101
|
Pilorget A, Conesa M, Sarray S, Michaud-Levesque J, Daoud S, Kim KS, Demeule M, Marvaldi J, El Ayeb M, Marrakchi N, Béliveau R, Luis J. Lebectin, aMacrovipera lebetina venom-derived C-type lectin, inhibits angiogenesis both in vitro and in vivo. J Cell Physiol 2007; 211:307-15. [PMID: 17323383 DOI: 10.1002/jcp.20935] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Integrins play an essential role in endothelial cell motility processes during angiogenesis and thus present interesting targets for the development of new anti-angiogenic agents. Snake venoms naturally contain a variety of proteins that can affect integrin-ligand interactions. Recently, the C-type lectin proteins (CLPs) have been characterized as efficient modulators of integrin functions. In this study, we investigated the anti-angiogenic activity of lebectin, a newly discovered CLP from Macrovipera lebetina venom. Human brain microvascular endothelial cells (HBMEC), used as an in vitro model, express alphavbeta3, alphavbeta5, and alpha5beta1 integrins, as well as the alpha2, alpha3, alpha6, and beta4 subunits. Our data show that lebectin acts as a very potent inhibitor (IC(50) approximately 0.5 nM) of HBMEC adhesion and migration on fibronectin by blocking the adhesive functions of both the alpha5beta1 and alphaV integrins. In addition, lebectin strongly inhibits both HBMEC in vitro tubulogenesis on Matrigel trade mark (IC(50) = 0.4 nM) and proliferation. Finally, using both a chicken CAM assay and a Matrigel trade mark Plug assay in nude mice, our results show that lebectin displays potent anti-angiogenic activity in vivo. Lebectin thus represents a new C-type lectin with anti-angiogenic properties with great potential for the treatment of angiogenesis-related diseases.
Collapse
MESH Headings
- Angiogenesis Inhibitors/isolation & purification
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Brain/blood supply
- Capillaries/cytology
- Capillaries/drug effects
- Cell Adhesion/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Chick Embryo
- Chorioallantoic Membrane/blood supply
- Chorioallantoic Membrane/drug effects
- Collagen
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Combinations
- Embryo Culture Techniques
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Fibronectins/pharmacology
- Humans
- Integrins/antagonists & inhibitors
- Integrins/metabolism
- Laminin
- Lectins, C-Type/isolation & purification
- Lectins, C-Type/physiology
- Lectins, C-Type/therapeutic use
- Mice
- Mice, Nude
- Neovascularization, Pathologic/chemically induced
- Neovascularization, Pathologic/prevention & control
- Neovascularization, Physiologic/drug effects
- Proteoglycans
- Subcutaneous Tissue/blood supply
- Time Factors
- Viper Venoms/isolation & purification
- Viper Venoms/pharmacology
- Viper Venoms/therapeutic use
- Viperidae
Collapse
Affiliation(s)
- Anthony Pilorget
- CNRS FRE 2737, Faculté de Pharmacie, bd Jean Moulin, Marseille, Cedex 5, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Simon-Assmann P, Turck N, Sidhoum-Jenny M, Gradwohl G, Kedinger M. In vitro models of intestinal epithelial cell differentiation. Cell Biol Toxicol 2006; 23:241-56. [PMID: 17171431 DOI: 10.1007/s10565-006-0175-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Accepted: 10/23/2006] [Indexed: 11/28/2022]
Abstract
The intestinal epithelium is a particularly interesting tissue as (1) it is in a constant cell renewal from a stem cell pool located in the crypts which form, with the underlying fibroblasts, a stem cell niche and (2) the pluripotent stem cells give rise to four main cell types: enterocytes, mucus, endocrine, and Paneth cells. The mechanisms leading to the determination of phenotype commitment and cell-specific expressions are still poorly understood. Although transgenic mouse models are powerful tools for elucidating the molecular cascades implicated in these processes, cell culture approaches bring easy and elegant ways to study cellular behavior, cell interactions, and cell signaling pathways for example. In the present review, we will describe the major tissue culture technologies that allow differentiation of epithelial cells from undifferentiated embryonic or crypt cells. We will point to the necessity of the re-creation of a complex microenvironment that allows full differentiation process to occur. We will also summarize the characteristics and interesting properties of the cell lines established from human colorectal tumors.
Collapse
|
103
|
Khalili P, Arakelian A, Chen G, Plunkett ML, Beck I, Parry GC, Doñate F, Shaw DE, Mazar AP, Rabbani SA. A non-RGD-based integrin binding peptide (ATN-161) blocks breast cancer growth and metastasis in vivo. Mol Cancer Ther 2006; 5:2271-80. [PMID: 16985061 DOI: 10.1158/1535-7163.mct-06-0100] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Integrins are expressed by numerous tumor types including breast cancer, in which they play a crucial role in tumor growth and metastasis. In this study, we evaluated the ability of ATN-161 (Ac-PHSCN-NH2), a 5-mer capped peptide derived from the synergy region of fibronectin that binds to alpha5beta1 and alphavbeta3 in vitro, to block breast cancer growth and metastasis. EXPERIMENTAL DESIGN MDA-MB-231 human breast cancer cells were inoculated s.c. in the right flank, or cells transfected with green fluorescent protein (MDA-MB-231-GFP) were inoculated into the left ventricle of female BALB/c nu/nu mice, resulting in the development of skeletal metastasis. Animals were treated with vehicle alone or by i.v. infusion with ATN-161 (0.05-1 mg/kg thrice a week) for 10 weeks. Tumor volume was determined at weekly intervals and tumor metastasis was evaluated by X-ray, microcomputed tomography, and histology. Tumors were harvested for histologic evaluation. RESULT Treatment with ATN-161 caused a significant dose-dependent decrease in tumor volume and either completely blocked or caused a marked decrease in the incidence and number of skeletal as well as soft tissue metastases. This was confirmed histologically as well as radiographically using X-ray and microcomputed tomography. Treatment with ATN-161 resulted in a significant decrease in the expression of phosphorylated mitogen-activated protein kinase, microvessel density, and cell proliferation in tumors grown in vivo. CONCLUSION These studies show that ATN-161 can block breast cancer growth and metastasis, and provides a rationale for the clinical development of ATN-161 for the treatment of breast cancer.
Collapse
Affiliation(s)
- Parisa Khalili
- Department of Medicine and Oncology, McGill University Health Center, Room H4.61, 687 Pine Avenue West, Montreal, Quebec, Canada H3A 1A1
| | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Rosanò L, Spinella F, Di Castro V, Dedhar S, Nicotra MR, Natali PG, Bagnato A. Integrin-linked kinase functions as a downstream mediator of endothelin-1 to promote invasive behavior in ovarian carcinoma. Mol Cancer Ther 2006; 5:833-42. [PMID: 16648553 DOI: 10.1158/1535-7163.mct-05-0523] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The endothelin-1 (ET-1) axis represents a novel target in several malignancies, including ovarian carcinoma. Upon being activated, the endothelin A receptor (ET(A)R) mediates multiple tumor-promoting activities, including mitogenesis, escape from apoptosis, angiogenesis, metastasis-related protease activation, epithelial-mesenchymal transition, and invasion. Integrin-linked kinase (ILK) is a multidomain focal adhesion protein that conveys intracellular signaling elicited by beta1-integrin and growth factor receptors. In this study, we investigate whether the signaling triggered by ET(A)R leading to an aggressive phenotype is mediated by an ILK-dependent mechanism. In HEY and OVCA 433 ovarian carcinoma cell lines, activation of ET(A)R by ET-1 enhances the expression of alpha2beta1 and alpha3beta1 integrins. ILK activity increases as ovarian cancer cells adhere to type I collagen through beta1 integrin signaling, and do so to a greater extent on ET-1 stimulation. ET-1 increases ILK mRNA and protein expression and activity in a time- and concentration-dependent manner. An ILK small-molecule inhibitor (KP-392) or transfection with a dominant-negative ILK mutant effectively blocks the phosphorylation of downstream signals, Akt and glycogen synthase kinase-3beta. The blockade of ET-1/ET(A)R-induced ILK activity results in an inhibition of matrix metalloproteinase activation as well as of cell motility and invasiveness in a phosphoinositide 3 kinase-dependent manner. In ovarian carcinoma xenografts, ABT-627, a specific ET(A)R antagonist, suppresses ILK expression, Akt and glycogen synthase kinase-3beta phosphorylation, and tumor growth. These data show that ILK functions as a downstream mediator of the ET-1/ET(A)R axis to potentiate aggressive cellular behavior. Thus, the ILK-related signaling cascade can be efficiently targeted by pharmacologic blockade of ET(A)R.
Collapse
Affiliation(s)
- Laura Rosanò
- Laboratory of Molecular Pathology and Ultrastructure, Regina Elena Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
105
|
Zeng ZZ, Jia Y, Hahn NJ, Markwart SM, Rockwood KF, Livant DL. Role of Focal Adhesion Kinase and Phosphatidylinositol 3′-Kinase in Integrin Fibronectin Receptor-Mediated, Matrix Metalloproteinase-1–Dependent Invasion by Metastatic Prostate Cancer Cells. Cancer Res 2006; 66:8091-9. [PMID: 16912186 DOI: 10.1158/0008-5472.can-05-4400] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
alpha(5)beta(1) Integrin interacts with the PHSRN sequence of plasma fibronectin, causing constitutive invasion by human prostate cancer cells. Inhibition of this process reduces tumorigenesis and prevents metastasis and recurrence. In this study, naturally serum-free basement membranes were used as in vitro invasion substrates. Immunoassays were employed to dissect the roles of focal adhesion kinase (FAK), phosphatidylinositol 3'-kinase (PI3K), and protein kinase Cdelta (PKC delta) in alpha(5)beta(1)-mediated, matrix metalloproteinase-1 (MMP-1)-dependent invasion by metastatic human DU 145 prostate cancer cells. We found that a peptide composed of the PHSRN sequence induced rapid FAK phosphorylation at Tyr(397) (Y397), a site whose phosphorylation is associated with kinase activation. The technique of RNA silencing [small interfering RNA (siRNA)] confirmed the role of FAK in PHSRN-induced invasion. PHSRN also induced the association of the p85-regulatory subunit of PI3K with FAK at a time corresponding to FAK phosphorylation and activation, and maximal PI3K activity occurred at this same time. The necessity of PI3K activity in both PHSRN-induced invasion and MMP-1 expression was confirmed by using specific PI3K inhibitors. By employing a specific inhibitor, Rottlerin, and by using siRNA, we also found that PKC delta, a PI3K substrate found in focal adhesions, functions in PHSRN-induced invasion. In addition, the induction of MMP-1 in PHSRN-treated DU 145 cells was shown by immunoblotting, and the role of MMP-1 in PHSRN-induced invasion was confirmed by the use of blocking anti-MMP-1 monoclonal antibody. Finally, a close temporal correspondence was observed between PHSRN-induced invasion and PHSRN-induced MMP-1 activity in DU 145 cells.
Collapse
Affiliation(s)
- Zhao-Zhu Zeng
- Department of Radiation Oncology and Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109-0582, USA
| | | | | | | | | | | |
Collapse
|
106
|
Cheng ZJ, Singh RD, Marks DL, Pagano RE. Membrane microdomains, caveolae, and caveolar endocytosis of sphingolipids. Mol Membr Biol 2006; 23:101-10. [PMID: 16611585 DOI: 10.1080/09687860500460041] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Caveolae are flask-shape membrane invaginations of the plasma membrane that have been implicated in endocytosis, transcytosis, and cell signaling. Recent years have witnessed the resurgence of studies on caveolae because they have been found to be involved in the uptake of some membrane components such as glycosphingolipids and integrins, as well as viruses, bacteria, and bacterial toxins. Accumulating evidence shows that endocytosis mediated by caveolae requires unique structural and signaling machinery (caveolin-1, src kinase), which indicates that caveolar endocytosis occurs through a mechanism which is distinct from other forms of lipid microdomain-associated, clathrin-independent endocytosis. Furthermore, a balance of glycosphingolipids, cholesterol, and caveolin-1 has been shown to be important in regulating caveolae endocytosis.
