endothelial cell-derived factors including MMPs play a critical role in tubulogenesis and imply a potential role of peritubular capillary endothelium as a source of factor(s) required for tubular recovery after injury.
Collapse
Affiliation(s)
- Masaaki Miya
- Departments of 1Medicine and Clinical Science and
| | | | | | | | | | | | | | - Hideaki Yokoo
- Human Pathology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | |
Collapse
|
172
|
Pavlovich A, Boghaert E, Nelson CM. Mammary branch initiation and extension are inhibited by separate pathways downstream of TGFβ in culture. Exp Cell Res 2011; 317:1872-84. [PMID: 21459084 PMCID: PMC3123406 DOI: 10.1016/j.yexcr.2011.03.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/21/2011] [Accepted: 03/27/2011] [Indexed: 01/17/2023]
Abstract
During the branching morphogenesis process that builds epithelial trees, signaling from stimulatory and inhibitory growth factors is integrated to control branch initiation and extension into the surrounding stroma. Here, we examined the relative roles played by these stimulatory and inhibitory signals in the patterning of branch initiation and extension of model mammary epithelial tubules in culture. We found that although several growth factors could stimulate branching, they did not determine the sites at which new branches formed or the lengths to which branches extended. Instead, branch initiation and extension were defined by two separate signals downstream of the inhibitory morphogen, transforming growth factor (TGF)-β. Branch initiation was controlled by signaling through p38 mitogen-activated protein kinase, whereas branch extension was controlled by Smad-mediated induction of a second diffusible inhibitor, Wnt5a. These data suggest that mammary epithelial branching is patterned predominately by repulsive signaling, and that TGFβ activates multiple inhibitory pathways to refine the architecture of the tree.
Collapse
Affiliation(s)
- Amira Pavlovich
- Department of Chemical & Biological Engineering Princeton University, Princeton NJ 08544
| | - Eline Boghaert
- Department of Chemical & Biological Engineering Princeton University, Princeton NJ 08544
| | - Celeste M. Nelson
- Department of Chemical & Biological Engineering Princeton University, Princeton NJ 08544
- Department of Molecular Biology Princeton University, Princeton NJ 08544
| |
Collapse
|
173
|
Garner OB, Bush KT, Nigam KB, Yamaguchi Y, Xu D, Esko JD, Nigam SK. Stage-dependent regulation of mammary ductal branching by heparan sulfate and HGF-cMet signaling. Dev Biol 2011; 355:394-403. [PMID: 21586278 PMCID: PMC3118867 DOI: 10.1016/j.ydbio.2011.04.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 04/18/2011] [Accepted: 04/30/2011] [Indexed: 02/08/2023]
Abstract
Specific interactions of growth factors with heparan sulfate may function as "switches" to regulate stages of branching morphogenesis in developing mammalian organs, such as breast, lung, salivary gland and kidney, but the evidence derives mostly from studies of explanted tissues or cell culture (Shah et al., 2004). We recently provided in vivo evidence that inactivation of Ndst1, the predominant N-deacetylase/N-sulfotransferase gene essential for the formation of mature heparan sulfate, results in a highly specific defect in murine lobuloalveolar development (Crawford et al., 2010). Here, we demonstrate a highly penetrant dramatic defect in primary branching by mammary epithelial-specific inactivation of Ext1, a subunit of the copolymerase complex that catalyzes the formation of the heparan sulfate chain. In contrast to Ext1 deletion, inactivation of Hs2st (which encodes an enzyme required for 2-O-sulfation of uronic acids in heparan sulfate) did not inhibit ductal formation but displayed markedly decreased secondary and ductal side-branches as well as fewer bifurcated terminal end buds. Targeted conditional deletion of c-Met, the receptor for HGF, in mammary epithelial cells showed similar defects in secondary and ductal side-branching, but did not result in any apparent defect in bifurcation of terminal end buds. Although there is published evidence indicating a role for 2-O sulfation in HGF binding, primary epithelial cells isolated from Hs2st conditional deletions were able to activate Erk in the presence of HGF and there appeared to be only a slight reduction in HGF-mediated c-Met phosphorylation in these cells compared to control. Thus, both c-Met and Hs2st play important, but partly independent, roles in secondary and ductal side-branching. When considered together with previous studies of Ndst1-deficient glands, the data presented here raise the possibility of partially-independent regulation by heparan sulfate-dependent pathways of primary ductal branching, terminal end bud bifurcation, secondary branching, ductal side-branching and lobuloalveolar formation.
Collapse
Affiliation(s)
- Omai B. Garner
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, 92093
| | - Kevin T. Bush
- Department of Pediatrics, University of California, San Diego, La Jolla, California, 92093
| | - Kabir B. Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
| | - Yu Yamaguchi
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California, 92093
| | - Ding Xu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, 92093
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, 92093
| | - Sanjay K. Nigam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, 92093
- Department of Pediatrics, University of California, San Diego, La Jolla, California, 92093
- Department of Bioengineering, University of California, San Diego, La Jolla, California, 92093
| |
Collapse
|
174
|
Lim DL, Ko R, Pautler SE. Current understanding of the molecular mechanisms of kidney cancer: a primer for urologists. Can Urol Assoc J 2011; 1:S13-20. [PMID: 18542780 DOI: 10.5489/cuaj.63] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC), the fifth leading malignant condition for men and tenth for women, accounts for 3% of all malignancies in Canada. It is a heterogeneous epithelial malignancy with different subtypes and varied tumour biology. Although most cases of RCC are sporadic, up to 4% of patients have an inherited predisposition for the disease. In this article, we review the current molecular genetics of the different subtypes in hereditary and sporadic RCC. Significant developments in understanding the underlying genetic basis of RCC over the last 2 decades are attributed to intensive research about rare inherited renal cancer syndromes and the identification of the genes responsible for them. Many of these genes are also found in sporadic RCC. Understanding the molecular mechanisms involved in the pathogenesis of RCC has aided the development of molecular-targeted drugs for this disease.
