151
|
The role of β3-integrins in tumor angiogenesis: context is everything. Curr Opin Cell Biol 2011; 23:630-7. [PMID: 21565482 DOI: 10.1016/j.ceb.2011.03.014] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 03/26/2011] [Indexed: 02/07/2023]
Abstract
Integrins are a family of cell-extracellular matrix adhesion molecules that play important roles in tumor angiogenesis. αvβ3-Integrin has received much attention as a potential anti-angiogenic target because it is upregulated in tumor-associated blood vessels. Agents targeting αvβ3-integrin are now showing some success in phase III clinical trails for the treatment of glioblastoma, but the exact function of this integrin in tumor angiogenesis is still relatively unknown. This review highlights some of the recent data illustrating that β3-integrins play both pro-angiogenic and anti-angiogenic roles in tumor angiogenesis depending on the context. Specifically we will discuss how the following differentially influence β3-integrin's role in tumor angiogenesis: first, cell-matrix interactions, second, β3-integrin inhibitor doses, third, cell type, and fourth, other interacting molecules.
Collapse
|
152
|
Isthmin exerts pro-survival and death-promoting effect on endothelial cells through alphavbeta5 integrin depending on its physical state. Cell Death Dis 2011; 2:e153. [PMID: 21544092 PMCID: PMC3122116 DOI: 10.1038/cddis.2011.37] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Isthmin (ISM) is a 60 kDa secreted-angiogenesis inhibitor that suppresses tumor growth in mouse and disrupts vessel patterning in zebrafish embryos. It selectively binds to alphavbeta5 (αvβ5) integrin on the surface of endothelial cells (ECs), but the mechanism of its antiangiogenic action remains unknown. In this work, we establish that soluble ISM suppresses in vitro angiogenesis and induces EC apoptosis by interacting with its cell surface receptor αvβ5 integrin through a novel 'RKD' motif localized within its adhesion-associated domain in MUC4 and other proteins domain. ISM induces EC apoptosis through integrin-mediated death (IMD) by direct recruitment and activation of caspase-8 without causing anoikis. On the other hand, immobilized ISM loses its antiangiogenic function and instead promotes EC adhesion, survival and migration through αvβ5 integrin by activating focal adhesion kinase (FAK). ISM unexpectedly has both a pro-survival and death-promoting effect on ECs depending on its physical state. This dual function of a single antiangiogenic protein may impact its antiangiogenic efficacy in vivo.
Collapse
|
153
|
Copland IB, Galipeau J. Death and inflammation following somatic cell transplantation. Semin Immunopathol 2011; 33:535-50. [DOI: 10.1007/s00281-011-0274-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 04/14/2011] [Indexed: 12/13/2022]
|
154
|
Yubero N, Jiménez-Marín A, Barbancho M, Garrido JJ. Two cDNAs coding for the porcine CD51 (αv) integrin subunit: cloning, expression analysis, adhesion assays and chromosomal localization. Gene 2011; 481:29-40. [PMID: 21549183 DOI: 10.1016/j.gene.2011.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 04/11/2011] [Accepted: 04/15/2011] [Indexed: 02/06/2023]
Abstract
CD51 (α(v)) is an integrin chain that associates with multiple β integrin chains to form different receptor complexes that mediate important human processes. Pigs show substantial physiological, immunological and anatomical similarities to humans, and are therefore a good model system to study immunological and pathological processes. Here we report the cloning and characterization of two cDNAs produced by alternative splicing that encode two different porcine CD51 proteins that differ in five amino acid residues. Pig CD51 cDNAs encode polypeptides of 1046 or 1041 amino acid residues, respectively, that share with other mammalian homologous proteins a high percentage amino acid identity and the functional domains. Expression analysis of CD51 was carried out at two different levels. RT-PCR analysis revealed that both CD51 transcripts were expressed ubiquitously but heterogeneously, with the exception of some platelets in which only the smallest CD51 transcript was detected. A specific monoclonal antibody against a pig CD51 recombinant protein was made and used in the immunohistochemical localization of CD51 proteins. It showed that CD51 was mainly expressed in hematopoietic cells of myeloid linage, epithelial and endothelial cells, osteoclasts, nervous fibers and smooth muscle. Adhesion assays showed that in the presence of Mn(++) pig α(v)-CHO-B2 transfected cells increased their attachment to fibronectin and vitonectin, but not to fibrinogen. Finally, we localized the CD51 gene on the porcine chromosome 15 (SSC15), q23-q26.
Collapse
Affiliation(s)
- Noemí Yubero
- Unidad de Genómica y Mejora Animal, Departamento de Genética, Universidad de Córdoba, Campus de Rabanales, 14071 Córdoba, Spain
| | | | | | | |
Collapse
|
155
|
Mas-Moruno C, Rechenmacher F, Kessler H. Cilengitide: the first anti-angiogenic small molecule drug candidate design, synthesis and clinical evaluation. Anticancer Agents Med Chem 2011; 10:753-68. [PMID: 21269250 PMCID: PMC3267166 DOI: 10.2174/187152010794728639] [Citation(s) in RCA: 480] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 02/02/2011] [Indexed: 12/29/2022]
Abstract
Cilengitide, a cyclic RGD pentapeptide, is currently in clinical phase III for treatment of glioblastomas and in phase II for several other tumors. This drug is the first anti-angiogenic small molecule targeting the integrins αvβ3, αvβ5 and αvβ1. It was developed by us in the early 90s by a novel procedure, the spatial screening. This strategy resulted in c(RGDfV), the first superactive αvβ3 inhibitor (100 to 1000 times increased activity over the linear reference peptides), which in addition exhibited high selectivity against the platelet receptor αIIbβ3. This cyclic peptide was later modified by N-methylation of one peptide bond to yield an even greater antagonistic activity in c(RGDf(NMe)V). This peptide was then dubbed Cilengitide and is currently developed as drug by the company Merck-Serono (Germany). This article describes the chemical development of Cilengitide, the biochemical background of its activity and a short review about the present clinical trials. The positive anti-angiogenic effects in cancer treatment can be further increased by combination with "classical" anti-cancer therapies. Several clinical trials in this direction are under investigation.
Collapse
Affiliation(s)
- Carlos Mas-Moruno
- Institute for Advance Study, Department Chemie, Technische Universität München, Garching, Germany
| | | | | |
Collapse
|
156
|
Tauszig-Delamasure S, Bouzas-Rodriguez J. Targeting neurotrophin-3 and its dependence receptor tyrosine kinase receptor C: a new antitumoral strategy. Expert Opin Ther Targets 2011; 15:847-58. [DOI: 10.1517/14728222.2011.575361] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
157
|
Abstract
As materials technology and the field of tissue engineering advance, the role of cellular adhesive mechanisms, in particular, interactions with implantable devices, becomes more relevant in both research and clinical practice. A key tenet of medical device technology is to use the exquisite ability of biological systems to respond to the material surface or chemical stimuli in order to help to develop next-generation biomaterials. The focus of this review is on recent studies and developments concerning focal adhesion formation in osteoneogenesis, with an emphasis on the influence of synthetic constructs on integrin-mediated cellular adhesion and function.
Collapse
Affiliation(s)
- M J P Biggs
- Nanotechnology Center for Mechanics in Regenerative Medicine, Department of Applied Physics and Applied Mathematics, Columbia University, New York 10027, USA.
| | | |
Collapse
|
158
|
Abstract
Malignant gliomas are the most destructive type of brain cancer. In order to gain a better understanding of the molecular mechanisms of glioma cell death and survival, we previously established an alkylating agent 1, 3-bis(2-chloroethyl)-1-nitrosourea (BCNU)-resistant variant of C6 rat glioma cells. Proteomic analysis indicated a significant down-regulation of integrin beta 3 (ITGB3) in the BCNU-resistant C6R cells. Re-expression of ITGB3 in C6R cells restored the BCNU sensitivity. In U87MG, U373MG, and T98G human glioma cells, there was a positive correlation between ITGB3 expression and the sensitivity to BCNU and etoposide, suggesting an important role of ITGB3 in glioma cell death. Over-expression of ITGB3 cDNA significantly increased the sensitivity of the human glioma cells to the anticancer drug-induced apoptosis. Nitric oxide showed an additive effect on the anticancer drug-induced glioma cell death by increasing ITGB3 expression. Subsequent dissection of signaling pathways indicated that extracellular signal-regulated kinase and unligated integrin-mediated cell death pathway may be involved in the pro-apoptotic role of ITGB3 in glioma cells. These results implicate ITGB3 in glioma cell death/survival and drug resistance.