Collapse
Affiliation(s)
- Zhi-Jie Cheng
- Department of Biochemistry and Molecular Biology, Thoracic Diseases Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
107
|
Mitra A, Chakrabarti J, Banerji A, Das S, Chatterjee A. Culture of human cervical cancer cells, SiHa, in the presence of fibronectin activates MMP-2. J Cancer Res Clin Oncol 2006; 132:505-13. [PMID: 16788844 DOI: 10.1007/s00432-006-0096-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2005] [Accepted: 03/10/2006] [Indexed: 01/28/2023]
Abstract
PURPOSE Several studies indicate that integrin receptors are involved in the regulation of matrix metalloproteinase (MMP) expression. Integrin-ECM ligand interaction leads to phosphorylation of focal adhesion kinase (FAK) and activation of mitogen activated protein kinase pathways. In this present communication, we cultured human cervical cancer cells, SiHa, in the presence of fibronectin to study fibronectin-integrin mediated modulation of MMP activity. METHODS SiHa cells were cultured in serum-free medium (SFCM) in the presence of fibronectin, SFCM was collected and gelatin zymography was performed. Western blot, RT-PCR and immunocytochemistry were performed with SiHa cells cultured in the presence of fibronectin. RESULTS The culture of SiHa cells in the presence of 50 microg/1.5 ml fibronectin led to expression of pro-MMP-9 and activation of MMP-2 within 2 h. When cells were treated with ERK inhibitor (PD98059) and grown in the presence of fibronectin MMP-2 activation was partially inhibited, but when cells were treated with PI-3K inhibitor (LY294002) and grown in the presence of fibronectin MMP-2 activation was appreciably reduced. Tyrosine phosphorylation of FAK, PI-3K and ERK and nuclear trafficking of ERK were increased in SiHa cells grown in the presence of fibronectin. Increased MT1-MMP mRNA expression and processing of MT1-MMP were also observed in SiHa cells grown in the presence of fibronectin. CONCLUSIONS Our findings indicate that the culture of SiHa cells in SFCM in the presence of fibronectin perhaps generates a signalling cascade which leads to the expression of pro-MMP-9 and the activation of MMP-2 within 2 h. The signalling pathways activated seem to be the FAK/ERK/PI-3K pathway.
Collapse
Affiliation(s)
- Aparna Mitra
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata, 700 026, India
| | | | | | | | | |
Collapse
|
108
|
Elkaim R, Obrecht-Pflumio S, Tenenbaum H. Paxillin phosphorylation and integrin expression in osteoblasts infected by Porphyromonas gingivalis. Arch Oral Biol 2006; 51:761-8. [PMID: 16620778 DOI: 10.1016/j.archoralbio.2006.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 02/27/2006] [Accepted: 03/01/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE We investigated early biological events initiated by Porphyromonas gingivalis infection of human osteoblasts, focusing on tyrosine-phosphorylation and the expression of key components in focal adhesion and cell signalling. DESIGN Human primary osteoblasts were challenged for 1h with Porphyromonas gingivalis. Tyrosine-phosphorylation of paxillin and focal adhesion kinase (FAK) was examined by Western blotting. Changes in alpha3- and beta1-integrin mRNA expression were quantified by RT-PCR. RESULTS Tyrosine-phosphorylation of paxillin was proportional to the size of the Porphyromonas gingivalis inoculum. FAK, a potential kinase for paxillin, was not activated. The amount of alpha3- and beta1-integrins, determined by Western blotting, did not vary significantly, while the corresponding mRNA levels fell significantly when a large bacterial inoculum was used. CONCLUSIONS These results indicate that Porphyromonas gingivalis infection of osteoblasts in vitro triggers tyrosine-phosphorylation of paxillin but not FAK and modify alpha3- and beta1-integrin mRNA expression. This infection thus appears to have different effects on components with essential roles in focal adhesion (paxillin) and cell signalling (FAK and integrins).
Collapse
Affiliation(s)
- René Elkaim
- Parogène, 11 rue Humann, 67085 Strasbourg Cedex, France
| | | | | |
Collapse
|
109
|
Dehn D, Burbach GJ, Schäfer R, Deller T. NG2 upregulation in the denervated rat fascia dentata following unilateral entorhinal cortex lesion. Glia 2006; 53:491-500. [PMID: 16369932 DOI: 10.1002/glia.20307] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The chondroitin sulfate proteoglycan NG2 is a component of the glial scar following brain injury. Because of its growth inhibiting properties, it has been suggested to impede axonal regeneration. To study whether NG2 could also regulate axonal growth in denervated brain areas, changes in NG2 were studied in the rat fascia dentata following entorhinal deafferentation and were correlated with the post-lesional sprouting response. Laser microdissection was employed to selectively harvest the denervated molecular layer and combined with quantitative RT-PCR to measure changes in NG2 mRNA (6 h, 12 h, 2 days, 4 days, 7 days post-lesion). This revealed increases of NG2 mRNA at day 2 (2.5-fold) and day 4 (2-fold) post-lesion. Immunocytochemistry was used to detect changes in NG2 protein (1 days, 4 days, 7 days, 10 days, 14 days, 30 days, 6 months post-lesion). NG2 staining was increased in the denervated outer molecular layer at day 1 post-lesion, reached a maximum 10 days post-lesion, and returned to control levels thereafter. Electron microscopy revealed NG2 immunoprecipitate on glial surfaces and in the extracellular matrix around neuronal profiles, indicating that NG2 is secreted following denervation. Double labeling of NG2-immunopositive cells with markers for astrocytes, microglia/macrophages, and mature oligodendrocytes suggested that NG2 cells are a distinct glial subpopulation before and after entorhinal deafferentation. BrdU labeling revealed that some of the NG2-positive cells are generated post-lesion. Taken together, our data revealed a layer-specific upregulation of NG2 in the denervated fascia dentata that coincides with the sprouting response. This suggests that NG2 could regulate lesion-induced axonal growth in denervated areas of the brain.
Collapse
Affiliation(s)
- Doris Dehn
- Institute of Clinical Neuroanatomy, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | | | | | | |
Collapse
|
110
|
Bouvard D, Millon-Fremillon A, Dupe-Manet S, Block MR, Albiges-Rizo C. Unraveling ICAP-1 function: Toward a new direction? Eur J Cell Biol 2006; 85:275-82. [PMID: 16546571 DOI: 10.1016/j.ejcb.2005.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Cell adhesion to either the extracellular matrix (ECM) or to neighboring cells is of critical importance during both physiological and pathological situations. Integrins are a large family of cell adhesion receptors composed of two non-covalently linked alpha and beta subunits. They have a well-identified dual function of mediating both firm adhesion and signaling. The short cytoplasmic domain of integrin can interact with cytoplasmic proteins that are either shared by several different integrins or specific for one type of integrin. Integrin cytoplasmic domain-associated protein-1 (ICAP-1) is a small cytoplasmic protein that specifically interacts with the beta1 integrin subunit. In this review we will discuss recent findings on ICAP-1, not only at the structural and functional level, but also its possible interconnection in other signaling pathways such as those that control cell proliferation.
Collapse
Affiliation(s)
- Daniel Bouvard
- LEDAC, UMR CNRS/UJF 5538, Institut Albert Bonniot, Domaine de la Merci, Faculté de Médecine, F-38706 La Tronche Cedex, France.
| | | | | | | | | |
Collapse
|
111
|
Dutt S, Kléber M, Matasci M, Sommer L, Zimmermann DR. Versican V0 and V1 guide migratory neural crest cells. J Biol Chem 2006; 281:12123-31. [PMID: 16510447 DOI: 10.1074/jbc.m510834200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously showed the selective expression of the chondroitin sulfate proteoglycans versican V0 and V1 in barrier tissues that impede the migration of neural crest cells during embryonic trunk development (Landolt, R. M., Vaughan, L., Winterhalter, K. H., and Zimmermann, D. R. (1995) Development 212, 2303-2312). To test for an active involvement of these isoforms in the guidance process, we have now established protocols to isolate intact versican V0 and V1 in quantities sufficient for functional experiments. Using stripe choice assays, we demonstrate that pure preparations of either a mixture of versican V0/V1 or V1 alone strongly inhibit the migration of multipotent Sox10/p75NTR double-positive early neural crest stem cells on fibronectin by interfering with cell-substrate adhesion. We show that this inhibition is largely core glycoprotein-dependent, as the complete removal of the glycosaminoglycan chains has only a minor effect on the inhibitory capacity. Our findings support the notion that versican variants V0 and V1 act, possibly in concert with other inhibitory molecules such as aggrecan and ephrins, in directing the migratory streams of neural crest cells to their appropriate target tissues.
Collapse
Affiliation(s)
- Shilpee Dutt
- Laboratory of Molecular Biology, Department of Pathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
112
|
Hu B, Jarzynka MJ, Guo P, Imanishi Y, Schlaepfer DD, Cheng SY. Angiopoietin 2 induces glioma cell invasion by stimulating matrix metalloprotease 2 expression through the alphavbeta1 integrin and focal adhesion kinase signaling pathway. Cancer Res 2006; 66:775-83. [PMID: 16424009 PMCID: PMC2868063 DOI: 10.1158/0008-5472.can-05-1149] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Accumulating evidence reveals a significant correlation between angiopoietin 2 (Ang2) expression and tumor invasion and metastasis in various human cancers, but the major focus of recent studies has been on the angiogenic effects of Ang2. We recently reported that Ang2-stimulated glioma cell invasion results from the up-regulation and activation of matrix metalloprotease 2 (MMP-2) in tumor cells. In this study, we identify a novel mechanism by which Ang2 stimulates MMP-2 expression leading to glioma cell invasion. We show that Ang2 interacts with alpha(v)beta(1) integrin in Tie2-deficient human glioma cells, activating focal adhesion kinase (FAK), p130(Cas), extracellular signal-regulated protein kinase (ERK) 1/2, and c-jun NH(2)-terminal kinase (JNK) and substantially enhancing MMP-2 expression and secretion. The Ang2/alpha(v)beta(1) integrin signaling pathway was attenuated by functional inhibition of beta(1) and alpha(v) integrins, FAK, p130(Cas), ERK1/2, and JNK. Furthermore, expression of a negative regulator of FAK, FAK-related nonkinase, by U87MG/Ang2-expressing glioma xenografts suppressed Ang2-induced MMP-2 expression and glioma cell infiltration in the murine brain. These data establish a functional link between Ang2 interaction with alpha(v)beta(1) integrin and glioma cell invasion through the FAK/p130(Cas)/ERK1/2 and JNK-mediated signaling pathway.