Collapse
Affiliation(s)
- Darwin L Lim
- Divisions of Urology and Surgical Oncology, University of Western Ontario, London, Ont
| | | | | |
Collapse
|
175
|
Jia L, Liu F, Hansen SH, ter Beest MB, Zegers MM. Distinct roles of cadherin-6 and E-cadherin in tubulogenesis and lumen formation. Mol Biol Cell 2011; 22:2031-41. [PMID: 21508319 PMCID: PMC3113768 DOI: 10.1091/mbc.e11-01-0038] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/11/2011] [Accepted: 04/12/2011] [Indexed: 12/21/2022] Open
Abstract
Classic cadherins are important regulators of tissue morphogenesis. The predominant cadherin in epithelial cells, E-cadherin, has been extensively studied because of its critical role in normal epithelial development and carcinogenesis. Epithelial cells may also coexpress other cadherins, but their roles are less clear. The Madin Darby canine kidney (MDCK) cell line has been a popular mammalian model to investigate the role of E-cadherin in epithelial polarization and tubulogenesis. However, MDCK cells also express relatively high levels of cadherin-6, and it is unclear whether the functions of this cadherin are redundant to those of E-cadherin. We investigate the specific roles of both cadherins using a knockdown approach. Although we find that both cadherins are able to form adherens junctions at the basolateral surface, we show that they have specific and mutually exclusive roles in epithelial morphogenesis. Specifically, we find that cadherin-6 functions as an inhibitor of tubulogenesis, whereas E-cadherin is required for lumen formation. Ablation of cadherin-6 leads to the spontaneous formation of tubules, which depends on increased phosphoinositide 3-kinase (PI3K) activity. In contrast, loss of E-cadherin inhibits lumen formation by a mechanism independent of PI3K.
Collapse
Affiliation(s)
- Liwei Jia
- Department of Surgery, Committee on Cancer Biology, University of Chicago, Chicago IL 60637
| | - Fengming Liu
- Department of Surgery, Committee on Cancer Biology, University of Chicago, Chicago IL 60637
| | - Steen H. Hansen
- Gastrointestinal Cell Biology, Children's Hospital, Boston, MA 02115
| | - Martin B.A. ter Beest
- Department of Surgery, Committee on Cancer Biology, University of Chicago, Chicago IL 60637
| | - Mirjam M.P. Zegers
- Department of Surgery, Committee on Cancer Biology, University of Chicago, Chicago IL 60637
| |
Collapse
|
176
|
Cunha C, Panseri S, Villa O, Silva D, Gelain F. 3D culture of adult mouse neural stem cells within functionalized self-assembling peptide scaffolds. Int J Nanomedicine 2011; 6:943-55. [PMID: 21720506 PMCID: PMC3124398 DOI: 10.2147/ijn.s17292] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional (3D) in vitro models of cell culture aim to fill the gap between the standard two-dimensional cell studies and the in vivo environment. Especially for neural tissue regeneration approaches where there is little regenerative capacity, these models are important for mimicking the extracellular matrix in providing support, allowing the natural flow of oxygen, nutrients, and growth factors, and possibly favoring neural cell regrowth. We have previously demonstrated that a new self-assembling nanostructured biomaterial, based on matrigel, was able to support adult neural stem cell (NSC) culture. In this study, we developed a new 3D cell culture system that takes advantage of the nano- and microfiber assembling process, under physiologic conditions, of these biomaterials. The assembled scaffold forms an intricate and biologically active matrix that displays specifically designed functional motifs: RGD (Arg-Gly-Asp), BMHP1 (bone marrow homing peptide 1), and BMHP2, for the culture of adult NSCs. These scaffolds were prepared at different concentrations, and microscopic examination of the cell-embedded scaffolds showed that NSCs are viable and they proliferate and differentiate within the nanostructured environment of the scaffold. Such a model has the potential to be tailored to develop ad hoc designed peptides for specific cell lines.
Collapse
Affiliation(s)
- Carla Cunha
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Silvia Panseri
- Laboratory of Biomechanics and Technology Innovation, Rizzoli Orthopaedic Institute, Bologna
- Laboratory of Nano-Biomagnetism, Institute of Science and Technology for Ceramics, National Research Council, Faenza, Italy
| | - Omar Villa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Diego Silva
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Fabrizio Gelain
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| |
Collapse
|
177
|
Chambers KF, Pearson JF, Aziz N, O'Toole P, Garrod D, Lang SH. Stroma regulates increased epithelial lateral cell adhesion in 3D culture: a role for actin/cadherin dynamics. PLoS One 2011; 6:e18796. [PMID: 21533155 PMCID: PMC3078910 DOI: 10.1371/journal.pone.0018796] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 03/20/2011] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cell shape and tissue architecture are controlled by changes to junctional proteins and the cytoskeleton. How tissues control the dynamics of adhesion and cytoskeletal tension is unclear. We have studied epithelial tissue architecture using 3D culture models and found that adult primary prostate epithelial cells grow into hollow acinus-like spheroids. Importantly, when co-cultured with stroma the epithelia show increased lateral cell adhesions. To investigate this mechanism further we aimed to: identify a cell line model to allow repeatable and robust experiments; determine whether or not epithelial adhesion molecules were affected by stromal culture; and determine which stromal signalling molecules may influence cell adhesion in 3D epithelial cell cultures. METHODOLOGY/PRINCIPAL FINDINGS The prostate cell line, BPH-1, showed increased lateral cell adhesion in response to stroma, when grown as 3D spheroids. Electron microscopy showed that 9.4% of lateral membranes were within 20 nm of each other and that this increased to 54% in the presence of stroma, after 7 days in culture. Stromal signalling did not influence E-cadherin or desmosome RNA or protein expression, but increased E-cadherin/actin co-localisation on the basolateral membranes, and decreased paracellular permeability. Microarray analysis identified several growth factors and pathways that were differentially expressed in stroma in response to 3D epithelial culture. The upregulated growth factors TGFβ2, CXCL12 and FGF10 were selected for further analysis because of previous associations with morphology. Small molecule inhibition of TGFβ2 signalling but not of CXCL12 and FGF10 signalling led to a decrease in actin and E-cadherin co-localisation and increased paracellular permeability. CONCLUSIONS/SIGNIFICANCE In 3D culture models, paracrine stromal signals increase epithelial cell adhesion via adhesion/cytoskeleton interactions and TGFβ2-dependent mechanisms may play a key role. These findings indicate a role for stroma in maintaining adult epithelial tissue morphology and integrity.