Collapse
Affiliation(s)
- Jong-Heon Kim
- Department of Pharmacology, Brain Science and Engineering Institute, CMRI, Kyungpook National University School of Medicine, Daegu, Korea
| | | | | | | |
Collapse
|
159
|
Phipps LE, Hino S, Muschel RJ. Targeting cell spreading: a method of sensitizing metastatic tumor cells to TRAIL-induced apoptosis. Mol Cancer Res 2011; 9:249-58. [PMID: 21296861 DOI: 10.1158/1541-7786.mcr-11-0021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) is a current focus for the development of new cancer therapies, because of its selective induction of apoptosis in cancer cells. TRAIL has previously been shown to be important for tumor cell clearance from the liver; however, many cancer cell lines show some resistance toward TRAIL, posing a problem for the future use of TRAIL therapies. In this study, we show that interfering with a cell's ability to attach and spread onto a matrix can sensitize tumor cells to TRAIL-induced apoptosis in vitro. We targeted different members of the integrin signaling pathway using siRNA or inhibitors, including β-integrins, talin, Src, and downstream survival pathways PI3K and MAPK. Targeting any of these molecules could sensitize both MDA-MB-231 human breast cancer cells and TRAIL-resistant 1205Lu melanoma cells to TRAIL-induced apoptosis in vitro. Transcriptionally targeting the cytoskeleton, using myocardin-related transcription factor depletion to disrupt the transcription of cytoskeletal proteins, also caused TRAIL sensitization in MDA-MB-231 cells. We showed that this sensitivity to TRAIL correlated with increased activation of the intrinsic pathway of apoptosis. Manipulation of cell spreading therefore presents a potential method by which disseminated tumor cells could be sensitized to TRAIL therapies in vivo.
Collapse
Affiliation(s)
- Laura E Phipps
- Gray Institute for Radiation Oncology and Biology, ORCRB, Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| | | | | |
Collapse
|
160
|
Sammons V, Davidson A, Tu J, Stoodley MA. Endothelial cells in the context of brain arteriovenous malformations. J Clin Neurosci 2011; 18:165-70. [PMID: 21167719 DOI: 10.1016/j.jocn.2010.04.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 04/14/2010] [Indexed: 10/18/2022]
|
161
|
Abstract
The fourth meeting on dependence receptors featured descriptions of previously unknown dependence receptors. New mechanistic data were presented on the switch between the trophic, antiapoptotic response with the proapoptotic response that occurs with loss of trophic support. The possibility that the loss of trophic support may also involve the binding of an active antitrophin was also discussed. New in vivo data were presented on the roles of dependence receptors in development, angiogenesis, oncogenesis, and neurodegeneration, as well as new therapeutic approaches based on dependence receptor function. The next meeting on dependence receptors is scheduled for 2012.
Collapse
Affiliation(s)
- Patrick Mehlen
- Apoptose, Cancer et Développement, CNRS UMR5538, Centre Léon Bérard, University of Lyon, Lyon 69008, France. dbredesen@buckinstitute
| | | |
Collapse
|
162
|
Intratumoral drug delivery with nanoparticulate carriers. Pharm Res 2011; 28:1819-30. [PMID: 21213021 DOI: 10.1007/s11095-010-0360-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2010] [Accepted: 12/20/2010] [Indexed: 12/25/2022]
Abstract
Stiff extracellular matrix, elevated interstitial fluid pressure, and the affinity for the tumor cells in the peripheral region of a solid tumor mass have long been recognized as significant barriers to diffusion of small-molecular-weight drugs and antibodies. However, their impacts on nanoparticle-based drug delivery have begun to receive due attention only recently. This article reviews biological features of many solid tumors that influence transport of drugs and nanoparticles and properties of nanoparticles relevant to their intratumoral transport, studied in various tumor models. We also discuss several experimental approaches employed to date for enhancement of intratumoral nanoparticle penetration. The impact of nanoparticle distribution on the effectiveness of chemotherapy remains to be investigated and should be considered in the design of new nanoparticulate drug carriers.
Collapse
|
163
|
Abstract
Angiogenesis, the formation of new blood vessel, plays an important role for the growth and metastasis of malignant tumors. The recent identification of specific growth factors for lymphatic vessels and of new lymphatic-specific markers provided evidence for an active role of the lymphatic system during the tumor growth and metastasis processes. Tumor lymphangiogenesis has been shown to play a role in promoting tumor growth and metastasis of tumor cells to distant sites. Integrins play keys roles in the regulation of angiogenesis and lymphangiogenesis during normal development and several diseases. Indeed, integrins control vascular and lymphatic endothelial cell adhesion, migration, and survival. Importantly, integrin inhibitors can block angiogenesis and lymphangiogenesis. In this chapter, we will highlight the role of integrins during angiogenesis and lymphangiogenesis as well as the function of individual integrins during vascular development, postnatal angiogenesis, and lymphangiogenesis. We discuss the role of integrins as potential therapeutic targets for the control of tumor angiogenesis, lymphangiogenesis, and metastatic spread in the treatment of cancer. We also describe methods to analyze expression and function of integrins during angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Philippe Foubert
- Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
164
|
Fis1 and Bap31 bridge the mitochondria-ER interface to establish a platform for apoptosis induction. EMBO J 2010; 30:556-68. [PMID: 21183955 DOI: 10.1038/emboj.2010.346] [Citation(s) in RCA: 396] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/08/2010] [Indexed: 01/21/2023] Open
Abstract
The mitochondria and the endoplasmic reticulum (ER) are two organelles that critically contribute to apoptosis induction. While it is established that they communicate, how cell death signals are transmitted from the mitochondria to the ER is unknown. Here, we show that the mitochondrial fission protein Fission 1 homologue (Fis1) conveys an apoptosis signal from the mitochondria to the ER by interacting with Bap31 at the ER and facilitating its cleavage into the pro-apoptotic p20Bap31. Exogenous apoptosis inducers likewise use this signalling route and induce the procession of Bap31. Moreover, we show that the recruitment of procaspase-8 to the Fis1-Bap31 platform is an early event during apoptosis induction. The association of procaspase-8 with the Fis1-Bap31 complex is dependent on the variant of death effector domain (vDED) in Bap31 and is required for the activation of procaspase-8. This signalling pathway establishes a feedback loop by releasing Ca(2+) from the ER that activates the mitochondria for apoptosis. Hence, the Fis1-Bap31 complex (ARCosome) that spans the mitochondria-ER interface serves as a platform to activate the initiator procaspase-8, and thereby bridges two critical organelles for apoptosis signalling.
Collapse
|
165
|
Zavan B, Paffaro AMA, Joazeiro PP, Yamada AT, Paffaro VA. Immunocytochemical studies of adhesion molecules on mouse UNK cells and their extracellular matrix ligands during mouse pregnancy. Anat Rec (Hoboken) 2010; 293:1081-8. [PMID: 20201059 DOI: 10.1002/ar.21117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Uterine natural killer (uNK) cells are the dominant lymphocytes of pregnant mammals' uterus. Studies have identified four differentiation stage of mouse uNK cells based on Dolichos biflorus lectin cytochemistry, and their distribution showed preferential domain in the uterus through out the pregnancy. This work was done to investigate the expression of alpha5, alpha6, and beta7 integrins on uNK cells and their ligands distribution. Section of mouse uterus from sixth to seventeenth gestational days were submitted to immunocytochemistry and positive reactions for alpha5, alpha6, and beta7 integrins were found on uNK from eighth to tenth gestational days but not after twelfth gestational days. Fibronectin reactions were seemed from sixth to tenth gestational days around uNK from the myometrium and endometrium close to the myometrium. No reaction for fibronectin was seen in the decidualized and nondecidualized endometrium near the placenta. Laminin reaction was seen just in the antimesometrial side. beta7 integrin seems to be the active receptor to bind with VCAM-1 or MAdCAM-1 of endothelial cells, promoting the uNK cross through the vessels. The absence of laminin in an uNK domain suggests these cells are not dependent of laminin and alpha6 integrin for their establishment. However, fibronectin seems to support uNK migration, proliferation, differentiation, and survival in the uterus by binding with alpha5 integrin. The loss of alpha5 integrin ligation by the down regulation of fibronectin could inhibits these events and further studies are need to investigate whether unligated alpha5 can actively and initiate apoptosis, maybe in a caspase 8-dependent way that has been called integrin-mediated death.