Collapse
Affiliation(s)
- Bo Hu
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
- Department of Medicine, Research Pavilion at the Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Michael J. Jarzynka
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
- Department of Pathology, Research Pavilion at the Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Ping Guo
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
- Department of Pathology, Research Pavilion at the Hillman Cancer Center, Pittsburgh, Pennsylvania
- North China Coal Medical College, Tangshan, Hebei, P.R. China
| | - Yorihisha Imanishi
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
- Department of Pathology, Research Pavilion at the Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - David D. Schlaepfer
- Department of Immunology, The Scripps Research Institute, La Jolla, California
| | - Shi-Yuan Cheng
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
- Department of Pathology, Research Pavilion at the Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
113
|
Belvisi L, Riccioni T, Marcellini M, Vesci L, Chiarucci I, Efrati D, Potenza D, Scolastico C, Manzoni L, Lombardo K, Stasi MA, Orlandi A, Ciucci A, Nico B, Ribatti D, Giannini G, Presta M, Carminati P, Pisano C. Biological and molecular properties of a new alpha(v)beta3/alpha(v)beta5 integrin antagonist. Mol Cancer Ther 2006; 4:1670-80. [PMID: 16275988 DOI: 10.1158/1535-7163.mct-05-0120] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of the present study was to identify specific alpha(v)beta3/alpha(v)beta5 integrin antagonists active on tumor-induced angiogenesis. To this purpose, in vitro integrin-binding assays were used to screen a library of conformationally constrained bicyclic lactam Arg-Gly-Asp-containing pseudopeptides. The results identified ST1646 as a high-affinity specific ligand for alpha(v)beta3 and alpha(v)beta5 integrins with negligible interacting with alpha5beta1 integrin. In all the assays, ST1646 was equipotent to or more potent than the well-characterized integrin antagonists c(RGDfV) and cyclo(Arg-Gly-Asp-d-Phe-[NMe]Val) (EMD121974). In the chorioallantoic membrane assay, topical administration of ST1646 was able to prevent the angiogenic responses elicited by recombinant fibroblast growth factor-2 or vascular endothelial growth factor. In addition, systemic administration of ST1646 in mice exerted a significant antiangiogenic activity on neovascularization triggered by mammary carcinoma MDA-MB435 cells implanted s.c. in a dorsal air sac via a (Millipore Filter Corporation, Bedford, MA) chamber. Moreover, ST1646 delivery via an osmotic pump inhibited the growth and vascularization of tumor xenografts originating from the injection of alpha(v)beta3/alpha(v)beta5-expressing human ovarian carcinoma cells in nude mice. In agreement with the biochemical and pharmacologic studies, Monte Carlo/Stochastic Dynamics simulation showed that the bicyclic scaffold in ST1646 forced the compound to assume a preferred conformation superimposable to the X-ray conformation of alpha(v)beta3-bound EMD121974. Accordingly, computer-docking studies indicated that the ST1646-alpha(v)beta3 integrin complex maintains the ligand-receptor distances and interactions observed in the crystalline EMD121974-alpha(v)beta3 integrin complex. Taken together, these observations indicate that ST1646 represents a dual alpha(v)beta3/alpha(v)beta5 integrin antagonist with interesting biochemical and biological features to be tested in cancer therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Arginine/chemistry
- Aspartic Acid/chemistry
- Cattle
- Cell Adhesion
- Cell Line, Tumor
- Cell Proliferation
- Chickens
- Crystallography, X-Ray
- Dose-Response Relationship, Drug
- Endothelium, Vascular/cytology
- Fibroblast Growth Factor 2/metabolism
- Glycine/chemistry
- Guinea Pigs
- Humans
- Inhibitory Concentration 50
- Integrin alphaVbeta3/antagonists & inhibitors
- Integrins/antagonists & inhibitors
- Integrins/metabolism
- Ligands
- Mice
- Mice, Nude
- Microcirculation
- Models, Chemical
- Models, Molecular
- Molecular Conformation
- Monte Carlo Method
- Neoplasm Transplantation
- Neovascularization, Pathologic
- Oligopeptides/chemistry
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/pharmacology
- Platelet Aggregation
- Protein Binding
- Protein Conformation
- Receptors, Vitronectin/antagonists & inhibitors
- Recombinant Proteins/chemistry
- Stochastic Processes
- Vascular Endothelial Growth Factor A/metabolism
- Vitronectin/chemistry
Collapse
Affiliation(s)
- Laura Belvisi
- Organic and Industrial Chemistry Department, Centre for Biomolecular Interdisciplinary Studies and Industrial Applications, University of Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Goel HL, Moro L, King M, Teider N, Centrella M, McCarthy TL, Holgado-Madruga M, Wong AJ, Marra E, Languino LR. β1Integrins Modulate Cell Adhesion by Regulating Insulin-Like Growth Factor-II Levels in the Microenvironment. Cancer Res 2006; 66:331-42. [PMID: 16397247 DOI: 10.1158/0008-5472.can-05-2588] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The interactions between cancer cells and the extracellular matrix (ECM) regulate cancer progression. The beta1C and beta1A integrins, two cytoplasmic variants of the beta1 integrin subfamily, are differentially expressed in prostate cancer. Using gene expression analysis, we show here that the beta1C variant, an inhibitor of cell proliferation, which is down-regulated in prostate cancer, up-regulates insulin-like growth factor-II (IGF-II) mRNA and protein levels. In contrast, beta1A does not affect IGF-II levels. We provide evidence that beta1C-mediated up-regulation of IGF-II levels increases adhesion to Laminin-1, a basement membrane protein down-regulated in prostate cancer, and that the beta1C cytoplasmic domain contains the structural motif sufficient to increase cell adhesion to Laminin-1. This autocrine mechanism that locally supports cell adhesion to Laminin-1 via IGF-II is selectively regulated by the beta1 cytoplasmic domain via activation of the growth factor receptor binding protein 2-associated binder-1/SH2-containing protein-tyrosine phosphatase 2/phosphatidylinositol 3-kinase pathway. Thus, the concurrent local loss of beta1C integrin, of its ligand Laminin-1, and of IGF-II in the tumor microenvironment may promote prostate cancer cell invasion and metastasis by reducing cancer cell adhesive properties. It is, therefore, conceivable that reexpression of beta1C will be sufficient to revert a neoplastic phenotype to a nonproliferative and highly adherent normal phenotype.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology and the Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Sharma DK, Brown JC, Cheng Z, Holicky EL, Marks DL, Pagano RE. The glycosphingolipid, lactosylceramide, regulates beta1-integrin clustering and endocytosis. Cancer Res 2005; 65:8233-41. [PMID: 16166299 DOI: 10.1158/0008-5472.can-05-0803] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glycosphingolipids are known to play roles in integrin-mediated cell adhesion and migration; however, the mechanisms by which glycosphingolipids affect integrins are unknown. Here, we show that addition of the glycosphingolipid, C8-lactosylceramide (C8-LacCer), or free cholesterol to human fibroblasts at 10 degrees C causes the formation of glycosphingolipid-enriched plasma membrane domains as shown by visualizing a fluorescent glycosphingolipid probe, BODIPY-LacCer, incorporated into the plasma membrane of living cells. Addition of C8-LacCer or cholesterol to cells initiated the clustering of beta1-integrins within these glycosphingolipid-enriched domains and the activation of the beta1-integrins as assessed using a HUTS antibody that only binds activated integrin. On warming to 37 degrees C, beta1-integrins were rapidly internalized via caveolar endocytosis in cells treated with C8-LacCer or cholesterol, whereas little beta1-integrin was endocytosed in untreated fibroblasts. Incubation of cells with C8-LacCer or cholesterol followed by warm-up caused src activation, a reorganization of the actin cytoskeleton, translocation of RhoA GTPase away from the plasma membrane as visualized using total internal reflection fluorescence microscopy, and transient cell detachment. These studies show that LacCer can regulate integrin function both by modulating integrin clustering in microdomains and by regulating integrin endocytosis via caveolae. Our findings suggest the possibility that aberrant levels of glycosphingolipids found in cancer cells may influence cell attachment events by direct effects on integrin clustering and internalization.
Collapse
Affiliation(s)
- Deepak K Sharma
- Department of Biochemistry and Molecular Biology, Thoracic Diseases Research Unit, and Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, Minnesota 55905-0001, USA
| | | | | | | | | | | |
Collapse
|
116
|
Wang D, Anderson JC, Gladson CL. The role of the extracellular matrix in angiogenesis in malignant glioma tumors. Brain Pathol 2005; 15:318-26. [PMID: 16389944 PMCID: PMC8095805 DOI: 10.1111/j.1750-3639.2005.tb00117.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Angiogenesis is a promising target for the development of effective strategies for the treatment of malignant brain tumors in that it has the potential to starve large tumors and prevent the regrowth of residual margins. Two critical steps in angiogenesis, the proliferation of activated endothelial cells and their migration into the perivascular space (sprouting), require adherence of the endothelial cells to the extracellular matrix (ECM). Thus, the availability of the appropriate ligands within the ECM contributes to the regulation of angiogenesis. In addition, several components of the ECM can act through other mechanisms to further promote angiogenesis or inhibit it. Current evidence suggests that the regulation of angiogenesis is a dynamic process in which the endothelial cells can promote angiogenesis by secreting proteases that remodel the ECM, tumor cells can further promote angiogenesis by secreting ECM components and actively remodeling their environment, and stromal cells may respond to angiogenesis associated with tumors and inflammatory reactions by secreting inhibitory molecules. Here, we provide a critical review of the protein and proteoglycan components of the ECM that have been implicated in angiogenesis with an emphasis on their role in promoting or inhibiting angiogenesis in brain tumors.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| | - Joshua C. Anderson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| | - Candece L. Gladson
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham
| |
Collapse
|
117
|
Abdollahi A, Griggs DW, Zieher H, Roth A, Lipson KE, Saffrich R, Gröne HJ, Hallahan DE, Reisfeld RA, Debus J, Niethammer AG, Huber PE. Inhibition of αvβ3 Integrin Survival Signaling Enhances Antiangiogenic and Antitumor Effects of Radiotherapy. Clin Cancer Res 2005; 11:6270-9. [PMID: 16144931 DOI: 10.1158/1078-0432.ccr-04-1223] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The involvement of alpha(v)beta3 and alpha(v)beta5 integrins in angiogenesis and the use of integrin antagonists as effective antiangiogenic agents are documented. Radiotherapy is an important therapy option for cancer. It has been shown that ionizing radiation exerts primarily antiangiogenic effects in tumors but has also proangiogenic effects as the reaction of the tumor to protect its own vasculature from radiation damage. Here, we show that combined treatment with S247, an Arg-Gly-Glu peptidomimetic antagonist of alpha(v)beta3 integrin, and external beam radiotherapy are beneficial in local tumor therapy. We found that radiation up-regulates alpha(v)beta3 expression in endothelial cells and consecutively phosphorylates Akt, which may provide a tumor escape mechanism from radiation injury mediated by integrin survival signaling. In the presence of S247, the radiation-induced Akt phosphorylation is strongly inhibited. Our studies on endothelial cell proliferation, migration, tube formation, apoptosis, and clonogenic survival show that the radiosensitivity of endothelial cells is enhanced by the concurrent administration of the integrin antagonist. The in vitro data are successfully translated into human glioma (U87), epidermoid (A431), and prostate cancer (PC3) xenograft models growing s.c. on BALB/c-nu/nu mice. In vivo, the combination of S247 treatment and fractionated radiotherapy (5 x 2.5 Gy) leads to enhanced antiangiogenic and antitumor effects compared with either monotherapies. These results underline the importance of alpha(v)beta3 integrin when tumors protect their microvasculature from radiation-induced damage. The data also indicate that the combination of integrin antagonists and radiotherapy represents a rational approach in local cancer therapy.
Collapse
Affiliation(s)
- Amir Abdollahi
- Department of Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Affiliation(s)
- C Brakebusch
- Max Planck Institute of Biochemistry, Junior Group Regulation of Cytoskeletal Organization, Martinsried, Germany
| | | |
Collapse
|
119
|
Abu-Ali S, Sugiura T, Takahashi M, Shiratsuchi T, Ikari T, Seki K, Hiraki A, Matsuki R, Shirasuna K. Expression of the urokinase receptor regulates focal adhesion assembly and cell migration in adenoid cystic carcinoma cells. J Cell Physiol 2005; 203:410-9. [PMID: 15521066 DOI: 10.1002/jcp.20242] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adenoid cystic carcinoma (AdCC) cell lines (ACCS and ACCT) showed higher migration responses and adhesion to the extracellular matrix (ECM), especially types I and IV collagen, than did the oral squamous cell carcinoma (SCC) lines (NA and TF). The response to collagens was largely and exclusively inhibited by anti-alpha(2) integrin antibody. Moreover, AdCC cell lines expressed higher surface levels of urokinase-type plasminogen activator receptor (uPAR) than did SCC cell lines. When AdCC cells were plated on collagen, the surface level of uPAR was increased, and numerous focal adhesions consisting of uPAR, vinculin, and paxillin were assembled; whereas collagen-stimulated SCC cell counterparts or AdCC cells plated on other types of ECM, such as fibronectin, failed to assemble such definite focal adhesions. In order to elucidate the association of uPAR with collagen-induced events, an ACCS-AS cell line transfected with a vector expressing antisense uPAR RNA was established and shown to have reduced uPAR (about 10% that of parental ACCS at both the protein and mRNA levels). ACCS-AS showed a strong reduction of collagen-stimulated migration and focal adhesion assembly of alpha(2) integrin, vinculin, and paxillin. These findings suggest that AdCC has a proclivity for migrating to types I and IV collagens due to the overexpression of uPAR, which plays a key role in focal adhesion assembly and migration.