Collapse
Affiliation(s)
- Karen F. Chambers
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Joanna F. Pearson
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Naveed Aziz
- Genomics Lab, Technology Facility, Department of Biology, University of York, Heslington, York, United Kingdom
| | - Peter O'Toole
- Imaging and Cytometry Lab, Technology Facility, Department of Biology, University of York, Heslington, York, United Kingdom
| | - David Garrod
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
- King Saud University, Riyadh, Saudi Arabia
| | - Shona H. Lang
- YCR Cancer Research Unit, Department of Biology, University of York, Heslington, York, United Kingdom
| |
Collapse
|
178
|
De Bacco F, Luraghi P, Medico E, Reato G, Girolami F, Perera T, Gabriele P, Comoglio PM, Boccaccio C. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. J Natl Cancer Inst 2011; 103:645-61. [PMID: 21464397 DOI: 10.1093/jnci/djr093] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Ionizing radiation (IR) is effectively used in cancer therapy. However, in subsets of patients, a few radioresistant cancer cells survive and cause disease relapse with metastatic progression. The MET oncogene encodes the hepatocyte growth factor (HGF) receptor and is known to drive "invasive growth", a regenerative and prosurvival program unduly activated in metastasis. METHODS Human tumor cell lines (MDA-MB-231, MDA-MB-435S, U251) were subjected to therapeutic doses of IR. MET mRNA, and protein expression and signal transduction were compared in treated and untreated cells, and the involvement of the DNA-damage sensor ataxia telangiectasia mutated (ATM) and the transcription factor nuclear factor kappa B (NF-κB) in activating MET transcription were analyzed by immunoblotting, chromatin immunoprecipitation, and use of NF-κB silencing RNA (siRNA). Cell invasiveness was measured in wound healing and transwell assays, and cell survival was measured in viability and clonogenic assays. MET was inhibited by siRNA or small-molecule kinase inhibitors (PHA665752 or JNJ-38877605). Combinations of MET-targeted therapy and radiotherapy were assessed in MDA-MB-231 and U251 xenografts (n = 5-6 mice per group). All P values were from two-sided tests. RESULTS After irradiation, MET expression in cell lines was increased up to fivefold via activation of ATM and NF-κB. MET overexpression increased ligand-independent MET phosphorylation and signal transduction, and rendered cells more sensitive to HGF. Irradiated cells became more invasive via a MET-dependent mechanism that was further enhanced in the presence of HGF. MET silencing by siRNA or inhibition of its kinase activity by treatment with PHA665752 or JNJ-38877605 counteracted radiation-induced invasiveness, promoted apoptosis, and prevented cells from resuming proliferation after irradiation in vitro. Treatment with MET inhibitors enhanced the efficacy of IR to stop the growth of or to induce the regression of xenografts (eg, at day 13, U251 xenografts, mean volume increase relative to mean tumor volume at day 0: vehicle = 438%, 5 Gy IR = 151%, 5 Gy IR + JNJ-38877605 = 76%; difference, IR vs JNJ-38877604 + IR = 75%, 95% CI = 59% to 91%, P = .01). CONCLUSION IR induces overexpression and activity of the MET oncogene through the ATM-NF-κB signaling pathway; MET, in turn, promotes cell invasion and protects cells from apoptosis, thus supporting radioresistance. Drugs targeting MET increase tumor cell radiosensitivity and prevent radiation-induced invasiveness.
Collapse
Affiliation(s)
- Francesca De Bacco
- IRCC-Institute for Cancer Research at Candiolo, University of Turin Medical School, Candiolo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Kim CH, Koh YW, Han JH, Kim JW, Lee JS, Baek SJ, Hwang HS, Choi EC. c-Met expression as an indicator of survival outcome in patients with oral tongue carcinoma. Head Neck 2011; 32:1655-64. [PMID: 20848408 DOI: 10.1002/hed.21383] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hepatocyte growth factor (HGF) can promote proliferation, invasion, and angiogenesis in various tumor cells. However, the prognosis according to expression of HGF/c-Met has not been reported in patients with squamous cell carcinoma of the oral tongue (SCCOT). METHODS Tumors from 61 patients with SCCOT were evaluated for the expression of HGF and c-Met by immunohistochemistry. For Western blot, we used fresh normal and cancer tissues from 3 patients with SCCOT. RESULTS The positive rates of HGF and c-Met immunostaining in SCCOT were 57.3% and 54.1%, respectively. The c-Met staining was significantly correlated with lymph node metastasis (p = .005), tumor classification (p = .004), and recurrence (p = .018). Survival was significantly affected in patients with positive c-Met expression (p = .003). HGF and c-Met were strongly expressed in cancer tissues on Western blot. CONCLUSION The c-Met expression may play an important role in the progression and the survival outcome of patients with SCCOT.
Collapse
Affiliation(s)
- Chul-Ho Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | | | |
Collapse
|
180
|
Szponar A, Yusenko MV, Kuiper R, van Kessel AG, Kovacs G. Genomic profiling of papillary renal cell tumours identifies small regions of DNA alterations: a possible role of HNF1B in tumour development. Histopathology 2011; 58:934-43. [DOI: 10.1111/j.1365-2559.2011.03795.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
181
|
Mangold S, Wu S, Norwood S, Collins B, Hamilton N, Thorn P, Yap A. Hepatocyte Growth Factor Acutely Perturbs Actin Filament Anchorage at the Epithelial Zonula Adherens. Curr Biol 2011; 21:503-7. [DOI: 10.1016/j.cub.2011.02.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 01/31/2011] [Accepted: 02/15/2011] [Indexed: 01/13/2023]
|
182
|
Kwon SH, Nedvetsky PI, Mostov KE. Transcriptional profiling identifies TNS4 function in epithelial tubulogenesis. Curr Biol 2011; 21:161-6. [PMID: 21236678 PMCID: PMC3031161 DOI: 10.1016/j.cub.2010.12.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 11/22/2010] [Accepted: 12/16/2010] [Indexed: 12/21/2022]
Abstract
Hepatocyte growth factor (HGF) plays central roles in tubulogenesis and metastasis [1-4]. HGF treatment of Madin-Darby canine kidney (MDCK) cells grown as cysts in three-dimensional culture induces tubulogenesis [5, 6], which like most tubulogenic processes proceeds through distinct intermediate phases. Identification of genes associated with these phases is central to understanding the molecular mechanisms of tubulogensis; however, because of inefficient, asynchronous tubule formation, isolating such genes has been unfeasible. Here we developed a synchronous, efficient tubulogenesis system and used time-course transcriptional profiling to identify genes temporally regulated in developmental intermediates. Knockdown (KD) of tensin 4 (TNS4), a particularly highly upregulated gene, leads to a decrease in formation of extensions and tubules, two sequential intermediates in tubulogenesis. Exogenous expression of TNS4 marks invasive cells in an epithelial sheet. A mutation in the SH2 domain of TNS4 prevents the transition from extension formation to invasive migration during tubule formation and leads to increased basal activation of STAT3. Exogenous expression of a constitutively active STAT3 mimics the defect by the mutation. Our study highlights the role of the TNS4-STAT3 axis in epithelial sheet invasion and tubulogenesis.