Collapse
Affiliation(s)
- Bruno Zavan
- Department of Biomedical Science, UNIFAL-MG, Alfenas, Minas Gerais, Brazil
| | | | | | | | | |
Collapse
|
166
|
Galectin-1 sensitizes carcinoma cells to anoikis via the fibronectin receptor α5β1-integrin. Cell Death Differ 2010; 18:806-16. [PMID: 21113146 DOI: 10.1038/cdd.2010.148] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Anoikis resistance is a hallmark of transformed epithelial cells. Here, we show that treatment of anoikis-resistant carcinoma cell lines with the endogenous lectin galectin-1 (Gal-1) promoted apoptosis via interaction with the unligated fibronectin receptor α(5)β(1)-integrin. Gal-1 efficiency correlated with expression of α(5)β(1)-integrin, and transfection of the α(5)-subunit into deficient cell lines conferred Gal-1 binding and anoikis stimulation. Furthermore, Gal-1 and the α(5)- and β(1)-integrin subunits co-precipitated in Gal-1-stimulated cells undergoing anoikis. Other members of the galectin family failed to be active. The functional interaction between Gal-1 and α(5)β(1)-integrin was glycan dependent with α2,6-sialylation representing a switch-off signal. Desialylation of cell surface glycans resulted in increased electrophoretic mobility of α(5)β(1)-integrin and facilitated Gal-1 binding and anoikis stimulation. On the level of signaling, Gal-1-stimulated anoikis was prevented by filipin, which impaired the internalization of α(5)β(1)-integrin via cholesterol-enriched microdomains, and by pretreatment with a caspase-8 inhibitor. We propose that Gal-1/α(5)β(1)-integrin interaction participates in the control of epithelial integrity and integrin sialylation may enable carcinoma cells to evade this Gal-1-dependent control mechanism.
Collapse
|
167
|
Identification of subtilase cytotoxin (SubAB) receptors whose signaling, in association with SubAB-induced BiP cleavage, is responsible for apoptosis in HeLa cells. Infect Immun 2010; 79:617-27. [PMID: 21098100 DOI: 10.1128/iai.01020-10] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Subtilase cytotoxin (SubAB), which is produced by certain strains of Shiga-toxigenic Escherichia coli (STEC), causes the 78-kDa glucose-regulated protein (GRP78/BiP) cleavage, followed by induction of endoplasmic reticulum (ER) stress, leading to caspase-dependent apoptosis via mitochondrial membrane damage by Bax/Bak activation. The purpose of the present study was to identify SubAB receptors responsible for HeLa cell death. Four proteins, NG2, α2β1 integrin (ITG), L1 cell adhesion molecule (L1CAM), and hepatocyte growth factor receptor (Met), were identified to be SubAB-binding proteins by immunoprecipitation and purification, followed by liquid chromatography-tandem mass spectrometry analysis. SubAB-induced Bax conformational change, Bax/Bak complex formation, caspase activation, and cell death were decreased in β1 ITG, NG2, and L1CAM small interfering RNA-transfected cells, but unexpectedly, BiP cleavage was still observed. Pretreatment of cells with a function-blocking β1 ITG antibody (monoclonal antibody [MAb] P5D2) enhanced SubAB-induced caspase activation; MAb P5D2 alone had no effect on caspase activation. Furthermore, we found that SubAB induced focal adhesion kinase fragmentation, which was mediated by a proteasome-dependent pathway, and caspase activation was suppressed in the presence of proteasome inhibitor. Thus, β1 ITG serves as a SubAB-binding protein and may interact with SubAB-signaling pathways, leading to cell death. Our results raise the possibility that although BiP cleavage is necessary for SubAB-induced apoptotic cell death, signaling pathways associated with functional SubAB receptors may be required for activation of SubAB-dependent apoptotic pathways.
Collapse
|
168
|
Zhang CY, Hu P, Fu D, Wu W, Jia CY, Zhu XC, Wu XZ. 3'-Sulfo-Le(x) is important for regulation of integrin subunit alphaV. Biochemistry 2010; 49:7811-20. [PMID: 20695481 DOI: 10.1021/bi101040k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Carbohydrate structures with a 3'-sulfo betaGal linkage, such as 3'-sulfo-Le(x), can be synthesized by Gal:3-O-sulfotransferase-2 (Gal3ST-2) catalysis, but little is known about their roles in many biological processes. To investigate the role of Gal3ST-2 and its product 3'-sulfo-Le(x), we depleted Gal3ST-2 via siRNA and added exogenous Lewis-x trisaccharide 3'-sulfate sodium salt in human SMMC7721 hepatoma cells. After siRNA transfection, a striking morphological change in SMMC7721 hepatoma cells from polygon to shuttle shape and a significant decrease in the level of adhesion to sL-selectin, HUVEC, fibronectin, vitronectin, and fibrinogen were observed. The expression of integrin subunit alphaV was markedly downregulated, and 3'-sulfated subunit alphaV almost disappeared in the transfectants. The level of cell surface integrin alphaVbeta3 was reduced simultaneously, although total subunit beta3 underwent almost no change. After treatment with exogenous Lewis-x 3'-sulfate, cellular integrin subunit alphaV was upregulated and the level of cell surface integrin alphaVbeta3 was elevated. Interestingly, knockdown of Gal3ST-2 expression effectively inhibited cell proliferation, and the result was significantly correlated with the decrease in the levels of ILK, phosphorylated AKT, and ERK. On the other hand, treatment with Lewis-x trisaccharide 3'-sulfate sodium salt greatly upregulated the phosphorylation of AKT and ERK. Our results also indicated that downregulation of Gal3ST-2 via siRNA transfection was associated with the decrease in the level of expression of anti-apoptotic protein, Bcl-2, with a consequent decrease in the ratios for Bcl-2 to Bax. By exposure to Lewis-x trisaccharide 3'-sulfate sodium salt, the apoptotic response of cells was inhibited. Therefore, Gal3ST-2 and its product, 3'-sulfo-Le(x), were involved in regulation of integrin subunit alphaV and might be associated with cancer cell regulation.
Collapse
Affiliation(s)
- Chun-Yi Zhang
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
169
|
Tung JJ, Kitajewski J. Chloride intracellular channel 1 functions in endothelial cell growth and migration. JOURNAL OF ANGIOGENESIS RESEARCH 2010; 2:23. [PMID: 21040583 PMCID: PMC2993651 DOI: 10.1186/2040-2384-2-23] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 11/01/2010] [Indexed: 12/29/2022]
Abstract
Background Little is known about the role of CLIC1 in endothelium. These studies investigate CLIC1 as a regulator of angiogenesis by in vitro techniques that mimic individual steps in the angiogenic process. Methods Using shRNA against clic1, we determined the role of CLIC1 in primary human endothelial cell behavior. Results Here, we report that reduced CLIC1 expression caused a reduction in endothelial migration, cell growth, branching morphogenesis, capillary-like network formation, and capillary-like sprouting. FACS analysis showed that CLIC1 plays a role in regulating the cell surface expression of various integrins that function in angiogenesis including β1 and α3 subunits, as well as αVβ3 and αVβ5. Conclusions Together, these results indicate that CLIC1 is required for multiple steps of in vitro angiogenesis and plays a role in regulating integrin cell surface expression.
Collapse
Affiliation(s)
- Jennifer J Tung
- Department of Obstetrics/Gynecology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, 1130 St, Nicholas Ave, 926, New York, NY 10032, USA.
| | | |
Collapse
|
170
|
El-Bikai R, Welman M, Margaron Y, Côté JF, Macqueen L, Buschmann MD, Fahmi H, Shi Q, Maghni K, Fernandes JC, Benderdour M. Perturbation of adhesion molecule-mediated chondrocyte-matrix interactions by 4-hydroxynonenal binding: implication in osteoarthritis pathogenesis. Arthritis Res Ther 2010; 12:R201. [PMID: 20977750 PMCID: PMC2991038 DOI: 10.1186/ar3173] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 08/13/2010] [Accepted: 10/26/2010] [Indexed: 12/12/2022] Open
Abstract
Introduction Objectives were to investigate whether interactions between human osteoarthritic chondrocytes and 4-hydroxynonenal (HNE)-modified type II collagen (Col II) affect cell phenotype and functions and to determine the protective role of carnosine (CAR) treatment in preventing these effects. Methods Human Col II was treated with HNE at different molar ratios (MR) (1:20 to 1:200; Col II:HNE). Articular chondrocytes were seeded in HNE/Col II adduct-coated plates and incubated for 48 hours. Cell morphology was studied by phase-contrast and confocal microscopy. Adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and α1β1 integrin at protein and mRNA levels were quantified by Western blotting, flow cytometry and real-time reverse transcription-polymerase chain reaction. Cell death, caspases activity, prostaglandin E2 (PGE2), metalloproteinase-13 (MMP-13), mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB) were assessed by commercial kits. Col II, cyclooxygenase-2 (COX-2), MAPK, NF-κB-p65 levels were analyzed by Western blotting. The formation of α1β1 integrin-focal adhesion kinase (FAK) complex was revealed by immunoprecipitation. Results Col II modification by HNE at MR approximately 1:20, strongly induced ICAM-1, α1β1 integrin and MMP-13 expression as well as extracellular signal-regulated kinases 1 and 2 (ERK1/2) and NF-κB-p65 phosphorylation without impacting cell adhesion and viability or Col II expression. However, Col II modification with HNE at MR approximately 1:200, altered chondrocyte adhesion by evoking cell death and caspase-3 activity. It inhibited α1β1 integrin and Col II expression as well as ERK1/2 and NF-κB-p65 phosphorylation, but, in contrast, markedly elicited PGE2 release, COX-2 expression and p38 MAPK phosphorylation. Immunoprecipitation assay revealed the involvement of FAK in cell-matrix interactions through the formation of α1β1 integrin-FAK complex. Moreover, the modification of Col II by HNE at a 1:20 or approximately 1:200 MR affects parameters of the cell shape. All these effects were prevented by CAR, an HNE-trapping drug. Conclusions Our novel findings indicate that HNE-binding to Col II results in multiple abnormalities of chondrocyte phenotype and function, suggesting its contribution in osteoarthritis development. CAR was shown to be an efficient HNE-snaring agent capable of counteracting these outcomes.