Collapse
Affiliation(s)
- Samah Abu-Ali
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Liang X, Zhou Q, Li X, Sun Y, Lu M, Dalton N, Ross J, Chen J. PINCH1 plays an essential role in early murine embryonic development but is dispensable in ventricular cardiomyocytes. Mol Cell Biol 2005; 25:3056-62. [PMID: 15798193 PMCID: PMC1069610 DOI: 10.1128/mcb.25.8.3056-3062.2005] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PINCH1, an adaptor protein composed of five LIM domains, mediates protein-protein interactions and functions as a component of the integrin-integrin-linked kinase (ILK) complex. The integrin-ILK signaling complex plays a pivotal role in cell motility, proliferation, and survival during embryonic development of many animal species. To elucidate the physiological function of PINCH1 in mouse embryonic development, we have deleted the mouse PINCH1 gene by homologous recombination. Mice heterozygous for PINCH1 are viable and indistinguishable from wild-type littermates. However, no viable homozygous offspring were observed from PINCH1+/- intercrosses. Histological analysis of homozygous mutant embryos revealed that they had a disorganized egg cylinder by E5.5, which degenerated by E6.5. Furthermore, E5.5 PINCH1-/- embryos exhibited decreased cell proliferation and excessive cell death. We have also generated and analyzed mice in which PINCH1 has been specifically deleted in ventricular cardiomyocytes. These mice exhibit no basal phenotype, with respect to mouse survival, cardiac histology, or cardiac function as measured by echocardiography. Altogether, these data indicate that PINCH1 plays an essential role in early murine embryonic development but is dispensable in ventricular cardiomyocytes.
Collapse
Affiliation(s)
- Xingqun Liang
- Department of Medicine, University of California at San Diego, School of Medicine, 9500 Gilman Dr., La Jolla, CA 92093-0613, USA
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Abstract
The movement of cancer cells into tissue surrounding the tumour and the vasculature is the first step in the spread of metastatic cancers. Recent advances in imaging, the use of 3D model systems and the application of microarray technologies have yielded new insights into these processes. This work has challenged our views about what causes cancer cells to become motile in the first place, and has demonstrated that cancer cells can move in many different ways.
Collapse
Affiliation(s)
- Erik Sahai
- Tumour Cell Biology Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, UK.
| |
Collapse
|
122
|
Banas B, Wörnle M, Merkle M, Gonzalez-Rubio M, Schmid H, Kretzler M, Pietrzyk MC, Fink M, Perez de Lema G, Schlöndorff D. Binding of the chemokine SLC/CCL21 to its receptor CCR7 increases adhesive properties of human mesangial cells. Kidney Int 2005; 66:2256-63. [PMID: 15569314 DOI: 10.1111/j.1523-1755.2004.66037.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Adherence of human mesangial cells to the surrounding matrix contributes to glomerular homeostasis and is important for the maintenance of glomerular architecture and function in normal adult human kidney. The expression of chemokines and corresponding chemokine receptors on adjacent intrinsic renal cells indicates a novel chemokine/chemokine receptor function on nonimmune cells important for glomerular homeostasis. A constitutive expression of the chemokine SLC/CCL21 on human podocytes and of its corresponding receptor CCR7 on mesangial cells was shown before. SLC/CCL21 has a positive effect on proliferation and migration of mesangial cells and leads to increased cell survival in Fas-induced apoptosis. In leukocytes chemokines mediate integrin-dependent firm adhesion. Therefore, we examined the influence of chemokine receptor CCR7 activation by SLC/CCL21 on adhesive properties of human mesangial cells to matrix molecules. METHODS Adhesion assays, mechanical detachment assays, and evaluation of integrin activation by integrin-linked kinase activity were performed. Changes in the cytoskeletal F-actin were illustrated by phalloidin immunofluorescence staining. RESULTS SLC/CCL21 stimulation enhanced adhesiveness to fibronectin in a time- and concentration-dependent manner. SLC/CCL21 also increased the firmness of mesangial cells adhesion as judged by detachment assays. Furthermore activation of integrin-linked kinase occurred with SLC/CCL21 addition to mesangial cells, resulting in increased phosphorylation of glycogen synthase kinase-3 (GSK-3) and protein kinase B (PKB/Akt). Exposure of mesangial cells to SLC/CCL21 also resulted in F-actin rearrangements with membrane ruffling and extensions leading to bridging between mesangial cells. CONCLUSION Activation of CCR7 on mesangial cells by SLC/CCL21 enhances the degree and firmness of cell adhesion and increases cell spreading and the formation of cell-cell contacts. This includes integrin-linked kinase activation and F-actin rearrangements. Thus, local chemokine generation and chemokine receptor expression on mesangial cells may play an important role in the maintenance of glomerular homeostasis and in local remodeling processes.
Collapse
Affiliation(s)
- Bernhard Banas
- Nephrologisches Zentrum, Medizinische Poliklinik, Ludwig-Maximilians-Universität München, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Meyer A, van Golen CM, Boyanapalli M, Kim B, Soules ME, Feldman EL. Integrin-linked kinase complexes with caveolin-1 in human neuroblastoma cells. Biochemistry 2005; 44:932-8. [PMID: 15654749 DOI: 10.1021/bi048619r] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Integrin-linked kinase (ILK) and caveolin-1 (cav-1) are implicated in the pathogenesis of cancer. Overexpression of ILK leads to altered expression of cell cycle regulators, a decreased level of cell adhesion to the extracellular matrix, a decreased level of apoptosis, in vitro phosphorylation of Akt, and tumor formation in nude mice. Conversely, cav-1 expression is frequently downregulated in many forms of cancer. We examined whether ILK and cav-1 interact in SHEP human neuroblastoma cells because ILK is present in caveolae-enriched membranes and contains a putative cav-binding domain. SHEP cells were stably transfected with vector, wild-type ILK (ILK-wt), kinase-deficient ILK (ILK-kd), or mutant cav-binding domain ILK (ILK-mutCavbd). Control SHEP cells and ILK transfectants express high levels of ILK and cav-1. Immunoprecipitation with anti-cav-1 co-immunoprecipitates a 59 kDa protein that is immunoreactive with the anti-ILK antibody, and this interaction is partially prevented in cells expressing ILK-mutCavbd. Cav-1 and ILK partially colocalize in SHEP cells, also supporting these data. Last, affinity chromatography with a biotinylated cav-scaffolding domain peptide precipitates ILK-wt but not ILK-mutCavbd. These data suggest that the cav-binding domain of ILK and the cav-scaffolding domain of cav-1 mediate complex formation in human neuroblastoma cells.
Collapse
Affiliation(s)
- Amy Meyer
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | |
Collapse
|
124
|
Meyer A, van Golen CM, Kim B, van Golen KL, Feldman EL. Integrin expression regulates neuroblastoma attachment and migration. Neoplasia 2005; 6:332-42. [PMID: 15256055 PMCID: PMC1502107 DOI: 10.1593/neo.03445] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neuroblastoma (NBL) is the most common malignant disease of infancy, and children with bone metastasis have a mortality rate greater than 90%. Two major classes of proteins, integrins and growth factors, regulate the metastatic process. We have previously shown that tumorigenic NBL cells express higher levels of the type I insulin-like growth factor receptor (IGF-IR) and that beta1 integrin expression is inversely proportional to tumorigenic potential in NBL. In the current study, we analyze the effect of beta1 integrin and IGF-IR on NBL cell attachment and migration. Nontumorigenic S-cells express high levels of beta1 integrin, whereas tumorigenic N-cells express little beta1 integrin. Alterations in beta1 integrin are due to regulation at the protein level, as translation is decreased in N-type cells. Moreover, inhibition of protein synthesis shows that beta1 integrin is degraded more slowly in S-type cells (SHEP) than in N-type cells (SH-SY5Y and IMR32). Inhibition of alpha5beta1 integrin prevents SHEP (but not SH-SY5Y or IMR32) cell attachment to fibronectin and increases SHEP cell migration. Increases in IGF-IR decrease beta1 integrin expression, and enhance SHEP cell migration, potentially through increased expression of alphavbeta3. These data suggest that specific classes of integrins in concert with IGF-IR regulate NBL attachment and migration.
Collapse
Affiliation(s)
- Amy Meyer
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth L. van Golen
- Department of Internal Medicine, Hematology and Oncology Division, University of Michigan, Ann Arbor, MI, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
125
|
Cho HJ, Youn SW, Cheon SI, Kim TY, Hur J, Zhang SY, Lee SP, Park KW, Lee MM, Choi YS, Park YB, Kim HS. Regulation of endothelial cell and endothelial progenitor cell survival and vasculogenesis by integrin-linked kinase. Arterioscler Thromb Vasc Biol 2005; 25:1154-60. [PMID: 15802621 DOI: 10.1161/01.atv.0000164312.20008.93] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE New vessel formation is a dynamic process of attachment, detachment, and reattachment of endothelial cells (ECs) and endothelial progenitor cells (EPCs) with each other and with the extracellular matrix (ECM). Integrin-linked kinase (ILK) plays a pivotal role in ECM-mediated signaling. Therefore, we investigated the role of ILK in ECs and EPCs during neovascularization. METHODS AND RESULTS In human umbilical cord vein ECs and EPCs, endogenous ILK expression, along with subsequent cell survival signals phospho-Akt and phospho-glycogen synthase kinase 3beta, was reduced after anchorage or nutrient deprivation. Even brief anchorage deprivation resulted in retarded capillary tube formation by ECs. Adenoviral ILK gene transfer in ECs and EPCs reversed the decrease in cell survival signals after anchorage or nutrient deprivation, leading to enhanced survival, reduced apoptosis, and significantly accelerated the functional recovery after reattachment. And ILK overexpressing EPCs significantly improved blood flow recovery and prevented limb loss in nude mice hindlimb ischemia model. Furthermore, the efficacy of systemic delivery was equivalent to local injection of ILK-EPCs. CONCLUSIONS ILK overexpression protects ECs and EPCs from anchorage- or nutrient-deprived stress and enhances neovascularization, suggesting that ILK is an optimal target gene for genetically modified cell-based therapy. Neovascularization is a dynamic process of detachment and reattachment of ECs and EPCs. Endogenous ILK expression was decreased in various stress conditions, and the gene transfer of ILK protected ECs and EPCs from temporary anchorage or nutrient deprivation. Furthermore, ILK gene transfer in EPCs significantly enhanced neovascularization in vivo.