Collapse
Affiliation(s)
- Sang-Ho Kwon
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143-2140, USA
| | | | | |
Collapse
|
183
|
Guglielmo MC, Ricci G, Catizone A, Barberi M, Galdieri M, Stefanini M, Canipari R. The effect of hepatocyte growth factor on the initial stages of mouse follicle development. J Cell Physiol 2011; 226:520-9. [PMID: 20683913 DOI: 10.1002/jcp.22361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Interactions between theca and granulosa cells of the follicle are critical for the coordination of ovarian follicle development. The cell-cell interactions are mediated through the local production and actions of a variety of factors. The current study is designed to investigate the expression of Hgf and its receptor, c-Met, in the mouse ovary during in vivo folliculogenesis. We found that Hgf and c-Met mRNAs were already expressed in 2-day-old ovaries, and that, while c-Met levels remained constant until 22-day-old, Hgf levels slightly but not significantly increased with age. The expression of Hgf mRNA in theca/interstitial cells was higher than in granulosa cells in 22-day-old ovaries. Immunohistochemistry analysis confirmed the expression pattern demonstrated by RT-PCR. We investigated the role of hepatocyte growth factor (HGF) at the beginning of mouse folliculogenesis and its possible interaction with kit ligand (KL). Interestingly, both KL and HGF were able to increase the expression of each other, creating a positive feedback loop. In the presence of HGF, we observed an increase of granulosa cell proliferation and an increase in the number of pre-antral and early antral follicles in ovary organ cultures. We also observed a significant increase in the diameters of follicles in individual follicle cultures. Moreover, HGF stimulated the expression of the FSH receptors, both in the whole ovary and in isolated pre-antral follicle cultures. Based on the data presented, we concluded that HGF exerts multiple levels of control over follicular cell functions, which collectively enable the progression of follicular development.
Collapse
Affiliation(s)
- M C Guglielmo
- Department of Histology and Medical Embryology, La Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
184
|
Nakamura T, Sakai K, Nakamura T, Matsumoto K. Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol 2011; 26 Suppl 1:188-202. [PMID: 21199531 DOI: 10.1111/j.1440-1746.2010.06549.x] [Citation(s) in RCA: 356] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver regeneration depends on the proliferation of mature hepatocytes. In the 1980s, the method for the cultivation of mature hepatocytes provided an opportunity for the discovery of hepatocyte growth factor (HGF) as a protein that is structurally and functionally different from other growth factors. In 1991, the scatter factor, tumor cytotoxic factor, and 3-D epithelial morphogen were identified as HGF, and Met tyrosine kinase was identified as the receptor for HGF. Thus, the connection of apparently unrelated research projects rapidly enriched the research on HGF in different fields. The HGF-Met pathway plays important roles in the embryonic development of the liver and the placenta, in the migration of myogenic precursor cells, and in epithelial morphogenesis. The use of tissue-specific knockout mice demonstrated that in mature tissues the HGF-Met pathway plays a critical role in tissue protection and regeneration, and in providing less susceptibility to chronic inflammation and fibrosis. In various injury and disease models, HGF promotes cell survival, regeneration of tissues, and suppresses and improves chronic inflammation and fibrosis. Drug development using HGF has been challenging, but extensive preclinical studies to address its therapeutic effects have provided significant results sufficient for the development of HGF as a biological drug in the regeneration-based therapy of diseases. Clinical trials using recombinant human HGF protein, or HGF genes, are in progress for the treatment of diseases.
Collapse
|
185
|
Subramanian B, Rudym D, Cannizzaro C, Perrone R, Zhou J, Kaplan DL. Tissue-engineered three-dimensional in vitro models for normal and diseased kidney. Tissue Eng Part A 2010; 16:2821-31. [PMID: 20486787 DOI: 10.1089/ten.tea.2009.0595] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Morphogenesis of epithelial cells involves processes by which kidney shape and function are regulated. The lack of in vitro models that are sustainable for longer time periods and emulating complex intercellular interactions of the kidney have limited understanding about epithelial tissue morphogenesis and its aberrations in diseases such as autosomal dominant polycystic kidney disease (ADPKD). A sustainable three-dimensional (3D) coculture system for normal and diseased kidney tissues is reported here. Tubule- and ADPKD cyst-derived cells were cultured in extracellular matrix molecules infused into 3D porous silk scaffolds, and these cultures were subsequently extended into a perfusion bioreactor. The results indicated collagen-matrigel-mediated morphogenesis for both (normal and disease) cell types and also supported coculturing with fibroblasts. The structural and functional features of the kidney-like tissue structures were validated based on the distribution of E-cadherin, N-cadherin, Na+ K+ ATPase pump, and cellular uptake of the organic anion (6-carboxy fluorescein). Further, the structures were sustained for longer time periods using a perfusion bioreactor to demonstrate the potential utility of this 3D in vitro coculture system for ADPKD research, other epithelial tissue systems, and for in vitro drug screening.
Collapse
|
186
|
Wakamatsu Y, Sakai D, Suzuki T, Osumi N. FilaminB is required for the directed localization of cell-cell adhesion molecules in embryonic epithelial development. Dev Dyn 2010; 240:149-61. [DOI: 10.1002/dvdy.22518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
187
|
MET signalling: principles and functions in development, organ regeneration and cancer. Nat Rev Mol Cell Biol 2010; 11:834-48. [PMID: 21102609 DOI: 10.1038/nrm3012] [Citation(s) in RCA: 958] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The MET tyrosine kinase receptor (also known as the HGF receptor) promotes tissue remodelling, which underlies developmental morphogenesis, wound repair, organ homeostasis and cancer metastasis, by integrating growth, survival and migration cues in response to environmental stimuli or cell-autonomous perturbations. The versatility of MET-mediated biological responses is sustained by qualitative and quantitative signal modulation. Qualitative mechanisms include the engagement of dedicated signal transducers and the subcellular compartmentalization of MET signalling pathways, whereas quantitative regulation involves MET partnering with adaptor amplifiers or being degraded through the shedding of its extracellular domain or through intracellular ubiquitylation. Controlled activation of MET signalling can be exploited in regenerative medicine, whereas MET inhibition might slow down tumour progression.