Collapse
Affiliation(s)
- Rana El-Bikai
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur de Montréal, Department of Surgery, University of Montreal, 5400 Gouin Blvd, West, Montreal, QC H4J 1C5, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Stupack DG. Caspase-8 as a therapeutic target in cancer. Cancer Lett 2010; 332:133-40. [PMID: 20817393 DOI: 10.1016/j.canlet.2010.07.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/08/2010] [Accepted: 07/28/2010] [Indexed: 01/25/2023]
Abstract
Caspase-8 is an apical caspase which initiates programmed cell death following death receptor ligation. This central role in apoptosis has prompted significant clinical interest in regulating caspase-8 expression and proteolytic activity. However, caspase-8 has also been found to play a number of non-apoptotic roles in cells, such as promoting activation NF-κB signaling, regulating autophagy and altering endosomal trafficking, and enhancing cellular adhesion and migration. Therefore, depending upon the specific cellular context, caspase-8 may either potentiate or suppress tumor malignancy. Accordingly, a marked heterogeneity exists in the expression patterns of caspase-8 among different tumor types. Therapeutics have been developed which can increase caspase-8 expression, yet it remains unclear whether this approach will be beneficial in all cases. Care is warranted, and the role of caspase-8 should be addressed on a case by case basis.
Collapse
Affiliation(s)
- Dwayne G Stupack
- Department of Pathology & the Moores Cancer Center, University of California San Diego, 3855 Health Sciences Drive MC0803, La Jolla, CA 92093-0803, USA.
| |
Collapse
|
172
|
Seck TM, Melchels FPW, Feijen J, Grijpma DW. Designed biodegradable hydrogel structures prepared by stereolithography using poly(ethylene glycol)/poly(D,L-lactide)-based resins. J Control Release 2010; 148:34-41. [PMID: 20659509 DOI: 10.1016/j.jconrel.2010.07.111] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 07/14/2010] [Accepted: 07/18/2010] [Indexed: 11/28/2022]
Abstract
Designed three-dimensional biodegradable poly(ethylene glycol)/poly(D,L-lactide) hydrogel structures were prepared for the first time by stereolithography at high resolutions. A photo-polymerisable aqueous resin comprising PDLLA-PEG-PDLLA-based macromer, visible light photo-initiator, dye and inhibitor in DMSO/water was used to build the structures. Porous and non-porous hydrogels with well-defined architectures and good mechanical properties were prepared. Porous hydrogel structures with a gyroid pore network architecture showed narrow pore size distributions, excellent pore interconnectivity and good mechanical properties. The structures showed good cell seeding characteristics, and human mesenchymal stem cells adhered and proliferated well on these materials.
Collapse
Affiliation(s)
- Tetsu M Seck
- Institute of Organic Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, D-55099 Mainz, Germany; MIRA Institute for Biomedical Technology and Technical Medicine, and Department of Biomaterials Science and Technology, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - Ferry P W Melchels
- MIRA Institute for Biomedical Technology and Technical Medicine, and Department of Biomaterials Science and Technology, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - Jan Feijen
- MIRA Institute for Biomedical Technology and Technical Medicine, and Department of Biomaterials Science and Technology, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - Dirk W Grijpma
- MIRA Institute for Biomedical Technology and Technical Medicine, and Department of Biomaterials Science and Technology, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands; Department of Biomedical Engineering, University Medical Centre Groningen and University of Groningen, P.O. Box 196, 9700 AD Groningen, The Netherlands.
| |
Collapse
|
173
|
A previously unreported function of β(1)B integrin isoform in caspase-8-dependent integrin-mediated keratinocyte death. J Invest Dermatol 2010; 130:2569-77. [PMID: 20613771 DOI: 10.1038/jid.2010.195] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Integrins regulate adhesive cell-matrix interactions and mediate survival signals. On the other hand, unligated or free cytoplasmic fragments of integrins induce apoptosis in many cell types (integrin-mediated death). We have previously shown that β(1) integrin expression protects keratinocyte stem cells from anoikis, whereas the role of the β(1)B integrin isoform has not been clarified. In this study we report that suspended keratinocytes undergo apoptosis through the activation of caspase-8, independently of the Fas/Fas ligand system. Indeed, anti-β(1) integrin-neutralizing antibodies induced apoptosis in short hairpin RNA Fas-associated death domain-treated cells. Moreover, before and during suspension, caspase-8 directly associated with β(1) integrin, which in turn internalized and progressively degraded, shedding the cytoplasmic domain. β(1)B was expressed only in the cytoplasm in a perinuclear manner and remained unaltered during suspension. At 24 hours, as β(1)A was located close to the nucleus, β(1)B colocalized with β(1)A and coimmunoprecipitated with caspase-8. Caspase-8 was activated earlier in β(1)B integrin-transfected keratinocytes, and these cells underwent a higher rate of apoptosis than mock cells. In contrast, caspase-8 was not activated in small interfering RNA (siRNA) β(1)B-transfected cells. These results indicate that when β(1)A is unligated, β(1)B is responsible for "integrin-mediated death" in human keratinocytes.
Collapse
|
174
|
Cupido T, Spengler J, Ruiz-Rodriguez J, Adan J, Mitjans F, Piulats J, Albericio F. Amide-to-ester substitution allows fine-tuning of the cyclopeptide conformational ensemble. Angew Chem Int Ed Engl 2010; 49:2732-7. [PMID: 20213784 DOI: 10.1002/anie.200907274] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Tommaso Cupido
- Institute for Research in Biomedicine (IRB), Barcelona Science Park (PCB) and CIBER-BBN Networking Centre on Bioengineering, Biomaterials and Nanomedicine, PCB Baldiri Reixac 10, 08028 Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
175
|
Abstract
Dependence receptors form a family of functionally related receptors which are all able to induce two completely opposite intracellular signals depending on the availability of their ligand. Indeed, in its presence, they mediate a positive, classical signal transduction of survival, differentiation or migration but without it, they trigger a negative signal which leads to cell death. The molecular mechanisms involved in triggering cell death in the absence of ligand are starting to be unravelled: dependence receptors are recruited at well-defined domains at the plasma membrane, they trigger cell death through a monomeric form, they are cleaved by caspases and they recruit a caspase activating complex.
Collapse
Affiliation(s)
- Chantal Thibert
- Apoptosis, Cancer and Development Laboratory, Equipe labellisée La Ligue, Université de Lyon, CNRS UMR5238, Lyon, France.
| | | |
Collapse
|
176
|
Irigoyen M, Pajares MJ, Agorreta J, Ponz-Sarvisé M, Salvo E, Lozano MD, Pío R, Gil-Bazo I, Rouzaut A. TGFBI expression is associated with a better response to chemotherapy in NSCLC. Mol Cancer 2010; 9:130. [PMID: 20509890 PMCID: PMC2900244 DOI: 10.1186/1476-4598-9-130] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 05/28/2010] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Lung cancer is one of the most prevalent neoplasias in developed countries. Advances in patient survival have been limited and the identification of prognostic molecules is needed. Resistance to treatment is strongly related to tumor cell adhesion to the extracellular matrix and alterations in the quantity and nature of molecules constituting the tumor cell niche. Recently, transforming growth factor beta-induced protein (TGFBI), an extracellular matrix adaptor protein, has been reported to be differentially expressed in transformed tissues. Loss of TGFBI expression has been described in several cancers including lung carcinoma, and it has been suggested to act as a tumor suppressor gene. RESULTS To address the importance of TGFBI expression in cancer progression, we determined its expression in NSCLC clinical samples using immunohistochemistry. We identified a strong association between elevated TGFBI expression and the response to chemotherapy. Furthermore, we transiently over-expressed and silenced TGFBI in human NSCLC cell lines. Cells over-expressing TGFBI displayed increased sensitivity to etoposide, paclitaxel, cisplatin and gemcitabine. We observed that TGFBI-mediated induction of apoptosis occurred through its binding to alphavbeta3 integrin. We also determined that full-length TGFBI did not induce caspase 3/7 activation but its proteolytic fragments that were < 3 kDa in size, were able to activate caspase 3, 7 and 8. This pro-apoptotic effect was blocked by anti-alphavbeta3 integrin antibodies. CONCLUSIONS The results shown here indicate that TGFBI is a predictive factor of the response to chemotherapy, and suggest the use of TGFBI-derived peptides as possible therapeutic adjuvants for the enhancement of responses to chemotherapy.