Collapse
Affiliation(s)
- Hyun-Jai Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Arbel-Goren R, Levy Y, Ronen D, Zick Y. Cyclin-dependent kinase inhibitors and JNK act as molecular switches, regulating the choice between growth arrest and apoptosis induced by galectin-8. J Biol Chem 2005; 280:19105-14. [PMID: 15753078 DOI: 10.1074/jbc.m502060200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Galectin-8, a mammalian beta-galactoside binding lectin, functions as an extracellular matrix protein that forms high affinity interactions with integrins. Here we demonstrated that soluble galectin-8 inhibits cell cycle progression and induces growth arrest. These effects cannot be attributed to interference with cell adhesion but can be attributed to a 4-5-fold increase in the cellular content of the cyclin-dependent kinase inhibitor p21, which was already evident following a 4-h incubation of H1299 cells with galectin-8. The increase in p21 levels was preceded by a 3-5-fold increase in JNK and protein kinase B (PKB) activities. Accordingly, SP600125, the inhibitor of JNK, and wortmannin, the inhibitor of phosphatidylinositol 3-kinase, which is the upstream activator of PKB, inhibited the increase in the cellular content of p21. Furthermore, overexpression of a dominant inhibitory form of SEK1, the upstream kinase regulator of JNK, inhibited both JNK activation and p21 accumulation. When p21 expression was inhibited by cycloheximide, galectin-8 directed the cells toward apoptosis, which involves induction of poly(ADP-ribose) polymerase cleavage. Indeed, galectin-8-induced apoptosis was 2-fold higher in HTC (p21-null) cells when compared with parental HTC cells. Because overexpression of galectin-8 attenuates the rate of DNA synthesis, stable colonies that overexpress and secrete galectin-8 can be generated only in cells overexpressing a growth factor receptor, such as the insulin receptor. These results implicate galectin-8 as a modulator of cellular growth through up-regulation of p21. This process involves activation of JNK, which enhances the synthesis of p21, combined with the activation of PKB, which inhibits p21 degradation. These effects of the lectin depended upon protein-sugar interactions and were induced when galectin-8 was present as a soluble ligand or when it was overexpressed in cells.
Collapse
Affiliation(s)
- Rinat Arbel-Goren
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | |
Collapse
|
127
|
Gruber G, Hess J, Stiefel C, Aebersold DM, Zimmer Y, Greiner RH, Studer U, Altermatt HJ, Hlushchuk R, Djonov V. Correlation between the tumoral expression of beta3-integrin and outcome in cervical cancer patients who had undergone radiotherapy. Br J Cancer 2005; 92:41-6. [PMID: 15597101 PMCID: PMC2361731 DOI: 10.1038/sj.bjc.6602278] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Integrins are cell-surface receptors, which mediate cell-to-cell and cell-to-extracellular matrix adhesion. Besides playing an important role in tumour angiogenesis, β3-integrin is also expressed in several types of epithelial cancer cells. It was the purpose of the present study to evaluate the prognostic value of β3-integrin expression in patients with cervical cancer. Biopsies were taken from 82 patients with squamous cell or adenocarcinomas of the uterine cervix who had undergone external-beam radiotherapy with or without brachytherapy. These tissue samples were analysed immunohistochemically for the expression of β3-integrin. The impact of immunoreactivity for β3-integrin on survival end points was assessed by univariate and multivariate analyses, and its correlation with clinicopathological characteristics evaluated by crosstabulations. β3-integrin was expressed in 61% (50 of 82) of the patients. Kaplan–Meier curves revealed local progression-free survival, distant metastasis-free survival and cause-specific survival to be significantly shorter (P-values according to the log-rank test: 0.002, 0.04 and 0.01, respectively) in patients with β3-integrin expression. The prognostic impact of this parameter was even higher than for other well-known prognostic parameters and remained statistically significant in the multivariate analyses. β3-integrin, which is expressed in the majority of patients with advanced cervical cancer, has a significant prognostic impact on outcome according to univariate and multivariate analyses.
Collapse
Affiliation(s)
- G Gruber
- Department of Radiation Oncology, University of Bern, Switzerland
| | - J Hess
- Department of Radiation Oncology, University of Bern, Switzerland
| | - C Stiefel
- Department of Radiation Oncology, University of Bern, Switzerland
| | - D M Aebersold
- Department of Radiation Oncology, University of Bern, Switzerland
| | - Y Zimmer
- Department of Radiation Oncology, University of Bern, Switzerland
| | - R H Greiner
- Department of Radiation Oncology, University of Bern, Switzerland
| | - U Studer
- Department of Radiation Oncology, University of Bern, Switzerland
| | | | - R Hlushchuk
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, CH-3000 Bern 9, Switzerland
| | - V Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, CH-3000 Bern 9, Switzerland
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, CH-3000 Bern 9, Switzerland. E-mail:
| |
Collapse
|
128
|
Bengtsson T, Aszodi A, Nicolae C, Hunziker EB, Lundgren-Akerlund E, Fässler R. Loss of alpha10beta1 integrin expression leads to moderate dysfunction of growth plate chondrocytes. J Cell Sci 2005; 118:929-36. [PMID: 15713743 DOI: 10.1242/jcs.01678] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integrin alpha10beta1 is a collagen-binding integrin expressed on chondrocytes. In order to unravel the role of the alpha10 integrin during development, we generated mice carrying a constitutive deletion of the alpha10 integrin gene. The mutant mice had a normal lifespan and were fertile but developed a growth retardation of the long bones. Analysis of the skeleton revealed defects in the growth plate after birth characterized by a disturbed columnar arrangement of chondrocytes, abnormal chondrocyte shape and reduced chondrocyte proliferation. Electron microscopy of growth plates from newborn mice revealed an increased number of apoptotic chondrocytes and reduced density of the collagen fibrillar network compared to these structures in control mice. These results demonstrate that integrin alpha10beta1 plays a specific role in growth plate morphogenesis and function.
Collapse
Affiliation(s)
- Therese Bengtsson
- Department of Experimental Pathology, Lund University Hospital, Box 117, 22185 Lund, Sweden
| | | | | | | | | | | |
Collapse
|
129
|
Gontero P, Banisadr S, Frea B, Brausi M. Metastasis markers in bladder cancer: a review of the literature and clinical considerations. Eur Urol 2005; 46:296-311. [PMID: 15306099 DOI: 10.1016/j.eururo.2004.04.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2004] [Indexed: 12/22/2022]
Abstract
Cancer invasion and metastasis develop through a sequence of processes involving loss of cell-cell and cell-matrix adhesions, proteolysis and induction of angiogenesis. We reviewed the current literature on the molecules that have been shown to play a significant role in these three steps of metastatisation in bladder cancer (BC) cells and their host microenvironment. Particular emphasis was given to markers that are assessable through immunohistochemistry and for which an additional prognostic value over the TNM variables has been recognized, in order to identify a subset of tumour markers readily available for application in daily clinical practice. We conclude that markers such as E-cadherin, Sialosyl-LeX, laminin, collagen IV, TSP-1 and MVD are useful prognostic markers, alpha, beta, and gamma catenin, MMP-2 and -9, uPAR, PD-ECGF and Bfgf can be considered potentially useful, while research on CD44, MMP-1 and -3, uPA, cathepsin D and VEGF has proved inconclusive. Further research in this field should concentrate on the molecules listed in the first group.
Collapse
Affiliation(s)
- Paolo Gontero
- Department of Urology, Università del Piemonte Orientale, Novara, Italy.
| | | | | | | |
Collapse
|
130
|
Pereira AL, Veras SS, Silveira ÉJ, Seabra FR, Pinto LP, Souza LB, Freitas RA. O papel das proteínas da matriz extracelular e das metaloproteinases em carcinomas de cabeça e pescoço: uma atualização bibliográfica. ACTA ACUST UNITED AC 2005. [DOI: 10.1590/s0034-72992005000100014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interações entre células neoplásicas e constituintes da matriz extracelular (MEC) interferem fortemente no desenvolvimento tumoral, incluindo os localizados em cabeça e pescoço, pois influenciam a proliferação e sobrevivência celular, bem como a sua capacidade de migrar do sítio primário para outros tecidos e formar metástases. Essa migração celular é facilitada pela destruição parcial da MEC, a qual é realizada pelas metaloproteinases (MMPs), que representam uma família de mais de vinte endopeptidases, com atividade controlada pela expressão de inibidores específicos (TIMPs). Diversos estudos utilizando-se de marcadores para constituintes da MEC bem como pelas MMPs têm fornecido informações adicionais sobre o diagnóstico e prognóstico em carcinomas de cabeça e pescoço. Nesta revisão consideraremos o papel da MEC e das MMPs na progressão desses tumores, enfatizando que não somente a degradação proteolítica está envolvida neste processo, como também interações entre vários constituintes da MEC fornecem substrato para regulação e crescimento destes tumores.
Collapse
Affiliation(s)
| | - Simone S.L. Veras
- Universidade Federal do Rio Grande do Norte; Universidade Federal do Piauí
| | | | | | | | | | | |
Collapse
|
131
|
Bialkowska K, Saido TC, Fox JEB. SH3 domain of spectrin participates in the activation of Rac in specialized calpain-induced integrin signaling complexes. J Cell Sci 2005; 118:381-95. [PMID: 15632109 DOI: 10.1242/jcs.01625] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In this study, we used cultured cells spreading on beta3 integrin substrates to examine the possibility that spectrin is involved in signal transduction. Spectrin clustered with specialized calpain-induced beta3 integrin signaling complexes that mediate the initial attachment of cells and initiate Rac activation and lamellipodia extension. It was absent from focal complexes and focal adhesions, the integrin complexes that mediate adhesion in lamellipodia and fully spread cells. Spectrin contains a Src homology (SH3) domain of unknown function. Cells overexpressing this domain adhered and calpain-induced integrin signaling complexes formed. However, Rac activation, lamellipodia extension and cell spreading were inhibited. Spreading was restored by overexpression of constitutively active Rac. These studies point to a previously unrecognized role for spectrin and its SH3 domain in initiating Rac activation in the specialized integrin clusters that initiate cell adhesion and spreading. Thus, spectrin may have a pivotal role in initiating integrin-induced physiological and pathological events such as development, proliferation, cell survival, wound healing, metastasis and atherosclerosis.
Collapse
Affiliation(s)
- Katarzyna Bialkowska
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Department of Molecular Cardiology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
132
|
Pereira AL, Veras SS, Silveira ÉJ, Seabra FR, Pinto LP, Souza LB, Freitas RA. The role of matrix extracellular proteins and metalloproteinases in head and neck carcinomas: an updated review. Braz J Otorhinolaryngol 2005; 71:81-6. [PMID: 16446896 PMCID: PMC9443490 DOI: 10.1016/s1808-8694(15)31289-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interactions involving tumor cells and the extracellular matrix (ECM) strongly influence tumor development, including head and neck tumors, affecting cell proliferation and survival as well as the ability to migrate beyond the original location into other tissues to form metastases. These cell migration is often facilitated by partial destruction of the surrounding ECM, which is catalyzed by matrix metalloproteinases (MMPs), a family of more than 20 endopeptidases that is controlled by regulated expression of specific inhibitors (TIMPs). Several studies of ECM and MMPs markers have provided additional diagnostic and prognostic information in head and neck carcinomas. In this review, we are considering the role of ECM and MMPs in tumor progression, emphasizing its proteolytic contributors to this process, and interactions between several members of ECM providing substrate to regulation of this process.
