401
|
Affiliation(s)
- Erkki Ruoslahti
- The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
402
|
Miura M, Garcia FL, Crawford SE, Rowley AH. Cell adhesion molecule expression in coronary artery aneurysms in acute Kawasaki disease. Pediatr Infect Dis J 2004; 23:931-6. [PMID: 15602193 DOI: 10.1097/01.inf.0000142171.91235.fc] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The pathogenesis of coronary artery aneurysm (CAA) formation in acute Kawasaki disease (KD) remains unclear. Cell adhesion molecules mediate cell-cell and cell-matrix interactions and regulate leukocyte migration, angiogenesis and tissue remodeling. We hypothesized that cell adhesion molecules are expressed in acute KD CAA. METHODS : P-selectin, E-selectin, vascular cell adhesion molecule-1 (VCAM-1) and integrin beta1 were immunolocalized in coronary arteries from 6 acute KD patients and 7 controls. RESULTS In endothelial cells of adventitial neovasculature in KD CAA, P-selectin and integrin beta1 were expressed in all of 6 patients, and E-selectin and/or VCAM-1 were expressed in 4 of 6. Endothelial cells in controls and in nonaneurysmal KD coronary arteries expressed P-selectin and integrin beta1, but not E-selectin or VCAM-1. Integrin beta1 was expressed on infiltrating leukocytes in 5 of 6 KD CAA and on fibroblasts in 6 of 6; these findings were absent in controls and in nonaneurysmal KD coronary arteries. CONCLUSIONS The lack of widespread expression of E-selectin or VCAM-1 on endothelial cells of acute KD coronary arteries was surprising and suggests that inflammatory cell infiltration into KD coronaries is not simply the result of widespread up-regulation of cell adhesion molecules on endothelial cells by circulating cytokines. Rather, inflammatory cells may be directed to specific areas of the coronary arteries targeted by a pathogen causing KD. Our results suggest that E-selectin and VCAM-1 expression on neovasculature may contribute to neoangiogenesis and prolonged CAA inflammation and that integrin beta1 might be involved in fibroblastic remodeling of acute KD CAA.
Collapse
Affiliation(s)
- Masaru Miura
- Department of Pediatrics, Northwestern University, The Feinberg School of Medicine and The Children's Memorial Hospital, Chicago, IL, USA
| | | | | | | |
Collapse
|
403
|
Abstract
Integrins are cell adhesion molecules that play an important role in the regulation of angiogenesis. In this overview, the vascular integrins and their mechanisms of action are outlined. Integrins have been evaluated in preclinical and clinical studies for the treatment of cancer and as diagnostic markers of angiogenesis. Furthermore, integrins are the basis for targeted therapy for solid tumors and novel imaging techniques to assess the angiogenic response of tumors.
Collapse
Affiliation(s)
- Rosa Hwang
- Department of Surgery, University of California San Diego, Moores Comprehensive Cancer Center, 9500 Gilman Drive, La Jolla, CA 92093-0987, USA
| | | |
Collapse
|
404
|
Xu H, Buck SM, Kopelman R, Philbert MA, Brasuel M, Ross BD, Rehemtulla A. Photoexcitation-Based Nano-Explorers: Chemical Analysis inside Live Cells and Photodynamic Therapy. Isr J Chem 2004. [DOI: 10.1560/wa5h-kbgv-pr13-nevn] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
405
|
Morohashi H, Abe F, Saiga K, Toyoda E, Ichimura E, Nishikawa K. NK95806, a newly synthesized microtubule-disrupting agent that suppresses collagen-induced arthritis in mice. Int Immunopharmacol 2004; 4:1437-43. [PMID: 15351313 DOI: 10.1016/j.intimp.2004.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Accepted: 06/18/2004] [Indexed: 11/20/2022]
Abstract
We investigated the anti-arthritic effects of NK95806, a novel inhibitor of microtubule polymerization, on collagen-induced arthritis in mice. The suppressive effect of NK95806 on the induction and development of arthritis was shown as a significant reduction in clinical arthritis scores. Histological analysis of the hind paws confirmed the improvement in clinical severity and showed marked decreases in granulomatous formation and further bone destruction. Further, under the experimental conditions in which methotrexate had little, if any, effect, NK95806 significantly suppressed the development of arthritis. These results suggest that the disruption of microtubules might be a novel target for anti-rheumatic drugs and NK95806 may be a candidate for further development.