Collapse
Affiliation(s)
- Marta Irigoyen
- Division of Oncology, Center for Applied Medical Research, Universidad de Navarra, Pío XII 55, Pamplona, 31008, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Basu B, Biswas S, Wrigley J, Sirohi B, Corrie P. Angiogenesis in cutaneous malignant melanoma and potential therapeutic strategies. Expert Rev Anticancer Ther 2010; 9:1583-98. [PMID: 19895243 DOI: 10.1586/era.09.135] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Metastatic melanoma (MM) carries a dismal prognosis, as it is largely resistant to conventional cytotoxic chemotherapy, biochemotherapy and immunotherapy. There is, therefore, a pressing need to identify new, effective treatments to improve outcomes from MM. Innovative approaches in oncology drug development include anti-angiogenic strategies, in the form of monoclonal antibodies and small-molecule kinase inhibitors. In this review we aim to present current concepts and controversies surrounding the role of angiogenesis and anti-angiogenic therapies in MM, alluding to other tumor types in which increasing knowledge may supply avenues for future directions in melanoma research and management. An overview of angiogenesis and its importance in melanoma progression is presented, highlighting the key molecules that represent potential therapeutic targets. The results of using anti-angiogenic strategies in preclinical and clinical trials are discussed and future perspectives for anti-angiogenic therapies in MM are considered.
Collapse
Affiliation(s)
- Bristi Basu
- Department of Oncology, Oncology Centre, Addenbrooke's Hospital, Cambridge, UK.
| | | | | | | | | |
Collapse
|
178
|
Ford WE, Ren W, Blackmore PF, Schoenbach KH, Beebe SJ. Nanosecond pulsed electric fields stimulate apoptosis without release of pro-apoptotic factors from mitochondria in B16f10 melanoma. Arch Biochem Biophys 2010; 497:82-9. [PMID: 20346344 DOI: 10.1016/j.abb.2010.03.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 03/11/2010] [Accepted: 03/13/2010] [Indexed: 01/01/2023]
Abstract
Nanosecond pulsed electric fields (nsPEFs) eliminates B16f10 melanoma in mice, but cell death mechanisms and kinetics of molecular events of cell death are not fully characterized. Treatment of B16f10 cells in vitro resulted in coordinate increases in active caspases with YO-PRO-1 uptake, calcium mobilization, decreases in mitochondria membrane potential with decreases in forward light scatter (cell size), increases in ADP/ATP ratio, degradation of actin cytoskeleton and membrane blebbing. However, there was no mitochondrial release of cytochrome c, AIF or Smac/DIABLO or generation of reactive oxygen species. Phosphatidylserine externalization was absent and propidium iodide uptake was delayed in small populations of cells. The results indicate that nsPEFs rapidly recruit apoptosis-like mechanisms through the plasma membrane, mimicking the extrinsic apoptosis pathway without mitochondrial amplification yet include activation of initiator and executioner caspases. nsPEFs provide a new cancer therapy that can bypass cancer-associated deregulation of mitochondria-mediated apoptosis in B16f10 melanoma.
Collapse
Affiliation(s)
- Wentia E Ford
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | | | | | | | | |
Collapse
|
179
|
D'Amico G, Robinson SD, Germain M, Reynolds LE, Thomas GJ, Elia G, Saunders G, Fruttiger M, Tybulewicz V, Mavria G, Hodivala-Dilke KM. Endothelial-Rac1 is not required for tumor angiogenesis unless alphavbeta3-integrin is absent. PLoS One 2010; 5:e9766. [PMID: 20339539 PMCID: PMC2842301 DOI: 10.1371/journal.pone.0009766] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 03/01/2010] [Indexed: 12/25/2022] Open
Abstract
Endothelial cell migration is an essential aspect of tumor angiogenesis. Rac1 activity is needed for cell migration in vitro implying a requirement for this molecule in angiogenesis in vivo. However, a precise role for Rac1 in tumor angiogenesis has never been addressed. Here we show that depletion of endothelial Rac1 expression in adult mice, unexpectedly, has no effect on tumor growth or tumor angiogenesis. In addition, repression of Rac1 expression does not inhibit VEGF-mediated angiogenesis in vivo or ex vivo, nor does it affect chemotactic migratory responses to VEGF in 3-dimensions. In contrast, the requirement for Rac1 in tumor growth and angiogenesis becomes important when endothelial beta3-integrin levels are reduced or absent: the enhanced tumor growth, tumor angiogenesis and VEGF-mediated responses in beta3-null mice are all Rac1-dependent. These data indicate that in the presence of alphavbeta3-integrin Rac1 is not required for tumor angiogenesis.
Collapse
Affiliation(s)
- Gabriela D'Amico
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Stephen D. Robinson
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Mitchel Germain
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Louise E. Reynolds
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Gareth J. Thomas
- Centre for Tumour Biology, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - George Elia
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Garry Saunders
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Victor Tybulewicz
- Division of Immune Cell Biology, National Institute for Medical Research, London, United Kingdom
| | - Georgia Mavria
- Cancer Research UK Centre for Cell and Molecular Biology, Institute of Cancer Research, London, United Kingdom
| | - Kairbaan M. Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Institute of Cancer, Bart's and The London, Queen Mary's School of Medicine and Dentistry, London, United Kingdom
| |
Collapse
|
180
|
Cupido T, Spengler J, Ruiz-Rodriguez J, Adan J, Mitjans F, Piulats J, Albericio F. Amide-to-Ester Substitution Allows Fine-Tuning of the Cyclopeptide Conformational Ensemble. Angew Chem Int Ed Engl 2010. [DOI: 10.1002/ange.200907274] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
181
|
Glucose deprivation induces an atypical form of apoptosis mediated by caspase-8 in Bax-, Bak-deficient cells. Cell Death Differ 2010; 17:1335-44. [PMID: 20203689 DOI: 10.1038/cdd.2010.21] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Apoptosis induced by most stimuli proceeds through the mitochondrial pathway. One such stimulus is nutrient deprivation. In this study we studied death induced by glucose deprivation in cells deficient in Bax and Bak. These cells cannot undergo mitochondrial outer membrane permeabilization (MOMP) during apoptosis, but they undergo necrosis when treated with MOMP-dependent apoptotic stimuli. We find in these cells that glucose deprivation, rather than inducing necrosis, triggered apoptosis. Cell death required caspase activation as inhibition of caspases with peptidic inhibitors prevented death. Glucose deprivation-induced death displayed many hallmarks of apoptosis, such as caspase cleavage and activity, phosphatidyl-serine exposure and cleavage of caspase substrates. Neither overexpression of Bcl-xL nor knockdown of caspase-9 prevented death. However, transient or stable knockdown of caspase-8 or overexpression of CrmA inhibited apoptosis. Cell death was not inhibited by preventing death receptor-ligand interactions, by overexpression of c-FLIP or by knockdown of RIPK1. Glucose deprivation induced apoptosis in the human tumor cell line HeLa, which was prevented by knockdown of caspase-8. Thus, we have found that glucose deprivation can induce a death receptor-independent, caspase-8-driven apoptosis, which is engaged to kill cells that cannot undergo MOMP.