Collapse
Affiliation(s)
- Antonio L.A. Pereira
- Professor of Periodontics, Department of Dental Sciences, Federal University of Maranhão; Ph.D. studies in Oral Pathology under course, Federal University of Rio Grande do Norte (UFRN)
| | - Simone S.L. Veras
- Professor of Oral Pathology, Department of Pathology and Dental Clinical Practice, Federal University of Piauí, Ph.D. in Oral Pathology under course,/UFRN, Sponsored by CNPq
| | | | - Flávio R.G. Seabra
- Professor of Periodontics and Surgery, Universidade Potiguar, Ph.D. in Oral Pathology under course, UFRN
| | | | - Lélia B. Souza
- Professor, Ph.D., Post-graduation in Oral Pathology, UFRN
| | | |
Collapse
|
133
|
Jia Y, Zeng ZZ, Markwart SM, Rockwood KF, Ignatoski KMW, Ethier SP, Livant DL. Integrin Fibronectin Receptors in Matrix Metalloproteinase-1–Dependent Invasion by Breast Cancer and Mammary Epithelial Cells. Cancer Res 2004; 64:8674-81. [PMID: 15574776 DOI: 10.1158/0008-5472.can-04-0069] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Integrins contribute to progression in many cancers, including breast cancer. For example, the interaction of alpha(5)beta(1) with plasma fibronectin causes the constitutive invasiveness of human prostate cancer cells. Inhibition of this process reduces tumorigenesis and prevents metastasis and recurrence. In this study, naturally serum-free basement membranes were used as invasion substrates. Immunoassays were used to compare the roles of alpha(5)beta(1) and alpha(4)beta(1) fibronectin receptors in regulating matrix metalloproteinase (MMP)-1-dependent invasion by human breast cancer and mammary epithelial cells. We found that a peptide consisting of fibronectin PHSRN sequence, Ac-PHSRN-NH(2), induces alpha(5)beta(1)-mediated invasion of basement membranes in vitro by human breast cancer and mammary epithelial cells. PHSRN-induced invasion requires interstitial collagenase MMP-1 activity and is suppressed by an equimolar concentration of a peptide consisting of the LDV sequence of the fibronectin connecting segment, Ac-LHGPEILDVPST-NH(2), in mammary epithelial cells, but not in breast cancer cells. This sequence interacts with alpha(4)beta(1), an integrin that is often down-regulated in breast cancer cells. Immunoblotting shows that the PHSRN peptide stimulates MMP-1 production by serum-free human breast cancer and mammary epithelial cells and that the LDV peptide represses PHSRN-stimulated MMP-1 production only in mammary epithelial cells. Furthermore, PHSRN stimulates MMP-1 activity in breast cancer cells and mammary epithelial cells with a time course that closely parallels invasion induction. Thus, down-regulation of surface alpha(4)beta(1) during oncogenic transformation may be crucial for establishment of the alpha(5)beta(1)-induced, MMP-1-dependent invasive phenotype of breast cancer cells.
Collapse
Affiliation(s)
- Yifeng Jia
- Department of Radiation Oncology and Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | |
Collapse
|
134
|
Ang J, Lijovic M, Ashman LK, Kan K, Frauman AG. CD151 Protein Expression Predicts the Clinical Outcome of Low-Grade Primary Prostate Cancer Better than Histologic Grading: A New Prognostic Indicator? Cancer Epidemiol Biomarkers Prev 2004. [DOI: 10.1158/1055-9965.1717.13.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Abstract
Objective: CD151 is the first member of the tetraspanin family to be associated as a promoter of human tumor metastasis. However, its biological function and expression phenotype among different tumors has not been well investigated.
Method: Tissue specimens from 76 primary prostate cancers and 30 benign prostate hyperplasia (BPH) controls were obtained from the Department of Anatomical Pathology at the Austin and Repatriation Medical Centre (now Austin Health) from 1984 to 1993. We used quantitative immunohistochemical analysis to measure CD151 protein expression. Analyses of differences among BPH and prostate cancer groups were done with one-way ANOVA and Newman-Keuls test. The Kaplan-Meier method and the log-rank test were used to estimate the overall survival.
Results: CD151 expression was found to be significantly higher in prostate cancer specimens compared with BPH specimens (P < 0.001). Poorly differentiated cancers expressed the strongest staining, whereas well-differentiated cancers expressed the weakest staining for CD151 (P < 0.001). The overall survival rate for cases in which CD151 expression was reduced was significantly higher than for cases in which CD151 expression was increased (P = 0.039) especially in well and moderately differentiated cancers (P = 0.014). This effect was independent of the patients' age or preoperative prostate-specific antigen values and superior in the predictive ability of the Gleason score.
Conclusions: CD151 has an increasing expression pattern in prostate cancer progression, and higher levels of CD151 are associated with poorer prognosis. CD151 had better predicting value for the clinical outcome of prostate cancer patients than does the traditional histologic grading method (Gleason grading).
Collapse
Affiliation(s)
- Jian Ang
- 1Clinical Pharmacology and Therapeutics Unit, Heidelberg, Victoria, Australia
| | - Marijana Lijovic
- 1Clinical Pharmacology and Therapeutics Unit, Heidelberg, Victoria, Australia
| | - Leonie K. Ashman
- 3Department of Nephrology, Austin Health, Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Kathleen Kan
- 2Experimental Oncology Unit, University of Newcastle, Callaghan, New South Wales, Australia and
| | - Albert G. Frauman
- 1Clinical Pharmacology and Therapeutics Unit, Heidelberg, Victoria, Australia
| |
Collapse
|
135
|
Lowrie AG, Salter DM, Ross JA. Latent effects of fibronectin, alpha5beta1 integrin, alphaVbeta5 integrin and the cytoskeleton regulate pancreatic carcinoma cell IL-8 secretion. Br J Cancer 2004; 91:1327-34. [PMID: 15354211 PMCID: PMC2409896 DOI: 10.1038/sj.bjc.6602132] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Interactions between tumour cells and the microenvironment are increasingly recognised to have an influence on cancer progression. In pancreatic carcinoma, a highly desmoplastic stroma with abnormal extracellular matrix (ECM) protein and interleukin-8 (IL-8) expression is seen. To investigate whether the ECM may further contribute to abnormalities in the microenvironment by influencing IL-8 secretion, we cultured the Mia PaCa2 pancreatic carcinoma cell line on fibronectin. This resulted in a dose-dependent increase in IL-8 secretion, which was RGD dependent and accompanied by cell spreading and proliferation. The role of spreading was assessed by disruption of the cytoskeleton with cytochalasin D, resulting in a large increase in IL-8 secretion, which was reduced from 31- to 24-fold by fibronectin. This remarkable response was associated with inhibition of spreading and proliferation and represents a novel cytoskeletal function. To investigate whether it could be accounted for by the loss of integrin-mediated signalling, the expressed α5β1, αVβ5 and α3β1 integrins were inhibited. α5β1 inhibition prevented spreading and proliferation but produced a much smaller rise in IL-8 secretion than cytochalasin D. αVβ5 inhibition alone had only minor effects but when inhibited in combination with α5β1 completely abolished the response to fibronectin. These results reveal latent stimulatory effects of the αVβ5 integrin on IL-8 secretion and suggest that integrin crosstalk may limit the induction of IL-8 secretion by fibronectin. However, the magnitude of IL-8 secretion induced by cytochalasin cannot be accounted for by integrin signalling and may reflect the influence of another signalling pathway or a novel, intrinsic cytoskeletal function.
Collapse
Affiliation(s)
- A G Lowrie
- Tissue Injury and Repair Group, 6th Floor, Centre for Inflammation Research, University of Edinburgh Medical School, Teviot Place, Edinburgh EH8 9AG, UK
| | - D M Salter
- Department of Pathology, University of Edinburgh, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
| | - J A Ross
- Tissue Injury and Repair Group, 6th Floor, Centre for Inflammation Research, University of Edinburgh Medical School, Teviot Place, Edinburgh EH8 9AG, UK
- Tissue Injury and Repair Group, 6th Floor, Centre for Inflammation Research, University of Edinburgh Medical School, Teviot Place, Edinburgh EH8 9AG, UK. E-mail:
| |
Collapse
|
136
|
Pasco S, Ramont L, Maquart FX, Monboisse JC. Control of melanoma progression by various matrikines from basement membrane macromolecules. Crit Rev Oncol Hematol 2004; 49:221-33. [PMID: 15036262 DOI: 10.1016/j.critrevonc.2003.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2003] [Indexed: 11/25/2022] Open
Abstract
Many biological processes such as cell differentiation, cell migration or gene expression are tightly controlled by cell-cell interactions or by various cytokines. During tumor progression, cancer cells are in contact with extracellular matrix (ECM) macromolecules involving specific receptors such as integrins. The different stages of tumor progression, and mainly the proteolytic cascades implicated in extracellular matrix degradation and cell migration, may be controlled by the extracellular matrix macromolecules or by domains released by directed and limited proteolysis of these molecules. In this review, we summarise the biological effects of various peptides, named matrikines, derived from basement membranes (BM) components, such as laminins (LN), proteoglycans or collagens. These peptides may control tumor progression by regulating the proteolytic cascades leading to cancer cell dissemination and metastasis.
Collapse
Affiliation(s)
- Sylvie Pasco
- Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS FRE 2534, Faculté de Médecine, IFR 53 Biomolécules, 51 Rue Cognac Jay, 51095 Reims Cedex, France
| | | | | | | |
Collapse
|
137
|
Aguzzi MS, Facchiano F, Ribatti D, Gaeta R, Casadio R, Rossi I, Capogrossi MC, Facchiano A. A novel RGDS-analog inhibits angiogenesis in vitro and in vivo. Biochem Biophys Res Commun 2004; 321:809-14. [PMID: 15358099 DOI: 10.1016/j.bbrc.2004.07.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2004] [Indexed: 11/28/2022]
Abstract
In this study the anti-angiogenic action of a novel non-peptide RGDS-analog named RAM was tested in vitro and in vivo. RAM inhibited FGF-2-induced chemotaxis by 80% in an adhesion-independent way. Further, it induced HUVEC-apoptosis in collagen-seeded HUVEC, indicating that such pro-apoptotic effect was adhesion-independent. In vivo studies revealed that RAM inhibited FGF-2 induced angiogenesis by 60% in the mouse Matrigel-assay and in the chicken-egg chorion-allantoic membrane assay. Finally, RAM was markedly more stable in serum as compared to the template RGDS and after 24 h incubation in 100% serum was significantly more active than RGDS. Taken together these results show that RAM exerts anti-chemotactic and pro-apoptotic effects, by an unexpected adhesion-independent mechanism, as we have recently shown for the template RGDS molecule [Blood 103 (2004) 4180], and has in vivo relevant anti-angiogenic properties, with marked stability in serum; therefore, RAM represents a novel promising anti-angiogenic molecule.
Collapse
Affiliation(s)
- M S Aguzzi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico della Immacolata, IDI, IRCCS, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Muramatsu H, Zou P, Suzuki H, Oda Y, Chen GY, Sakaguchi N, Sakuma S, Maeda N, Noda M, Takada Y, Muramatsu T. alpha4beta1- and alpha6beta1-integrins are functional receptors for midkine, a heparin-binding growth factor. J Cell Sci 2004; 117:5405-15. [PMID: 15466886 DOI: 10.1242/jcs.01423] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Midkine is a heparin-binding growth factor that promotes the growth, survival, migration and differentiation of various target cells. So far, receptor-type protein tyrosine phosphatase zeta, low-density-lipoprotein-receptor-related protein and anaplastic lymphoma kinase have been identified as receptors for midkine. We found beta1 integrin in midkine-binding proteins from 13-day-old mouse embryos. beta1-Integrin bound to a midkine-agarose column and was eluted mostly with EDTA. Further study revealed that the alpha-subunits capable of binding to midkine were alpha4 and alpha6. Purified alpha4beta1- and alpha6beta1-integrins bound midkine. Anti-alpha4 antibody inhibited the midkine-dependent migration of osteoblastic cells, and anti-alpha6 antibody inhibited the midkine-dependent neurite outgrowth of embryonic neurons. After midkine treatment, tyrosine phosphorylation of paxillin, an integrin-associated molecule, was transiently increased in osteoblastic cells. Therefore, we concluded that alpha4beta1- and alpha6beta1-integrins are functional receptors for midkine. We observed that the low-density-lipoprotein-receptor-related-protein-6 ectodomain was immunoprecipitated with alpha6beta1-integrin and alpha4beta1-integrin. The low-density-lipoprotein-receptor-related-protein-6 ectodomain was also immunoprecipitated with receptor-type protein tyrosine phosphatase zeta. alpha4beta1- and alpha6beta1-Integrins are expected to co-operate with other midkine receptors, possibly in a multimolecular complex that contains other midkine receptors.