Collapse
|
188
|
Sánchez A, Fabregat I. Growth factor- and cytokine-driven pathways governing liver stemness and differentiation. World J Gastroenterol 2010; 16:5148-61. [PMID: 21049549 PMCID: PMC2975086 DOI: 10.3748/wjg.v16.i41.5148] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [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
Liver is unique in its capacity to regenerate in response to injury or tissue loss. Hepatocytes and other liver cells are able to proliferate and repopulate the liver. However, when this response is impaired, the contribution of hepatic progenitors becomes very relevant. Here, we present an update of recent studies on growth factors and cytokine-driven intracellular pathways that govern liver stem/progenitor cell expansion and differentiation, and the relevance of these signals in liver development, regeneration and carcinogenesis. Tyrosine kinase receptor signaling, in particular, c-Met, epidermal growth factor receptors or fibroblast growth factor receptors, contribute to proliferation, survival and differentiation of liver stem/progenitor cells. Different evidence suggests a dual role for the transforming growth factor (TGF)-β signaling pathway in liver stemness and differentiation. On the one hand, TGF-β mediates progression of differentiation from a progenitor stage, but on the other hand, it contributes to the expansion of liver stem cells. Hedgehog family ligands are necessary to promote hepatoblast proliferation but need to be shut off to permit subsequent hepatoblast differentiation. In the same line, the Wnt family and β-catenin/T-cell factor pathway is clearly involved in the maintenance of liver stemness phenotype, and its repression is necessary for liver differentiation during development. Collectively, data indicate that liver stem/progenitor cells follow their own rules and regulations. The same signals that are essential for their activation, expansion and differentiation are good candidates to contribute, under adequate conditions, to the paradigm of transformation from a pro-regenerative to a pro-tumorigenic role. From a clinical perspective, this is a fundamental issue for liver stem/progenitor cell-based therapies.
Collapse
|
189
|
Marmaras A, Berge U, Ferrari A, Kurtcuoglu V, Poulikakos D, Kroschewski R. A mathematical method for the 3D analysis of rotating deformable systems applied on lumen-forming MDCK cell aggregates. Cytoskeleton (Hoboken) 2010; 67:224-40. [PMID: 20183868 DOI: 10.1002/cm.20438] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell motility contributes to the formation of organs and tissues, into which multiple cells self-organize. However such mammalian cellular motilities are not characterized in a quantitative manner and the systemic consequences are thus unknown. A mathematical tool to decipher cell motility, accounting for changes in cell shape, within a three-dimensional (3D) cell system was missing. We report here such a tool, usable on segmented images reporting the outline of clusters (cells) and allowing the time-resolved 3D analysis of circular motility of these as parts of a system (cell aggregate). Our method can analyze circular motility in sub-cellular, cellular, multi-cellular, and also non-cellular systems for which time-resolved segmented cluster outlines are available. To exemplify, we characterized the circular motility of lumen-initiating MDCK cell aggregates, embedded in extracellular matrix. We show that the organization of the major surrounding matrix fibers was not significantly affected during this cohort rotation. Using our developed tool, we discovered two classes of circular motion, rotation and random walk, organized in three behavior patterns during lumen initiation. As rotational movements were more rapid than random walk and as both could continue during lumen initiation, we conclude that neither the class nor the rate of motion regulates lumen initiation. We thus reveal a high degree of plasticity during a developmentally critical cell polarization step, indicating that lumen initiation is a robust process. However, motility rates decreased with increasing cell number, previously shown to correlate with epithelial polarization, suggesting that migratory polarization is converted into epithelial polarization during aggregate development.
Collapse
Affiliation(s)
- Anastasios Marmaras
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
190
|
Xu D, Matsuo Y, Ma J, Koide S, Ochi N, Yasuda A, Funahashi H, Okada Y, Takeyama H. Cancer cell-derived IL-1α promotes HGF secretion by stromal cells and enhances metastatic potential in pancreatic cancer cells. J Surg Oncol 2010; 102:469-477. [PMID: 20872950 DOI: 10.1002/jso.21530] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Interleukin (IL)-1α and hepatocyte growth factor (HGF) play an important role in pancreatic cancer proliferation, angiogenesis, and invasiveness. The aim of this study was to investigate the cooperative role of HGF and IL-1α in metastatic processes promoted by interactions between pancreatic cancer cells and stromal cells. METHODS Expression of IL-1α and HGF mRNA and protein was determined by RT-PCR and ELISA. The effect of HGF on metastatic potential was evaluated by proliferation, invasion, and angiogenesis assays using an in vitro system consisting of co-cultured tumor cells and stromal cells. RESULTS IL-1α expression was closely correlated with metastatic potential, and cancer cell-derived IL-1α significantly promoted HGF expression by fibroblasts (P < 0.01). HGF not only enhanced the invasiveness and proliferation of pancreatic cancer cells, but also enhanced migration and proliferation of human umbilical vein endothelial cells (HUVECs). HGF significantly enhanced HUVEC tube formation (P < 0.01). Furthermore, the high liver-metastatic pancreatic cancer cell line (BxPC-3), which secretes IL-1α, significantly enhanced HUVEC tube formation compared with the low liver-metastatic cell line (Capan-2), which does not produce IL-1α (P < 0.01). CONCLUSION Autocrine IL-1α and paracrine HGF co-enhance the metastatic potential of pancreatic cancer cells via both IL-1α and HGF signaling pathways.