Collapse
Affiliation(s)
- Hirohisa Morohashi
- R&D division, Pharmaceutical group, Nippon Kayaku Co., Ltd., 31-12 Shimo 3-chome, Kita-ku, Tokyo, 115-8588 Japan
| | | | | | | | | | | |
Collapse
|
406
|
Liu C, Dickinson C, Shobe J, Doñate F, Ruf W, Edgington T. A hybrid fibronectin motif protein as an integrin targeting selective tumor vascular thrombogen. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.793.3.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Targeted thrombotic eradication of solid tumors is a novel therapeutic strategy. The feasibility, efficacy, selectivity, and safety are dependent on multiple variables of protein design, molecular assembly, vascular target, and exclusive restriction of function to the tumor vasculature. To advance this strategy, we describe a design of an integrin targeting selective tumor vascular thrombogen. We adopted the fibronectin structural motif of tandem repeating modules with four type III repeat modules of fibronectin followed by two structurally homologous modules of the extracellular domain of tissue factor. This hybrid protein of six tandem modules recognizes integrins and selectively docks and initiates the thrombogenic protease cascade locally on the target cell surfaces. The protein is inactive in blood but is functionally active once assembled on integrin-positive cells. When administered i.v. to tumor-bearing mice, it selectively induces extensive local microthrombosis of the tumor microvasculature. The principles are addressed from the perspective of protein structural design for a class of selective tumor vascular thrombogen proteins that, through interaction with tumor angiogenic endothelium, elicit thrombotic occlusion rather than apoptosis or arrest of angiogenesis. This response can produce local tumor infarction followed by intratumoral ischemia-reperfusion injury, inflammation, and a local host tumor eradicative response.
Collapse
Affiliation(s)
- Cheng Liu
- 1Department of Immunology, Scripps Research Institute, La Jolla, California
| | | | - Justin Shobe
- 1Department of Immunology, Scripps Research Institute, La Jolla, California
| | | | - Wolfram Ruf
- 1Department of Immunology, Scripps Research Institute, La Jolla, California
| | - Thomas Edgington
- 1Department of Immunology, Scripps Research Institute, La Jolla, California
| |
Collapse
|
407
|
Sottile J. Regulation of angiogenesis by extracellular matrix. Biochim Biophys Acta Rev Cancer 2004; 1654:13-22. [PMID: 14984764 DOI: 10.1016/j.bbcan.2003.07.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2003] [Accepted: 07/04/2003] [Indexed: 10/26/2022]
Abstract
During angiogenesis, endothelial cell growth, migration, and tube formation are regulated by pro- and anti-angiogenic factors, matrix-degrading proteases, and cell-extracellular matrix interactions. Temporal and spatial regulation of extracellular matrix remodeling events allows for local changes in net matrix deposition or degradation, which in turn contributes to control of cell growth, migration, and differentiation during different stages of angiogenesis. Remodeling of the extracellular matrix can have either pro- or anti-angiogenic effects. Extracellular matrix remodeling by proteases promotes cell migration, a critical event in the formation of new vessels. Matrix-bound growth factors released by proteases and/or by angiogenic factors promote angiogenesis by enhancing endothelial migration and growth. Extracellular matrix molecules, such as thrombospondin-1 and -2, and proteolytic fragments of matrix molecules, such as endostatin, can exert anti-angiogenic effects by inhibiting endothelial cell proliferation, migration and tube formation. In contrast, other matrix molecules promote endothelial cell growth and morphogenesis, and/or stabilize nascent blood vessels. Hence, extracellular matrix molecules and extracellular matrix remodelling events play a key role in regulating angiogenesis.
Collapse
Affiliation(s)
- Jane Sottile
- Center for Cardiovascular Research, Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 679, Rochester, NY 14642, USA.
| |
Collapse
|
408
|
Rüegg C, Dormond O, Mariotti A. Endothelial cell integrins and COX-2: mediators and therapeutic targets of tumor angiogenesis. Biochim Biophys Acta Rev Cancer 2004; 1654:51-67. [PMID: 14984767 DOI: 10.1016/j.bbcan.2003.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2003] [Revised: 09/03/2003] [Accepted: 09/03/2003] [Indexed: 02/08/2023]
Abstract
Vascular integrins are essential regulators and mediators of physiological and pathological angiogenesis, including tumor angiogenesis. Integrins provide the physical interaction with the extracellular matrix (ECM) necessary for cell adhesion, migration and positioning, and induce signaling events essential for cell survival, proliferation and differentiation. Integrins preferentially expressed on neovascular endothelial cells, such as alphaVbeta3 and alpha5beta1, are considered as relevant targets for anti-angiogenic therapies. Anti-integrin antibodies and small molecular integrin inhibitors suppress angiogenesis and tumor progression in many animal models, and are currently tested in clinical trials as anti-angiogenic agents. Cyclooxygense-2 (COX-2), a key enzyme in the synthesis of prostaglandins and thromboxans, is highly up-regulated in tumor cells, stromal cells and angiogenic endothelial cells during tumor progression. Recent experiments have demonstrated that COX-2 promotes tumor angiogenesis. Chronic intake of nonsteroidal anti-inflammatory drugs and COX-2 inhibitors significantly reduces the risk of cancer development, and this effect may be due, at least in part, to the inhibition of tumor angiogenesis. Endothelial cell COX-2 promotes integrin alphaVbeta3-mediated endothelial cell adhesion, spreading, migration and angiogenesis through the prostaglandin-cAMP-PKA-dependent activation of the small GTPase Rac. In this article, we review the role of integrins and COX-2 in angiogenesis, their cross talk, and discuss implications relevant to their targeting to suppress tumor angiogenesis.