Collapse
|
182
|
Goldschneider D, Mehlen P. Dependence receptors: a new paradigm in cell signaling and cancer therapy. Oncogene 2010; 29:1865-82. [DOI: 10.1038/onc.2010.13] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
183
|
Integrin involvement in freeze resistance of androgen-insensitive prostate cancer. Prostate Cancer Prostatic Dis 2010; 13:151-61. [PMID: 20066006 PMCID: PMC2869388 DOI: 10.1038/pcan.2009.59] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cryoablation has emerged as a primary therapy to treat prostate cancer. While effective, the assumption that freezing serves as a ubiquitous lethal stress is challenged by clinical experience and experimental evidence demonstrating time-temperature related cell death dependence. The age-related transformation from an androgen-sensitive (AS) to an androgen-insensitive (AI) phenotype is a major challenge in the management of prostate cancer. AI cells exhibit morphological changes and treatment resistance to many therapies. Since this resistance has been linked with α6β4 integrin overexpression as a result of androgen receptor (AR) loss, we investigated whether α6β4 integrin expression, as a result AR loss, contributes to the reported increased freeze tolerance of AI prostate cancer. A series of studies using AS (LNCaP LP and PC-3 AR) and AI (LNCaP HP and PC-3) cell lines were designed to investigate the cellular mechanisms contributing to variations in freezing response. Investigation into α6β4 integrin expression revealed that AI cell lines overexpressed this protein, thereby altering morphological characteristics and increasing adhesion characteristics. Molecular investigations revealed a significant decrease in caspase 8, 9, and 3 levels AI cells following freezing. Inhibition of α6β4 integrin resulted in increased caspase activity following freezing (similar to AS cells) and enhanced cell death. These data demonstrate that AI cells show an increase in post-freeze susceptibility following inhibition of α6β4 integrin function. Further understanding the role of androgen-receptor related α6β4 integrin expression in prostate cancer cells responses to freezing might lead to novel options for neo-adjunctive treatments targeting the AR signaling pathway.
Collapse
|
184
|
Forsberg EC, Smith-Berdan S. Parsing the niche code: the molecular mechanisms governing hematopoietic stem cell adhesion and differentiation. Haematologica 2010; 94:1477-81. [PMID: 19880773 DOI: 10.3324/haematol.2009.013730] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
185
|
Abstract
The integrin family of cell adhesion receptors regulates a diverse array of cellular functions crucial to the initiation, progression and metastasis of solid tumours. The importance of integrins in several cell types that affect tumour progression has made them an appealing target for cancer therapy. Integrin antagonists, including the alphavbeta3 and alphavbeta5 inhibitor cilengitide, have shown encouraging activity in Phase II clinical trials and cilengitide is currently being tested in a Phase III trial in patients with glioblastoma. These exciting clinical developments emphasize the need to identify how integrin antagonists influence the tumour and its microenvironment.
Collapse
Affiliation(s)
- Jay S Desgrosellier
- Department of Pathology, Moores University of California at San Diego Cancer Center, La Jolla, 92093-0803, United States
| | | |
Collapse
|
186
|
Karoubi G, Ormiston ML, Stewart DJ, Courtman DW. Single-cell hydrogel encapsulation for enhanced survival of human marrow stromal cells. Biomaterials 2009; 30:5445-55. [PMID: 19595454 DOI: 10.1016/j.biomaterials.2009.06.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 06/21/2009] [Indexed: 02/08/2023]
Abstract
Inadequate extracellular matrix cues and subsequent apoptotic cell death are among crucial factors currently limiting cell viability and organ retention in cell-based therapeutic strategies for vascular regeneration. Here we describe the use of a single-cell hydrogel capsule to provide enhanced cell survival of adherent cells in transient suspension culture. Human marrow stromal cells (hMSCs) were singularly encapsulated in agarose capsules containing the immobilized matrix molecules, fibronectin and fibrinogen to ameliorate cell-matrix survival signals. MSCs in the enriched capsules demonstrated increased viability, greater metabolic activity and enhanced cell-cytoskeletal patterning. Increased cell viability resulted from the re-induction of cell-matrix interactions likely via integrin clustering and subsequent activation of the extracellular signal regulated MAPK (ERK)/mitogen activated protein kinase (MAPK) signaling cascade. Proof of principle in-vivo studies, investigating autologous MSC delivery into Fisher 344 rat hindlimb, depicted a significant increase in the number of engrafted cells using the single-cell encapsulation system. Incorporation of immobilized adhesion molecules compensates, at least in part, for the missing cell-matrix cues, thereby attenuating the initial anoikis stimuli and providing protection from subsequent apoptosis. Thus, this single-cell encapsulation strategy may markedly enhance therapeutic cell survival in targeted tissues.
Collapse
Affiliation(s)
- Golnaz Karoubi
- University of Toronto and St. Michael's Hospital, Toronto, ON, Canada
| | | | | | | |
Collapse
|
187
|
Meng Y, Eshghi S, Li YJ, Schmidt R, Schaffer DV, Healy KE. Characterization of integrin engagement during defined human embryonic stem cell culture. FASEB J 2009; 24:1056-65. [PMID: 19933311 DOI: 10.1096/fj.08-126821] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human embryonic stem (hES) cells are pluripotent, capable of differentiating into any cell type of the body, and therefore have the ability to provide insights into mechanisms of human development and disease, as well as to provide a potentially unlimited supply of cells for cell-based therapy and diagnostics. Knowledge of the adhesion receptors that hES cells employ to engage extracellular matrix (ECM) proteins is of basic biological interest and can enhance the design of cell culture and implantation systems to enable these biomedical applications. Although hES cells express a variety of cell surface receptors, little is known about which integrins are involved during subculture and passage. Matrigel is broadly used as a cell adhesive matrix for hES cell culture. Here, we sought to identify which integrins hES cells exploit for adhesion to Matrigel-coated surfaces in defined medium conditions. Using RT-PCR, flow cytometry, and fluorescence immunochemistry, we found that numerous integrins were expressed by H1 hES cells; however, antibody blocking assays indicated that only alpha(v)beta(3), alpha(6), beta(1), and alpha(2)beta(1) played a significant role in the initial adhesion of the hES cells to Matrigel in defined medium conditions. We subsequently identified a cohort of synthetic peptides that, when adsorbed to the culture surface, promoted H1 hES cell attachment and proliferation, as well as maintained a pluripotent phenotype. Peptides designed to engage with alpha(v)beta(3), alpha(6), beta(1), and alpha(2)beta(1) integrins and syndecan-1 were tested both individually and in various combinations. A combination of two integrin-engaging peptides (AG-10, C-16) and one syndecan-engaging peptide (AG-73) was sufficient to promote hES cell adhesion, maintenance, and proliferation. We propose that a specific integrin "fingerprint" is necessary for maintenance of hES cell self-renewal, and synthetic culture systems must capture this engagement profile for hES cells to remain undifferentiated.-Meng, Y., Eshghi, S., Li, Y. J., Schmidt, R., Schaffer, D. V., Healy, K. E. Characterization of integrin engagement during defined human embryonic stem cell culture.
Collapse
Affiliation(s)
- Ying Meng
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA 94720-1760, USA
| | | | | | | | | | | |
Collapse
|
188
|
Torres VA, Mielgo A, Barbero S, Hsiao R, Wilkins JA, Stupack DG. Rab5 mediates caspase-8-promoted cell motility and metastasis. Mol Biol Cell 2009; 21:369-76. [PMID: 19923319 PMCID: PMC2808229 DOI: 10.1091/mbc.e09-09-0769] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Integrins signaling promotes nonapoptotic functions of caspase-8 via activation of small GTPases from the Rab and Rac families. Integrin ligation promotes Rab5 activity, which mediates subsequent activation of Rac1, cytoskeletal remodeling, and enhanced cell motility. Caspase-8 is a key apical sensory protein that governs cell responses to environmental cues, alternatively promoting apoptosis, proliferation, and cell migration. The proteins responsible for integration of these pathways, however, have remained elusive. Here, we reveal that Rab5 regulates caspase-8–dependent signaling from integrins. Integrin ligation leads to Rab5 activation, association with integrins, and activation of Rac, in a caspase-8–dependent manner. Rab5 activation promotes colocalization and coprecipitation of integrins with caspase-8, concomitant with Rab5 recruitment to integrin-rich regions such as focal adhesions and membrane ruffles. Moreover, caspase-8 expression promotes Rab5-mediated internalization and the recycling of β1 integrins, increasing cell migration independently of caspase catalytic activity. Conversely, Rab5 knockdown prevented caspase-8–mediated integrin signaling for Rac activation, cell migration, and apoptotic signaling, respectively. Similarly, Rab5 was critical for caspase-8–driven cell migration in vivo, because knockdown of Rab5 compromised the ability of caspase-8 to promote metastasis under nonapoptotic conditions. These studies identify Rab5 as a key integrator of caspase-8–mediated signal transduction downstream of integrins, regulating cell survival and migration in vivo and in vitro.