Collapse
Affiliation(s)
- Donghui Xu
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Science, Mizuhoku, Nagoya, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Lee WC, Hough MT, Liu W, Ekiert R, Lindström NO, Hohenstein P, Davies JA. Dact2 is expressed in the developing ureteric bud/collecting duct system of the kidney and controls morphogenetic behavior of collecting duct cells. Am J Physiol Renal Physiol 2010; 299:F740-51. [PMID: 20685821 DOI: 10.1152/ajprenal.00148.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The overall pattern of the developing kidney is set in large part by the developing ureteric bud/collecting duct system, and dysgenesis of this system accounts for a variety of clinically significant renal diseases. Understanding how the behavior of cells in the developing ureteric bud/collecting duct is controlled is therefore important to understanding the normal and abnormal kidney. Dact proteins have recently been identified as cytoplasmic regulators of intracellular signaling. Dact1 inhibits Wnt signaling, and Dact2 inhibits transforming growth factor (TGF)-β signaling. Here, we report that Dact2 is expressed in developing and adult mouse kidneys, specifically in the ureteric bud/collecting duct epithelium, a structure whose morphogenesis is controlled partially by TGF-β. When small interfering RNA is used to knock down Dact2 expression in collecting duct cells, they show some constitutive phospho-Smad2, undetectable in controls, and elevated phospho-Smad2 in response to TGF-β. They also show defective migration and, in a monolayer wound-healing assay, they fail to assemble a leading edge "cable" of actomyosin and advance instead as a disorganized mass of lamellipodium-bearing cells. This effect is seriously exacerbated by exogenous TGF-β, although control cells tolerate it well. In three-dimensional culture, Dact2 knockdown cells form cysts and branching tubules, but the outlines of the cysts made by knockdown cells are ragged rather than smooth and the branching tubules are decorated with many fine spikes not seen in controls. These data suggest Dact2 plays a role in regulating morphogenesis by renal collecting duct cells, probably by protecting cells from overly strong TGF-β pathway activation.
Collapse
Affiliation(s)
- Wen-Chin Lee
- Centre for Integrative Physiology, Univ. of Edinburgh, Edinburgh, EH8 9XB, UK
| | | | | | | | | | | | | |
Collapse
|
192
|
Kim M, O'Brien LE, Kwon SH, Mostov KE. STAT1 is required for redifferentiation during Madin-Darby canine kidney tubulogenesis. Mol Biol Cell 2010; 21:3926-33. [PMID: 20861313 PMCID: PMC2982126 DOI: 10.1091/mbc.e10-02-0112] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Signal transducers and activators of transcription (STAT)1 is the key to the sequential control of Madin-Darby canine kidney tubulogenesis. Loss of STAT1 prevents redifferentiation. Constitutively active STAT1 is sufficient to restore cord formation but not mature lumens. These data suggest that STAT1 is necessary for the redifferentiation phase of tubulogenesis and that mature lumenogenesis requires a distinct signal. Tubule formation in vitro using Madin-Darby canine kidney (MDCK) epithelial cells consists mainly of two processes. First, the cells undergo a partial epithelial–mesenchymal transition (pEMT), losing polarity and migrating. Second, the cells redifferentiate, forming cords and then tubules with continuous lumens. We have shown previously that extracellular signal-regulated kinase activation is required for pEMT. However, the mechanism of how the pEMT phase is turned off and the redifferentiation phase is initiated is largely unknown. To address the central question of the sequential control of these two phases, we used MDCK cells grown as cysts and treated with hepatocyte growth factor to model tubulogenesis. We show that signal transducer and activator of transcription (STAT)1 controls the sequential progression from the pEMT phase to the redifferentiation phase. Loss of STAT1 prevents redifferentiation. Constitutively active STAT1 allows redifferentiation to occur even when cells are otherwise prevented from progressing beyond the pEMT phase by exogenous activation of Raf. Moreover, tyrosine phosphorylation defective STAT1 partially restored cord formation in such cells, suggesting that STAT1 functions in part as nonnuclear protein mediating signal transduction in this process. Constitutively active or inactive forms of STAT1 did not promote lumen maturation, suggesting this requires a distinct signal.
Collapse
Affiliation(s)
- Minji Kim
- Department of Anatomy, University of California-San Francisco, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
193
|
To WS, Midwood KS. Cryptic domains of tenascin-C differentially control fibronectin fibrillogenesis. Matrix Biol 2010; 29:573-85. [PMID: 20708078 DOI: 10.1016/j.matbio.2010.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 08/02/2010] [Accepted: 08/04/2010] [Indexed: 12/20/2022]
Abstract
The three-dimensional organization of the ubiquitous extracellular matrix glycoprotein fibronectin regulates cell fate and morphogenesis during development; in particular tubule formation that constitutes the vasculature, lung and kidney. Tenascin-C is a matrix protein with a restricted expression pattern; it is specifically up-regulated at sites of fibronectin fibril assembly during development and in remodeling adult tissues. Here we demonstrate that specific domains of tenascin-C inhibit fibronectin matrix assembly whereas full-length tenascin-C does not. These domains act via distinct mechanisms: TNfn1-8 blocks fibrillogenesis by binding to fibronectin fibrils and preventing intermolecular fibronectin interactions whilst FBG acts independently of binding to fibronectin and instead is internalized and causes cytoskeletal re-organization. We also show that TNfn1-8 disrupts epithelial cell tubulogenesis. Our data demonstrate that tenascin-C contains cryptic sites which can control tissue levels of fibrillar fibronectin either by preventing de novo fibril assembly or reducing levels of deposited fibronectin. Exposure of these domains during tissue remodeling may provide a novel means of controlling fibronectin assembly and tubulogenic processes dependent on the assembly of this matrix.
Collapse
Affiliation(s)
- Wing S To
- Department of Matrix Biology, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College, 65 Aspenlea Road, London, W6 8LH, UK
| | | |
Collapse
|
194
|
Togawa A, Sfakianos J, Ishibe S, Suzuki S, Fujigaki Y, Kitagawa M, Mellman I, Cantley LG. Hepatocyte Growth Factor stimulated cell scattering requires ERK and Cdc42-dependent tight junction disassembly. Biochem Biophys Res Commun 2010; 400:271-7. [PMID: 20728428 DOI: 10.1016/j.bbrc.2010.08.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 08/17/2010] [Indexed: 12/25/2022]
Abstract
The mechanism by which Hepatocyte Growth Factor (HGF) induces tight junction disassembly prior to cell scattering is largely unknown. Here, we show that HGF stimulates rapid loss of the TJ assembly protein Par6 from the TJ in an Erk-dependent manner. Erk activation by HGF is found to mediate the interaction of Par6 with GTP-loaded Cdc42. The Cdc42 GTPase activating protein cdGAP is shown to interact with Pkcζ at baseline and prevent Par6-Cdc42 association. Erk, by phosphorylating cdGAP at threonine776, can inhibit the GAP activity, thereby increasing Par6-Cdc42 association and TJ disassembly. Our findings reveal a novel pathway for regulating HGF signaling to the Par proteins through Erk-cdGAP, resulting in TJ disassembly and cell scattering.