Collapse
Affiliation(s)
- Hisako Muramatsu
- Department of Biochemistry and Division of Disease Models, Center for Neural Disease and Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Chavakis T, Athanasopoulos A, Rhee JS, Orlova V, Schmidt-Wöll T, Bierhaus A, May AE, Celik I, Nawroth PP, Preissner KT. Angiostatin is a novel anti-inflammatory factor by inhibiting leukocyte recruitment. Blood 2004; 105:1036-43. [PMID: 15383457 DOI: 10.1182/blood-2004-01-0166] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis and inflammation are closely related biologic processes in wound healing and the responses to vascular injury as well as in cardiovascular diseases; however, the molecular connections are poorly defined. In particular, it is yet unclear whether endogenous factors can regulate both angiogenesis and inflammation. Here, we show that the endogenous angiogenesis inhibitor, angiostatin (containing kringle domains 1-4 of plasminogen), serves an anti-inflammatory role, since the kringles 1-3 and its kringle 4 directly interact with leukocyte beta1- and beta2-integrins, respectively. In particular, a specific interaction between kringle 4 and alphaMbeta2-integrin (Mac-1) but not leukocyte function antigen 1 (LFA-1) was identified. Angiostatin thereby inhibited beta1- and beta2-integrin-mediated adhesion of leukocytes to extracellular matrix proteins and the endothelium as well as their transmigration through the endothelium in vitro. Moreover, angiostatin blocked the peritonitis-induced neutrophil emigration in vivo. In addition, through its interaction with Mac-1, angiostatin reduced activation of the proinflammatory transcription factor nuclear factor kappaB (NFkappaB), as well as the NFkappaB-related expression of tissue factor, a potent initiator of hemostasis following vascular injury. Finally, angiostatin forms were generated in vivo following skin injury/inflammation and were detectable during the following entire period of wound healing peaking at the terminal phase of the healing process. Taken together, over and above inhibition of neovascularization, angiostatin was identified as an antiadhesive/anti-inflammatory substance. These observations could provide the basis for new therapeutic applications of angiostatin to target chronic inflammatory processes in different pathologic situations.
Collapse
|
140
|
Zwartz G, Chigaev A, Foutz T, Larson RS, Posner R, Sklar LA. Relationship between molecular and cellular dissociation rates for VLA-4/VCAM-1 interaction in the absence of shear stress. Biophys J 2004; 86:1243-52. [PMID: 14747358 PMCID: PMC1303916 DOI: 10.1016/s0006-3495(04)74198-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The rate of leukocyte recruitment to and detachment from the vasculature contributes to cellular tethering, rolling, firm adherence, and migration across an endothelium layer. The molecular rates depend on the type and number of bound integrin or selectin adhesion molecules, shear force acting on the bound adhesion molecules, and affinity state of integrins. Although little is known of the effect that the number of adhesion molecules has on leukocyte recruitment, it has been shown that firm adhesion for cells in suspension may be mediated by small numbers of bound adhesion molecules. We studied the disaggregation of aggregates composed of B78H1 cells transfected with human vascular cell adhesion molecule-1 (VCAM-1) and human monoblastoid U937 cells expressing Very Late Antigen-4 (VLA-4). Aggregate disaggregation rates were obtained and compared to dissociation rates for soluble rhVCAM-1 ligand and monoblastoid U937 cells. Under conditions without shear stress, it was found that average cellular disaggregation rates were a factor of 1.3 +/- 0.4 times slower than molecular dissociation rates for the 1 mM Mn(2+) and 1 mM Mn(2+) + 1 mM Ca(2+) conditions. A simple mathematical model was used to predict how much smaller the dissociation constant would be if the number of bonds holding an aggregate varied from one bond to N bonds under conditions without shear stress. The average number of adhesion bonds holding the cell aggregates together was found to be 1.5 +/- 0.7. This suggests that a few bonds were needed to form cellular aggregates and that increased aggregation was related to integrin affinity changes and not due to clustering or increased bond numbers.
Collapse
Affiliation(s)
- Gordon Zwartz
- Department of Pathology and Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | | | | | | | | | | |
Collapse
|
141
|
Sava G, Frausin F, Cocchietto M, Vita F, Podda E, Spessotto P, Furlani A, Scarcia V, Zabucchi G. Actin-dependent tumour cell adhesion after short-term exposure to the antimetastasis ruthenium complex NAMI-A. Eur J Cancer 2004; 40:1383-96. [PMID: 15177498 DOI: 10.1016/j.ejca.2004.01.034] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2003] [Revised: 09/20/2003] [Accepted: 01/16/2004] [Indexed: 11/25/2022]
Abstract
Imidazolium trans-imidazoledimethylsulphoxidetrachlororuthenate (NAMI-A) was tested in vitro on the pro-adhesive properties, evaluated as resistance to trypsin treatment, which is a bona fide measure of adhesion strength, of KB and HeLa carcinoma cell lines and on human polymorphonuclear neutrophils (HPMN). NAMI-A increased the pro-adhesive activity of KB cells at 0.001 mM concentration, after few minutes incubation and this effect was not influenced by the vehicle used for cell challenge, neither did it depend on NAMI-A concentration or on temperature. The same effect occurred on HeLa cells at 0.01 mM NAMI-A. This effect, detected at concentrations up to 100 times lower than those necessary to block cells at the G(2)-M premitotic phase of cell cycle, or to inhibit matrix metalloproteinase release or cell invasion, was not related to ruthenium uptake by tumour cells. HeLa cells and healthy HPMN, following short exposure to 0.1 mM NAMI-A, assumed a different shape, with the extrusion of filopodia (HeLa) and of large lamellopodia (HPMN), which increased their interactions with the substrate. This effect was attributed to stabilisation, altered turnover and sensitivity to cytochalasin D of actin filaments. Provided that adhesion is associated with cell motility and invasion, these data suggest that NAMI-A may exert antimetastatic properties at concentrations lower than those observed in the lungs at the end of a conventional intraperitoneal treatment in vivo.
Collapse
Affiliation(s)
- G Sava
- Department of Biomedical Sciences, University of Trieste, Via L. Giorgieri 7, 34127 Trieste, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Pilorget A, Berthet V, Luis J, Moghrabi A, Annabi B, Béliveau R. Medulloblastoma cell invasion is inhibited by green tea (-)epigallocatechin-3-gallate. J Cell Biochem 2004; 90:745-55. [PMID: 14587030 DOI: 10.1002/jcb.10667] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Epigallocatechin-3-gallate (EGCG), the major green tea polyphenol, can reach the brain following oral intake and could thus act as an anti-tumoral agent targeting several key steps of brain cancer cells invasive activity. Because integrin-mediated extracellular matrix recognition is crucial during the cell adhesion processes involved in carcinogenesis, we have investigated the effects of EGCG on different cellular integrins of the pediatric brain tumor-derived medulloblastoma cell line DAOY. Using flow cytometry, we report the levels of expression of several cell surface integrins in DAOY. These include high expression of alpha2, alpha3, and beta1 integrins, as well as alphav and beta3 integrins. Moreover, we provide evidence that EGCG can antagonize DAOY cell migration specifically on collagen by increasing cell adhesive ability through specific gene and protein upregulation of the beta1 integrin subunit. Our results suggest that this naturally occurring green tea polyphenol may thus be used as a nutraceutical therapeutic agent in targeting the invasive character of medulloblastomas.
Collapse
Affiliation(s)
- Anthony Pilorget
- Laboratoire de Médecine Moléculaire, Centre de Cancérologie Charles Bruneau, Hôpital Sainte-Justine and Université du Québec à Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
143
|
Shields CJ, Winter DC, Wang JH, Andrews E, Laug WE, Redmond HP. Hypertonic saline impedes tumor cell–endothelial cell interaction by reducing adhesion molecule and laminin expression. Surgery 2004; 136:76-83. [PMID: 15232542 DOI: 10.1016/j.surg.2003.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Hypertonic saline infusion dampens inflammatory responses and suppresses neutrophil-endothelial interaction by reducing adhesion molecule expression. This study tested the hypothesis that hypertonic saline attenuates tumor cell adhesion to the endothelium through a similar mechanism. METHODS Human colon cancer cells (LS174T) were transfected with green fluorescent protein and exposed to lipopolysaccharide, tumor necrosis factor-alpha, and interleukin-6 under hypertonic and isotonic conditions for 1 and 4 hours. Confluent human umbilical vein endothelial cells were similarly exposed. Cellular apoptosis and expression of adhesion molecules and laminin were measured by flow cytometry. Tumor cell adhesion to endothelium and laminin was assessed with fluorescence microscopy. Data are represented as mean +/- standard error of mean, and an ANOVA test was performed to gauge statistical significance, with P <.05 considered significant. RESULTS Hypertonic exposure significantly reduced tumor cell adhesion despite the presence of the perioperative cell stressors (42 +/- 2.9 vs 172.5 +/- 12.4, P <.05), attenuated tumor cell beta-1 integrin (14.43 vs 23.84, P <.05), and endothelial cell laminin expression (22.78 +/- 2.2 vs 33.74 +/- 2.4, P <.05), but did not significantly alter cell viability. CONCLUSION Hypertonic saline significantly attenuates tumor cell adhesion to endothelium by inhibiting adhesion molecule and laminin expression. This may halt the metastatic behavior of tumor cells shed at surgery.
Collapse
Affiliation(s)
- Conor J Shields
- Department of Academic Surgery, Cork University Hospital and National University of Ireland, Wilton
| | | | | | | | | | | |
Collapse
|
144
|
Li DQ, Wang ZB, Bai J, Zhao J, Wang Y, Hu K, Du YH. Effects of mifepristone on invasive and metastatic potential of human gastric adenocarcinoma cell line MKN-45 in vitro and in vivo. World J Gastroenterol 2004; 10:1726-9. [PMID: 15188494 PMCID: PMC4572257 DOI: 10.3748/wjg.v10.i12.1726] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To investigate the effects of mifepristone on the invasive and metastatic potential of human gastric adenocarcinoma cell line MKN-45 and its mechanisms.
METHODS: After incubation with various concentrations of mifepristone (5, 10, 20 μmol/L), the adhesion to artificial basement membrane, Matrigel, and the migration of MKN-45 cells were assayed using MTT assay and Transwell cell culture chambers, respectively. Enzyme- linked immunoabsorbent assay (ELISA) and flow cytometry were used to determine the expression of vascular endothelial growth factor (VEGF) and integrin β3 in the cells. After subcutaneous transplantation of MKN-45 cells in nude mice, mifepristone (50 mg/kg·d) was administrated subcutaneously for 8 wk to assess its effects on tumor metastasis. Immunohistochemical analysis was used to detect the expression of VEGF and microvascular density (MVD) in xenografted tumors.
RESULTS: Mifepristone dose-dependently inhibited the heterotypic adhesion to Matrigel of MKN-45 cells. The inhibition was accompanied by a significant down-regulation of integrin β3 expression in the cells. After incubation with 5, 10, 20 μmol/L mifepristone, the number of migrated MKN-45 cells was 72 ± 8, 50 ± 6, 41 ± 5 in experiment group, and 94 ± 16 in control group (P < 0.01). Meanwhile, secreted VEGF protein of MKN-45 cells in mifepristone-treated group (14.2 ± 2.9, 8.9 ± 3.1, 5.4 ± 2.1 ng/g per liter) was significantly lower than that in control group (22.7 ± 4.3 ng/g per liter, P < 0.01). In vivo, mifepristone decreased the number of metastatic foci in lungs of nude mice and down-regulated the expression of VEGF and MVD in the xenograted tumors.
CONCLUSION: Mifepristone can effectively inhibit the invasive and metastatic potential of human gastric adenocarcinoma cell line MKN-45 in vitro and in vivo through inhibition of heterotypic adhesion to basement membrane, cell migration and angiogenesis.