Collapse
Affiliation(s)
- Vicente A Torres
- Department of Pathology, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
189
|
Degaki TL, Demasi MAA, Sogayar MC. Overexpression of Nrp/b (nuclear restrict protein in brain) suppresses the malignant phenotype in the C6/ST1 glioma cell line. J Steroid Biochem Mol Biol 2009; 117:107-16. [PMID: 19682578 DOI: 10.1016/j.jsbmb.2009.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 07/28/2009] [Accepted: 07/29/2009] [Indexed: 12/13/2022]
Abstract
Upon searching for glucocorticoid-regulated cDNA sequences associated with the transformed to normal phenotypic reversion of C6/ST1 rat glioma cells, we identified Nrp/b (nuclear restrict protein in brain) as a novel rat gene. Here we report on the identification and functional characterization of the complete sequence encoding the rat NRP/B protein. The cloned cDNA presented a 1767 nucleotides open-reading frame encoding a 589 amino acids residues sequence containing a BTB/POZ (broad complex Tramtrack bric-a-brac/Pox virus and zinc finger) domain in its N-terminal region and kelch motifs in its C-terminal region. Sequence analysis indicates that the rat Nrp/b displays a high level of identity with the equivalent gene orthologs from other organisms. Among rat tissues, Nrp/b expression is more pronounced in brain tissue. We show that overexpression of the Nrp/b cDNA in C6/ST1 cells suppresses anchorage independence in vitro and tumorigenicity in vivo, altering their malignant nature towards a more benign phenotype. Therefore, Nrp/b may be postulated as a novel tumor suppressor gene, with possible relevance for glioblastoma therapy.
Collapse
Affiliation(s)
- Theri Leica Degaki
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
190
|
Francavilla C, Maddaluno L, Cavallaro U. The functional role of cell adhesion molecules in tumor angiogenesis. Semin Cancer Biol 2009; 19:298-309. [DOI: 10.1016/j.semcancer.2009.05.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 05/20/2009] [Indexed: 12/18/2022]
|
191
|
An integrin alpha(v)beta(3)-c-Src oncogenic unit promotes anchorage-independence and tumor progression. Nat Med 2009; 15:1163-9. [PMID: 19734908 PMCID: PMC2759406 DOI: 10.1038/nm.2009] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 06/19/2009] [Indexed: 01/13/2023]
Abstract
Integrins regulate adhesion-dependent growth, survival and invasion of tumor cells. In particular, expression of integrin alpha(v)beta(3) is associated with progression of a variety of human tumors. Here we reveal a previously undescribed adhesion-independent role for integrin alpha(v)beta(3) in pancreatic cancer and other carcinomas. Specifically, alpha(v)beta(3) expressed in carcinoma cells enhanced anchorage-independent tumor growth in vitro and increased lymph node metastases in vivo. These effects required recruitment of c-Src to the beta(3) integrin cytoplasmic tail, leading to c-Src activation, Crk-associated substrate (CAS) phosphorylation and tumor cell survival that, unexpectedly, was independent of cell adhesion or focal adhesion kinase (FAK) activation. Pharmacological blockade of c-Src kinase activity or decreased expression of endogenous alpha(v)beta(3) integrin or c-Src not only inhibited anchorage-independent growth but also suppressed metastasis in vivo, yet these manipulations did not affect tumor cell migration or invasion. These data define an unexpected role for an integrin as a mediator of anchorage independence, suggesting that an alpha(v)beta(3)-c-Src signaling module may account for the aggressive behavior of integrin alpha(v)beta(3)-expressing tumors in humans.
Collapse
|
192
|
Westhoff MA, Fulda S. Adhesion-mediated apoptosis resistance in cancer. Drug Resist Updat 2009; 12:127-36. [PMID: 19726220 DOI: 10.1016/j.drup.2009.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 06/14/2009] [Accepted: 08/03/2009] [Indexed: 01/06/2023]
Abstract
Adhesion-mediated apoptosis resistance (AMAR) is an emerging concept that may explain the observed differences in survival between cells within the three-dimensional structure of a tumor and the standard monolayer culture conditions in the laboratory. Not only the cancer cells' motility and invasiveness are different in a three-dimensional tumor, but - crucially - the cells' sensitivity towards apoptosis, a form of programmed cell death, varies widely between the in vivo and in vitro situation. Tumor cells interacting either with a specific extracellular matrix protein substrate or with each other or with non-transformed cells, such as fibroblasts, exhibit increased resistance towards a wide variety of therapeutic approaches. In this review we discuss the molecular basis of these interactions and the main downstream effectors that are involved in the enhancement of the tumor cells' survival. In particular, we show that the pathways activated by adhesion are not unique, but involve the MAPK/ERK and PI3K/Akt pathways, which are reused between different forms of AMAR and are also found in adhesion-independent modes of resistance. Thus, the tools to overcome AMAR are already at our disposal and using them in this novel context of AMAR should lead to significant therapeutic benefit.
Collapse
|
193
|
Hasegawa A, Yamada C, Tani M, Hirano SI, Tokumoto Y, Miyake J. Caspase inhibitors increase the rate of recovery of neural stem/progenitor cells from post-mortem rat brains stored at room temperature. J Biosci Bioeng 2009; 107:652-7. [PMID: 19447344 DOI: 10.1016/j.jbiosc.2009.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 01/09/2009] [Accepted: 01/27/2009] [Indexed: 12/11/2022]
Abstract
To match the demand of regenerative medicine for nerve system, collection of stem cells from the post-mortem body is one of the most practical ways. In this study, the storage condition of the post-mortem body was examined. We prepared neural stem/progenitor cells (NSPCs) from post-mortem rat brains stored at different temperatures. When brains were stored at 4 degrees C, for one week, we were able to obtain neurospheres (a spheroid body containing NSPCs) by stimulation of cells with epidermal growth factor (EGF). Incremental increases in storage temperature decreased the rate of appearance of neurospheres. Within 48 h at 15 degrees C, 24 h at 25 degrees C, in both condition, we were able to recover NSPCs from post-mortem rat brains. At 15 degrees C, 90% of neurosphere-forming activity was lost within 24 h. However, even after 24 h at 25 degrees C, 2% neurosphere-forming activity remained. After 6 h of death, there was very little difference between the rates of NSPC recovery at 4 degrees C and 25 degrees C. Addition of caspase inhibitors to both the rat brain storage solution and the NSPC culture medium increased the rate of neurosphere-forming activity. In particular, an inhibitor of caspase-8 activity increased the NSPC recovery rate approximately three-fold, with no accompanying detrimental effects on neural differentiation in vitro.
Collapse
Affiliation(s)
- Atsuko Hasegawa
- Department of Tissue Engineering and Cell Therapy, Institute of Biomedical Research and Innovation, 1-5-4 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | | | | | | | | | | |
Collapse
|
194
|
Paclitaxel promotes a caspase 8-mediated apoptosis through death effector domain association with microtubules. Oncogene 2009; 28:3551-62. [PMID: 19668227 PMCID: PMC2851247 DOI: 10.1038/onc.2009.210] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microtubule-perturbing drugs have become front line chemotherapeutics, inducing cell cycle crisis as a major mechanism of action. However, these agents exhibit pleiotropic effects on cells, and can induce apoptosis via other means. Paclitaxel, a microtubule-stabilizing agent, induces a caspase-dependent apoptosis, though the precise mechanism(s) remain unclear. Here, we used genetic approaches to evaluate the role of caspase 8 in paclitaxel-mediated apoptosis. We observed that caspase 8-expressing cells are more sensitive to paclitaxel than caspase 8-deficient cells. Mechanistically, caspase 8 was found associated with microtubules, and this interaction increased following paclitaxel-treatment. The prodomains (DEDs) of caspase 8 were sufficient for interaction with microtubules, but the caspase 8 holoprotein was required for apoptosis. DED-only forms of caspase 8 were found in both primary and tumor cell lines, associating with perinuclear microtubules and the centrosome. Microtubule-association, and paclitaxel-sensitivity, depends upon a critical lysine (K156) within a microtubule-binding motif (KLD) in DED-b of caspase 8. The results reveal an unexpected pathway of apoptosis mediated by caspase 8.