Collapse
Affiliation(s)
- Akashi Togawa
- Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | | | | | |
Collapse
|
195
|
Raghavan S, Shen CJ, Desai RA, Sniadecki NJ, Nelson CM, Chen CS. Decoupling diffusional from dimensional control of signaling in 3D culture reveals a role for myosin in tubulogenesis. J Cell Sci 2010; 123:2877-83. [PMID: 20682635 DOI: 10.1242/jcs.055079] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We present a novel microfabricated platform to culture cells within arrays of micrometer-scale three-dimensional (3D) extracellular matrix scaffolds (microgels). These microscale cultures eliminate diffusion barriers that are intrinsic to traditional 3D culture systems (macrogels) and enable uniform cytokine stimulation of the entire culture population, as well as allow immunolabeling, imaging and population-based biochemical assays across the relatively coplanar microgels. Examining early signaling associated with hepatocyte growth factor (HGF)-mediated scattering and tubulogenesis of MDCK cells revealed that 3D culture modulates cellular responses both through dimensionality and altered stimulation rates. Comparing responses in 2D culture, microgels and macrogels demonstrated that HGF-induced ERK signaling was driven by the dynamics of stimulation and not by whether cells were in a 2D or 3D environment, and that this ERK signaling was equally important for HGF-induced cell scattering on 2D substrates and tubulogenesis in 3D. By contrast, we discovered a specific HGF-induced increase in myosin expression leading to sustained downregulation of myosin activity that occurred only within 3D contexts and was required for 3D tubulogenesis but not 2D scattering. Interestingly, although absent in cells on collagen-coated plates, downregulation of myosin activity also occurred for cells on collagen gels, but was transient and mediated by a combination of myosin dephosphorylation and enhanced myosin expression. Furthermore, upregulating myosin activity via siRNA targeted to a myosin phosphatase did not attenuate scattering in 2D but did inhibit tubulogenesis in 3D. Together, these results demonstrate that cellular responses to soluble cues in 3D culture are regulated by both rates of stimulation and by matrix dimensionality, and highlight the importance of decoupling these effects to identify early signals relevant to cellular function in 3D environments.
Collapse
Affiliation(s)
- Srivatsan Raghavan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
196
|
Talbot NC, Blomberg LA, Garrett WM, Caperna TJ. Feeder-independent continuous culture of the PICM-19 pig liver stem cell line. In Vitro Cell Dev Biol Anim 2010; 46:746-57. [PMID: 20607619 DOI: 10.1007/s11626-010-9336-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 06/16/2010] [Indexed: 01/28/2023]
Abstract
The PICM-19 pig liver stem cell line is a bipotent cell line, i.e., capable of forming either bile ductules or hepatocyte monolayers in vitro, that was derived from the primary culture of pig embryonic stem cells. The cell line has been strictly feeder-dependent in that cell replication, morphology, and function were lost if the cells were cultured without STO feeder cells. A method for the feeder-independent continuous culture of PICM-19 cells (FI-PICM-19) is presented. PICM-19 cells were maintained and grown without feeder cells on collagen I-coated tissue culture plastic for 26 passages (P26) with initial split ratios of 1:3 that diminished to split ratios of less than 1:2 after passage 16. Once plated, the FI-PICM-19 cells were overlaid with a 1:12 to 1:50 dilution of Matrigel or related extracellular matrix product. Growth of the cells was stimulated by daily refeedings with STO feeder-cell conditioned medium. The FI-PICM-19 cells grew to an approximate confluence of 50% prior to each passage at 2-wk intervals. Growth curve analysis showed their average cell number doubling time to be ~96 h. Morphologically, the feeder-independent cells closely resembled PICM-19 cells grown on feeder cells, and biliary canalicui were present at cell-to-cell junctions. However, no spontaneous multicellular ductules formed in the monolayers of FI-PICM-19 cells. Ultrastructural subcellular features of the FI-PICM-19 cells were similar to those of PICM-19 cells cultured on feeder cells. The FI-PICM-19 cells produced a spectrum of serum proteins and expressed many liver/hepatocyte-specific genes. Importantly, cytochrome P450 (EROD) activity, ammonia clearance, and urea production were maintained by the feeder-independent cells. This simple method for the propagation of the PICM-19 cell line without feeder cells should simplify the generation and selection of functional mutants within the population and enhances the cell line's potential for use in toxicological/pharmacological screening assays and for use in an artificial liver device.
Collapse
Affiliation(s)
- Neil C Talbot
- U. S. Department of Agriculture, Agricultural Research Service, Animal and Natural Resources Institute, Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Bldg. 200, Rm. 13, BARC-East, Beltsville, MD 20705, USA.
| | | | | | | |
Collapse
|
197
|
Hunter MP, Zegers MM. Pak1 regulates branching morphogenesis in 3D MDCK cell culture by a PIX and beta1-integrin-dependent mechanism. Am J Physiol Cell Physiol 2010; 299:C21-32. [PMID: 20457839 PMCID: PMC2904258 DOI: 10.1152/ajpcell.00543.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 03/17/2010] [Indexed: 12/22/2022]
Abstract
Branching morphogenesis is a fundamental process in the development of the kidney. This process gives rise to a network of ducts, which form the collecting system. Defective branching can lead to a multitude of kidney disorders including agenesis and reduced nephron number. The formation of branching tubules involves changes in cell shape, cell motility, and reorganization of the cytoskeleton. However, the exact intracellular mechanisms involved are far from understood. We have used the three-dimensional (3D) Madin-Darby canine kidney (MDCK) cell culture system to study how p21-activated kinase 1 (Pak1), which is an important regulator of the cytoskeleton, modulates branching. Our data reveal that Pak1 plays a crucial role in regulating branching morphogenesis. Expression of a dominant-negative Pak1 mutant (DN-Pak1) in MDCK cysts resulted in the spontaneous formation of extensions and branching tubules. Cellular contractility and levels of phosphorylated myosin light chain (pMLC) were increased in DN-Pak1 cells in collagen. Expression of a DN-Pak1 mutant that does not bind to PIX (DN-Pak1-DeltaPIX) failed to form extensions in collagen and did not have increased contractility. This shows that the DN-Pak1 mutant requires PIX binding to generate extensions and increased contractility in 3D culture. Furthermore, a beta1-integrin function-blocking antibody (AIIB2) inhibited the formation of branches and blocked the increased contractility in DN-Pak1 cysts. Taken together, our work shows that DN-Pak1-induced branching morphogenesis requires PIX binding and beta1-integrin signaling.