Collapse
Affiliation(s)
- Da-Qiang Li
- State Key Laboratory of Ultrasound Engineering in Medicine, Chongqing Medical University(PO Box 153), Chongqing 400016, China
| | | | | | | | | | | | | |
Collapse
|
145
|
Aguzzi MS, Giampietri C, De Marchis F, Padula F, Gaeta R, Ragone G, Capogrossi MC, Facchiano A. RGDS peptide induces caspase 8 and caspase 9 activation in human endothelial cells. Blood 2004; 103:4180-7. [PMID: 14982875 DOI: 10.1182/blood-2003-06-2144] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AbstractPeptides containing the Arg-Gly-Asp (RGD) motif inhibit cell adhesion and exhibit a variety of other biologic effects including anticoagulant and antimetastatic activities. The aim of the present study was to examine the anchorage-independent effects of an RGD-containing peptide, Arg-Gly-Asp-Ser (RGDS), on human umbilical vein endothelial cells (HUVECs). Assays were performed on HUVECs seeded onto collagen IV; under these experimental conditions RGDS did not exert antiadhesive effects but significantly reduced FGF-2-dependent chemotaxis after 4 hours of treatment and reduced proliferation after 24 hours of treatment. Experiments carried out with caspase-specific inhibitors indicated that the observed antichemotactic effects required caspase 8 and caspase 9 activation. RGDS activated both caspase 8 and caspase 9 after 4 hours of treatment and caspase 3 after 24 hours of treatment, and markedly enhanced HUVEC apoptosis by transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL)/Hoechst staining and fluorescence-activated cell sorting (FACS) analysis. Finally, confocal microscopy showed that RGDS localizes in the cytoplasm of live HUVECs within 4 hours and in vitro experiments showed that RGDS directly interacts with recombinant caspases 8 and 9 in a specific way. In summary, these results indicate that RGDS directly binds and activates caspases 8 and 9, inhibits chemotaxis, and induces apoptosis of HUVECs with a mechanism independent from its antiadhesive effect.
Collapse
Affiliation(s)
- Maria Simona Aguzzi
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Kjaer M. Role of extracellular matrix in adaptation of tendon and skeletal muscle to mechanical loading. Physiol Rev 2004; 84:649-98. [PMID: 15044685 DOI: 10.1152/physrev.00031.2003] [Citation(s) in RCA: 989] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The extracellular matrix (ECM), and especially the connective tissue with its collagen, links tissues of the body together and plays an important role in the force transmission and tissue structure maintenance especially in tendons, ligaments, bone, and muscle. The ECM turnover is influenced by physical activity, and both collagen synthesis and degrading metalloprotease enzymes increase with mechanical loading. Both transcription and posttranslational modifications, as well as local and systemic release of growth factors, are enhanced following exercise. For tendons, metabolic activity, circulatory responses, and collagen turnover are demonstrated to be more pronounced in humans than hitherto thought. Conversely, inactivity markedly decreases collagen turnover in both tendon and muscle. Chronic loading in the form of physical training leads both to increased collagen turnover as well as, dependent on the type of collagen in question, some degree of net collagen synthesis. These changes will modify the mechanical properties and the viscoelastic characteristics of the tissue, decrease its stress, and likely make it more load resistant. Cross-linking in connective tissue involves an intimate, enzymatical interplay between collagen synthesis and ECM proteoglycan components during growth and maturation and influences the collagen-derived functional properties of the tissue. With aging, glycation contributes to additional cross-linking which modifies tissue stiffness. Physiological signaling pathways from mechanical loading to changes in ECM most likely involve feedback signaling that results in rapid alterations in the mechanical properties of the ECM. In developing skeletal muscle, an important interplay between muscle cells and the ECM is present, and some evidence from adult human muscle suggests common signaling pathways to stimulate contractile and ECM components. Unaccostumed overloading responses suggest an important role of ECM in the adaptation of myofibrillar structures in adult muscle. Development of overuse injury in tendons involve morphological and biochemical changes including altered collagen typing and fibril size, hypervascularization zones, accumulation of nociceptive substances, and impaired collagen degradation activity. Counteracting these phenomena requires adjusted loading rather than absence of loading in the form of immobilization. Full understanding of these physiological processes will provide the physiological basis for understanding of tissue overloading and injury seen in both tendons and muscle with repetitive work and leisure time physical activity.
Collapse
Affiliation(s)
- Michael Kjaer
- Sports Medicine Research Unit, Department of Rheumatology, Copenhagen University Hospital at Bispebjerg, 23 Bispebjerg Bakke, DK-2400 Copenhagen NV, Denmark.
| |
Collapse
|
147
|
Yang FC, Ingram DA, Chen S, Hingtgen CM, Ratner N, Monk KR, Clegg T, White H, Mead L, Wenning MJ, Williams DA, Kapur R, Atkinson SJ, Clapp DW. Neurofibromin-deficient Schwann cells secrete a potent migratory stimulus for Nf1+/- mast cells. J Clin Invest 2004; 112:1851-61. [PMID: 14679180 PMCID: PMC296994 DOI: 10.1172/jci19195] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The NF1 tumor suppressor gene encodes a GTPase-activating protein called neurofibromin that negatively regulates Ras signaling. Mutations in NF1 cause neurofibromatosis type 1 (NF1). The development of neurofibromas, which are complex tumors composed of multiple cell types, is a hallmark of NF1. Somatic inactivation of murine Nf1 in Schwann cells is necessary, but not sufficient, to initiate neurofibroma formation. Neurofibromas occur with high penetrance in mice in which Nf1 is ablated in Schwann cells in the context of a heterozygous mutant (Nf1+/-) microenvironment. Mast cells infiltrate neurofibromas, where they secrete proteins that can remodel the ECM and initiate angiogenesis. Thus, identification of mechanisms responsible for mast cell migration to tumor microenvironments is important for understanding tumorigenesis and for designing potential therapies. Here, we show that homozygous Nf1 mutant (Nf1-/-) Schwann cells secrete Kit ligand (KitL), which stimulates mast cell migration, and that Nf1+/- mast cells are hypermotile in response to KitL. Furthermore, we link hyperactivation of the Ras-class IA-PI3K-Rac2 pathway to increased Nf1+/- mast cell migration. Thus, these studies identify a novel interaction between Nf1-/- Schwann cells and Nf1+/- mast cells that is likely to be important in neurofibroma formation.
Collapse
Affiliation(s)
- Feng-Chun Yang
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Abstract
Under normal circumstances, adhered cells die of anoikis when detached from their extracellular matrix (ECM). Resistance to anoikis has been implicated in the progression of many human malignancies by affording an increased survival time in the absence of matrix attachment, facilitating the migration and eventual colonisation of distant sites. In this study, an anoikis-resistant variant of the human osteosarcoma cell line, SAOS-2 (SAOSar), was generated by sequential cycles of culturing under adhered and suspended conditions. It was also shown that although parental SAOS (SAOSp) cells are a heterogeneous population with varying levels of sensitivity to anoikis, the establishment of anoikis-resistant clones was not necessarily the result of mere selection of a previously resistant subpopulation. Anoikis-resistant cells were also derived from anoikis-sensitive SAOS clones by exposure to anoikis-inducing culture conditions. This suggests that lack of the normal signalling generated by attachment to the ECM could represent a driving force towards anoikis resistance. Resistance to anoikis could not be attributed to a general defect in the apoptotic pathway since apoptosis in both sensitive and resistant populations was induced after treatment with staurosporine, cycloheximide and hydrogen peroxide. This suggests that the apoptotic machinery is intact in both anoikis-sensitive and -resistant SAOS cells and that the death signal in anoikis-sensitive cells is generated by the lack of attachment, most probably by unligated integrins. Anoikis-resistant cells have circumvented this death signal and remain viable despite suspended conditions.
Collapse
Affiliation(s)
- C M Díaz-Montero
- Department of Immunology, Unit 102, The University of Texas M.D. Anderson Cancer Center, PO Box 301402, Houston, TX 77030-1903, USA
| | | |
Collapse
|
149
|
Attwell S, Mills J, Troussard A, Wu C, Dedhar S. Integration of cell attachment, cytoskeletal localization, and signaling by integrin-linked kinase (ILK), CH-ILKBP, and the tumor suppressor PTEN. Mol Biol Cell 2003; 14:4813-25. [PMID: 12960424 PMCID: PMC284786 DOI: 10.1091/mbc.e03-05-0308] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Revised: 07/31/2003] [Accepted: 07/31/2003] [Indexed: 12/19/2022] Open
Abstract
Cell attachment and the assembly of cytoskeletal and signaling complexes downstream of integrins are intimately linked and coordinated. Although many intracellular proteins have been implicated in these processes, a new paradigm is emerging from biochemical and genetic studies that implicates integrin-linked kinase (ILK) and its interacting proteins, such as CH-ILKBP (alpha-parvin), paxillin, and PINCH in coupling integrins to the actin cytoskeleton and signaling complexes. Genetic studies in Drosophila, Caenorhabditis elegans, and mice point to an essential role of ILK as an adaptor protein in mediating integrin-dependent cell attachment and cytoskeletal organization. Here we demonstrate, using several different approaches, that inhibiting ILK kinase activity, or expression, results in the inhibition of cell attachment, cell migration, F-actin organization, and the specific cytoskeletal localization of CH-ILKBP and paxillin in human cells. We also demonstrate that the kinase activity of ILK is elevated in the cytoskeletal fraction and that the interaction of CH-ILKBP with ILK within the cytoskeleton stimulates ILK activity and downstream signaling to PKB/Akt and GSK-3. Interestingly, the interaction of CH-ILKBP with ILK is regulated by the Pi3 kinase pathway, because inhibition of Pi3 kinase activity by pharmacological inhibitors, or by the tumor suppressor PTEN, inhibits this interaction as well as cell attachment and signaling. These data demonstrate that the kinase and adaptor properties of ILK function together, in a Pi3 kinase-dependent manner, to regulate integrin-mediated cell attachment and signal transduction.
Collapse
Affiliation(s)
- Sarah Attwell
- BC Cancer Agency, Jack Bell Research Centre, Vancouver, BC, V6H 3Z6, Canada
| | | | | | | | | |
Collapse
|
150
|
Nakayamada S, Okada Y, Saito K, Tamura M, Tanaka Y. Beta1 integrin/focal adhesion kinase-mediated signaling induces intercellular adhesion molecule 1 and receptor activator of nuclear factor kappaB ligand on osteoblasts and osteoclast maturation. J Biol Chem 2003; 278:45368-74. [PMID: 12954625 DOI: 10.1074/jbc.m308786200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have assessed characteristics of primary human osteoblasts, shedding light on signaling mediated by beta1 integrin. beta1 integrins are major receptors for these matrix glycoproteins. 1) Integrins beta1, alpha2, alpha3, alpha4, alpha5, alpha6, and alphav were highly expressed on primary osteoblasts. 2) Engagement of beta1 integrins on osteoblasts by cross-linking with specific antibody or ligand matrices, such as fibronectin or collagen, augmented expression of intercellular adhesion molecule 1 (ICAM-1) and receptor activator of nuclear factor kappaB ligand (RANKL) on the surface. 3) Up-regulation of ICAM-1 and RANKL on osteoblasts by beta1 stimulation was completely abrogated by pretreatment with herbimycin A and genistein, tyrosine kinase inhibitors, or transfection of dominant negative truncations of focal adhesion kinase (FAK). 4) Engagement of beta1 integrins on osteoblasts induced tartrate-resistant acid phosphatase-positive multinuclear cell formation in the coculture system of osteoblasts and peripheral monocytes. 5) Up-regulation of tartrate-resistant acid phosphatase-positive multinuclear cell formation by beta1 stimulation was completely abrogated by transfection of dominant negative truncations of FAK. Our results indicate that beta1 integrin-dependent adhesion of osteoblasts to bone matrices induces ICAM-1 and RANKL expression and osteoclast formation via tyrosine kinase, especially FAK. We here propose that beta1 integrin/FAK-mediated signaling on osteoblasts could be involved in ICAM-1- and RANKL-dependent osteoclast maturation.
Collapse
Affiliation(s)
- Shingo Nakayamada
- First and Second Department of Internal Medicine, University of Occupational and Environmental Health, Japan, School of Medicine, Kitakyushu, 807-8555, Japan
| | | | | | | | | |
Collapse
|