Collapse
|
195
|
Hehlgans S, Eke I, Storch K, Haase M, Baretton GB, Cordes N. Caveolin-1 mediated radioresistance of 3D grown pancreatic cancer cells. Radiother Oncol 2009; 92:362-70. [PMID: 19665245 DOI: 10.1016/j.radonc.2009.07.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 06/30/2009] [Accepted: 07/03/2009] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Resistance of pancreatic ductal adenocarcinoma (PDAC) to chemo- and radiotherapy is a major obstacle. The integral membrane protein Caveolin-1 (Cav-1) has been suggested as a potent target in human pancreatic carcinoma cells. MATERIALS AND METHODS Human pancreatic tumor cells were examined in a three-dimensional (3D) cell culture model with regard to clonogenic survival, apoptosis, radiogenic DNA-double strand breaks and protein expression and phosphorylation under siRNA-mediated knockdown of Cav-1 without and in combination with irradiation (X-rays, 0-6Gy). Immunohistochemistry was used to assess Cav-1 expression in biopsies from patients with PDAC. RESULTS Tumor cells in PDAC showed significantly higher Cav-1 expression relative to tumor stroma. Cav-1 knockdown significantly reduced beta1 integrin expression and Akt phosphorylation, induced Caspase 3- and Caspase 8-dependent apoptosis and enhanced the radiosensitivity of 3D cell cultures. While cell cycling and Cav-1 promoter activity remained stable, Cav-1 knockdown-induced radiosensitization correlated with elevated numbers of residual DNA-double strand breaks. CONCLUSIONS Our data strongly support the concept of Cav-1 as a potent target in pancreatic carcinoma cells due to radiosensitization and Cav-1 overexpression in tumor cells of PDAC. 3D cell cultures are powerful and useful tools for the testing of novel targeting strategies to optimize conventional radio- and chemotherapy regimes for PDAC.
Collapse
Affiliation(s)
- Stephanie Hehlgans
- OncoRay-Center for Radiation Research in Oncology, Dresden University of Technology, Dresden, Germany
| | | | | | | | | | | |
Collapse
|
196
|
Gorlov IP, Byun J, Gorlova OY, Aparicio AM, Efstathiou E, Logothetis CJ. Candidate pathways and genes for prostate cancer: a meta-analysis of gene expression data. BMC Med Genomics 2009; 2:48. [PMID: 19653896 PMCID: PMC2731785 DOI: 10.1186/1755-8794-2-48] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 08/04/2009] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The genetic mechanisms of prostate tumorigenesis remain poorly understood, but with the advent of gene expression array capabilities, we can now produce a large amount of data that can be used to explore the molecular and genetic mechanisms of prostate tumorigenesis. METHODS We conducted a meta-analysis of gene expression data from 18 gene array datasets targeting transition from normal to localized prostate cancer and from localized to metastatic prostate cancer. We functionally annotated the top 500 differentially expressed genes and identified several candidate pathways associated with prostate tumorigeneses. RESULTS We found the top differentially expressed genes to be clustered in pathways involving integrin-based cell adhesion: integrin signaling, the actin cytoskeleton, cell death, and cell motility pathways. We also found integrins themselves to be downregulated in the transition from normal prostate tissue to primary localized prostate cancer. Based on the results of this study, we developed a collagen hypothesis of prostate tumorigenesis. According to this hypothesis, the initiating event in prostate tumorigenesis is the age-related decrease in the expression of collagen genes and other genes encoding integrin ligands. This concomitant depletion of integrin ligands leads to the accumulation of ligandless integrin and activation of integrin-associated cell death. To escape integrin-associated death, cells suppress the expression of integrins, which in turn alters the actin cytoskeleton, elevates cell motility and proliferation, and disorganizes prostate histology, contributing to the histologic progression of prostate cancer and its increased metastasizing potential. CONCLUSION The results of this study suggest that prostate tumor progression is associated with the suppression of integrin-based cell adhesion. Suppression of integrin expression driven by integrin-mediated cell death leads to increased cell proliferation and motility and increased tumor malignancy.
Collapse
Affiliation(s)
- Ivan P Gorlov
- Department of Genitourinary Medical Oncology, The University of Texas M, D, Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | | | |
Collapse
|
197
|
Alghisi GC, Rüegg C. Vascular Integrins in Tumor Angiogenesis: Mediators and Therapeutic Targets. ACTA ACUST UNITED AC 2009; 13:113-35. [PMID: 16728329 DOI: 10.1080/10623320600698037] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The notion that tumor angiogenesis may have therapeutic implications in the control of tumor growth was introduced by Dr. Judah Folkman in 1971. The approval of Avastin in 2004 as the first antiangiogenic systemic drug to treat cancer patients came as a validation of this visionary concept and opened new perspectives to the treatment of cancer. In addition, this success boosted the field to the quest for new therapeutic targets and antiangiogenic drugs. Preclinical and clinical evidence indicate that vascular integrins may be valid therapeutic targets. In preclinical studies, pharmacological inhibition of integrin function efficiently suppressed angiogenesis and inhibited tumor progression. alphaVbeta3 and alphaVbeta5 were the first vascular integrins targeted to suppress tumor angiogenesis. Subsequent experiments revealed that at least four additional integrins (i.e., alpha1beta1, alpha2beta1, alpha5beta1, and alpha6beta4) might be potential therapeutic targets. In clinical studies low-molecular-weight integrin inhibitors and anti-integrin function-blocking antibodies demonstrated low toxicity and good tolerability and are now being tested in combination with radiotherapy and chemotherapy for anticancer activity in patients. In this article the authors review the role of integrins in angiogenesis, present recent development in the use of alphaVbeta3 and alpha5beta1 integrin antagonists as potential therapeutics in cancer, and discuss future perspectives.
Collapse
Affiliation(s)
- Gian Carlo Alghisi
- Centre Pluridisciplinaire d'Oncologie (CePO), Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | | |
Collapse
|
198
|
Abstract
BACKGROUND Integrins are a family of transmembrane receptors that mediate cell-cell and cell-matrix adhesion. They are involved in stable cell adhesion and migration of cells. In addition, integrin-mediated interactions modulate the response to most, if not all growth factors, cytokines, and other soluble factors. PURPOSE In this review, we briefly explain how integrins can affect the multitude of signal transduction cascades in control of survival, proliferation, and differentiation. Subsequently, we primarily focus on targeting integrins alpha5beta1 and alphanubeta3 in disease and we discuss how antagonists of these integrins, including disintegrins, RGD peptides, small molecules, and function blocking antibodies, may be of therapeutical value either alone or, especially in the treatment of cancer, in combination with existing therapeutical strategies.
Collapse
Affiliation(s)
- Stephan Huveneers
- Division of Toxicology, Leiden/Amsterdam Center for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | |
Collapse
|
199
|
Bredesen DE. Neurodegeneration in Alzheimer's disease: caspases and synaptic element interdependence. Mol Neurodegener 2009; 4:27. [PMID: 19558683 PMCID: PMC2709109 DOI: 10.1186/1750-1326-4-27] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 06/26/2009] [Indexed: 11/10/2022] Open
Abstract
Extensive genetic, biochemical, and histological evidence has implicated the amyloid-β peptide (Aβ) in Alzheimer's disease pathogenesis, and several mechanisms have been suggested, such as metal binding, reactive oxygen species production, and membrane pore formation. However, recent evidence argues for an additional role for signaling mediated by the amyloid precursor protein, APP, in part via the caspase cleavage of APP at aspartate 664. Here we review the effects and implications of this cleavage event, and propose a model of Alzheimer's disease that focuses on the critical nature of this cleavage and its downstream effects.
Collapse
Affiliation(s)
- Dale E Bredesen
- Buck Institute for Age Research, 8001 Redwood Blvd,, Novato, CA USA 94945.
| |
Collapse
|
200
|
Interfering with multimerization of netrin-1 receptors triggers tumor cell death. Cell Death Differ 2009; 16:1344-51. [PMID: 19543238 DOI: 10.1038/cdd.2009.75] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Netrin-1 was recently proposed to control tumorigenesis by inhibiting apoptosis induced by the dependence receptors DCC (Deleted in colorectal cancer) and UNC5H. Although the loss of these dependence receptors' expression has been described as a selective advantage for tumor growth and progression in numerous cancers, recent observations have shown that some tumors may use an alternative strategy to block dependence receptor-induced programmed cell death: the autocrine expression of netrin-1. This alternative strategy has been observed in a large fraction of aggressive breast cancers, neuroblastoma, pancreatic adenocarcinoma, and lung cancer. This observation is of potential interest regarding future targeted therapy, as in such cases interfering with the ability of netrin-1 to inhibit DCC or UNC5H-induced cell death is associated with apoptosis of netrin-1-expressing tumor cells in vitro, and with inhibition of tumor growth or metastasis in different animal tumor models. The understanding of the mechanism by which netrin-1 inhibits cell death is therefore of interest. Here, we show that netrin-1 triggers the multimerization of both DCC and UNC5H2 receptors, and that multimerization of the intracellular domain of DCC and UNC5H2 is the critical step to inhibit the proapoptotic effects of both of these receptors. Taking advantage of this property, we utilized a recombinant specific domain of DCC that (i) interacts with netrin-1 and (ii) inhibits netrin-1-induced multimerization, to trigger apoptosis in netrin-dependent tumor cells.
Collapse
|