Collapse
Affiliation(s)
- Michael P Hunter
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
198
|
Afford SC, Kakoullis T, Oates J, Crocker J, Strain AJ. Effects of hepatocyte growth factor on differentiation and cMET receptor expression in the promyelocytic HL60 cell line. Mol Pathol 2010; 48:M23-7. [PMID: 16695971 PMCID: PMC407915 DOI: 10.1136/mp.48.1.m23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Aim-To determine the effects of hepatocyte growth factor (HGF) on myeloid cell differentiation and cMET expression using the promyelocytic HL60 cell line.Methods-HL60 cells cultured with purified recombinant HGF, dimethyl sulphoxide (DMSO), or 12-O tetradecanoylphorbol-13-acetate (TPA) were immunostained for the differentiation markers, human neutrophil elastase (HNE), cathepsin B, MAC387, or the receptor for hepatocyte growth factor (cMET).Results-HGF treated cells were positive on staining for cathepsin B and MAC387, but were negative for HNE, indicating monocytic differentiation. HGF treated cells had the morphology of monocytes but continued to divide at the same rate as control cells and remained non-adherent. DMSO treated cells were positive for HNE and cell numbers were reduced, confirming myeloid differentiation. TPA treated cells were positive for cathepsin B and MAC387, cell numbers were reduced, and the cells became adherent, confirming terminal monocytic differentiation. Untreated HL60 cells were weakly positive for cMET at the start of the culture period and expression increased after 72 hours. Cells treated with HGF, DMSO, or TPA were also positive for cMET.Conclusions-These data suggest that HGF induced partial monocytic differentiation in HL60 cells. In addition, expression of cMET by HL60 cells occurs at an early stage in myelomonocytic cells and is maintained after differentiation along either the myeloid or monocytic pathways.
Collapse
Affiliation(s)
- S C Afford
- The Liver Research Laboratories, Queen Elizabeth Hospital, Edgbaston, Birmingham B15 2TH
| | | | | | | | | |
Collapse
|
199
|
Zhang H, Lau SFT, Heng BF, Teo PY, Alahakoon PKDT, Ni M, Tasnim F, Ying JY, Zink D. Generation of easily accessible human kidney tubules on two-dimensional surfaces in vitro. J Cell Mol Med 2010; 15:1287-98. [PMID: 20586829 PMCID: PMC4373329 DOI: 10.1111/j.1582-4934.2010.01113.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The generation of tissue-like structures in vitro is of major interest for various fields of research including in vitro toxicology, regenerative therapies and tissue engineering. Usually 3D matrices are used to engineer tissue-like structures in vitro, and for the generation of kidney tubules, 3D gels are employed. Kidney tubules embedded within 3D gels are difficult to access for manipulations and imaging. Here we show how large and functional human kidney tubules can be generated in vitro on 2D surfaces, without the use of 3D matrices. The mechanism used by human primary renal proximal tubule cells for tubulogenesis on 2D surfaces appears to be distinct from the mechanism employed in 3D gels, and tubulogenesis on 2D surfaces involves interactions between epithelial and mesenchymal cells. The process is induced by transforming growth factor-β1, and enhanced by a 3D substrate architecture. However, after triggering the process, the formation of renal tubules occurs with remarkable independence from the substrate architecture. Human proximal tubules generated on 2D surfaces typically have a length of several millimetres, and are easily accessible for manipulations and imaging, which makes them attractive for basic research and in vitro nephrotoxicology. The experimental system described also allows for in vitro studies on how primary human kidney cells regenerate renal structures after organ disruption. The finding that human kidney cells organize tissue-like structures independently from the substrate architecture has important consequences for kidney tissue engineering, and it will be important, for instance, to inhibit the process of tubulogenesis on 2D surfaces in bioartificial kidneys.
Collapse
Affiliation(s)
- Huishi Zhang
- Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Hurley JR, Balaji S, Narmoneva DA. Complex temporal regulation of capillary morphogenesis by fibroblasts. Am J Physiol Cell Physiol 2010; 299:C444-53. [PMID: 20505042 DOI: 10.1152/ajpcell.00572.2009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interactions between endothelial and stromal cells are important for vascularization of regenerating tissue. Fibroblasts (FBs) are responsible for expression of angiogenic growth factors and matrix metalloproteinases, as well as collagen deposition and fibrotic myocardial remodeling. Recently, self-assembling peptide nanofibers were described as a promising environment for cardiac regeneration due to its synthetic nature and control over physiochemical properties. In this study, peptide nanofibers were used as a model system to quantify the dual role of fibroblasts in mediating angiogenesis chemically via expression of angiogenic factors and mechanically via cell-mediated scaffold disruption, extracellular matrix deposition, and remodeling. Human microvascular endothelial cells (ECs), FBs, or cocultures were cultured in three-dimensional nanofibers for up to 6 days. The peptide nanofiber microenvironment supported cell migration, capillary network formation, and cell survival in the absence of detectable scaffold contraction and proteolytic degradation. FBs enhanced early capillary network formation by "assisting" EC migration and increasing vascular endothelial growth factor and Angiopoietin-1 expression in a temporal manner. EC-FB interactions attenuated FB matrix metalloproteinase-2 expression while increasing collagen I deposition, resulting in greater construct stiffness and a more stable microenvironment in cocultures. Whereas FBs are critical for initial steps of angiogenesis in the absence of external angiogenic stimulation, coordinated efforts by ECs and FBs are required for a balance between cell-mediated scaffold disruption, extracellular matrix deposition, and remodeling at later time points. The findings of this study also emphasize the importance of developing a microenvironment that supports cell-cell interactions and cell migration, thus contributing toward an optimal environment for successful cardiac regeneration strategies.
Collapse
Affiliation(s)
- Jennifer R Hurley
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio 45221-0048, USA
| | | | | |
Collapse
|