Collapse
Affiliation(s)
- Curzio Rüegg
- Centre Pluridisciplinaire d'Oncologie, University of Lausanne Medical School, CH-1011 Lausanne, Switzerland.
| | | | | |
Collapse
|
409
|
Shannon KE, Keene JL, Settle SL, Duffin TD, Nickols MA, Westlin M, Schroeter S, Ruminski PG, Griggs DW. Anti-metastatic properties of RGD-peptidomimetic agents S137 and S247. Clin Exp Metastasis 2004; 21:129-38. [PMID: 15168730 DOI: 10.1023/b:clin.0000024764.93092.5f] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Integrins expressed on endothelial cells modulate cell migration and survival during angiogenesis. Integrins expressed on carcinoma cells potentiate metastasis by facilitating invasion and movement across blood vessels. We describe the activities of two synthetic low-molecular-weight peptidomimetics of the ligand amino acid sequence arg-gly-asp (RGD) in integrin-based functional assays in vitro. We also evaluate efficacy and potential mechanisms of action in models of both spontaneous and experimental metastasis. Broad-spectrum potency against the family of alpha v subunit-containing integrins was observed, with significantly less potency against alpha5beta1 and alpha(IIb)beta3. Both endothelial and tumor cell migration mediated by alpha(v)beta3 was inhibited, whereas proliferation of endothelial cells but not tumor cells was diminished. Continuous infusion of compound by minipumps or oral administration twice daily significantly reduced metastatic tumor burden in the lungs of mice despite no reduction in growth of 435/HAL primary tumors, and only a slight reduction in tumor cells detected in circulating blood. Delaying treatment in this model until after extensive dissemination of tumor cells to the lungs had occurred, and after primary tumor resection, still produced significant efficacy. Conversely, administration of the agent for only the first 18 h after tumor-cell inoculation into the tail vein also resulted in decreased metastases observed after several weeks. These data suggest these compounds or their relatives have potential to interfere with both early and late steps of metastasis involving tumor and endothelial cell functions. Furthermore, the metastatic process can be effectively inhibited independently of primary tumor growth using integrin antagonists.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Breast Neoplasms/pathology
- Carcinoma, Ductal/drug therapy
- Carcinoma, Ductal/pathology
- Carcinoma, Ductal/prevention & control
- Carcinoma, Ductal/secondary
- Carcinoma, Ductal/surgery
- Cell Division/drug effects
- Cell Line, Tumor/transplantation
- Cell Movement/drug effects
- Colonic Neoplasms/pathology
- Dipeptides/administration & dosage
- Dipeptides/pharmacology
- Dipeptides/therapeutic use
- Drug Administration Schedule
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelium, Vascular/cytology
- Female
- Humans
- Infusion Pumps, Implantable
- Integrin alphaVbeta3/antagonists & inhibitors
- Lung Neoplasms/drug therapy
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Male
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Neoplasm Proteins/antagonists & inhibitors
- Neoplastic Cells, Circulating
- Oligopeptides
- Organic Chemicals/administration & dosage
- Organic Chemicals/pharmacology
- Organic Chemicals/therapeutic use
- Pyrimidines/administration & dosage
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kristen E Shannon
- Pfizer Global Research and Development, Chesterfield, Missouri 63017, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
410
|
Chavakis T, Cines DB, Rhee JS, Liang OD, Schubert U, Hammes HP, Higazi AAR, Nawroth PP, Preissner KT, Bdeir K. Regulation of neovascularization by human neutrophil peptides (α‐defensins): a link between inflammation and angiogenesis. FASEB J 2004; 18:1306-8. [PMID: 15208269 DOI: 10.1096/fj.03-1009fje] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Angiogenesis, the growth of new blood vessels, is a complex biological process that is orchestrated by several growth factors and components of the extracellular matrix, including fibronectin (FN) and its receptor the integrin alpha5beta1. Angiogenesis is a critical part of inflammation and wound repair, but the mechanism by which vascular proliferation and migration is regulated by inflammatory cells is not completely understood. We have previously shown that human neutrophil peptides (HNPs), also known as alpha-defensins, which are secreted in high concentrations when neutrophils are activated, bind specifically to FN in the extracellular matrix and inhibit plasminogen activation. Therefore, we asked whether HNPs act as a link between inflammation and angiogenesis. Alpha5beta1-mediated endothelial cell adhesion and migration to FN, both under control conditions and under stimulation by vascular endothelial growth factor (VEGF), were inhibited specifically and in a dose-dependent manner by HNPs, whereas endothelial cell adhesion and migration to other components of the extracellular matrix, such as vitronectin, collagen, or fibrinogen/fibrin were not. Consistent with this finding, HNPs bound to and promoted the binding of fibronectin to alpha5beta1 integrin in arginine-glycine-aspartic acid (RGD)-independent manner. HNPs also completely inhibited VEGF-induced proliferation and induced apoptosis of endothelial cells in a dose-dependent manner. Moreover, HNPs inhibited capillary tube formation in three-dimensional fibrin-matrices as well as neovascularization in vivo in the chicken chorioallantoic membrane assay. Taken together, these data indicate that HNPs can regulate angiogenesis by affecting endothelial cell adhesion and migration in an FN-dependent manner as well as endothelial cell proliferation. These findings provide new insight into the role of inflammatory cells in angiogenesis and might provide a platform for developing a novel class of anti-angiogenesis drugs.
Collapse
|
411
|
Davis FB, Mousa SA, O'Connor L, Mohamed S, Lin HY, Cao HJ, Davis PJ. Proangiogenic action of thyroid hormone is fibroblast growth factor-dependent and is initiated at the cell surface. Circ Res 2004; 94:1500-6. [PMID: 15117822 DOI: 10.1161/01.res.0000130784.90237.4a] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The effects of thyroid hormone analogues on modulation of angiogenesis have been studied in the chick chorioallantoic membrane model. Generation of new blood vessels from existing vessels was increased 3-fold by either l-thyroxine (T4; 10(-7) mol/L) or 3,5,3'-triiodo-l-thyronine (10(-9) mol/L). T4-agarose reproduced the effects of T4, and tetraiodothyroacetic acid (tetrac) inhibited the effects of both T4 and T4-agarose. Tetrac itself was inactive and is known to block actions of T4 on signal transduction that are initiated at the plasma membrane. T4 and basic fibroblast growth factor (FGF2) were comparably effective as inducers of angiogenesis. Low concentrations of FGF2 combined with submaximal concentrations of T4 produced an additive angiogenic response. Anti-FGF2 inhibited the angiogenic effect of T4. The proangiogenic effects of T4 and FGF2 were blocked by PD 98059, a mitogen-activated protein kinase (MAPK) pathway inhibitor. Endothelial cells (ECV304) treated with T4 or FGF2 for 15 minutes demonstrated activation of MAPK, an effect inhibited by PD 98059 and the protein kinase C inhibitor CGP41251. Reverse transcription-polymerase chain reaction of RNA extracted from endothelial cells treated with T4 revealed increased abundance of FGF2 transcript at 6 to 48 hours, and after 72 hours, the medium of treated cells showed increased FGF2 content, an effect inhibited by PD 98059. Thus, thyroid hormone is shown to be a proangiogenic factor. This action, initiated at the plasma membrane, is MAPK dependent and mediated by FGF2.
Collapse
Affiliation(s)
- Faith B Davis
- Department of Veterans Affairs Medical Center, Albany, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
412
|
Trochon-Joseph V, Martel-Renoir D, Mir LM, Thomaïdis A, Opolon P, Connault E, Li H, Grenet C, Fauvel-Lafève F, Soria J, Legrand C, Soria C, Perricaudet M, Lu H. Evidence of antiangiogenic and antimetastatic activities of the recombinant disintegrin domain of metargidin. Cancer Res 2004; 64:2062-9. [PMID: 15026344 DOI: 10.1158/0008-5472.can-03-3272] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metargidin, a transmembrane protein of the adamalysin family, and integrins, e.g., alpha5beta1 and alphav, are preferentially expressed on endothelial cells on angiogenesis. Furthermore, metargidin interacts with these integrins via its disintegrin domain. In this study, recombinant human disintegrin domain (RDD) was produced in Escherichia coli by subcloning its cDNA into the pGEX-2T vector, and the effect of purified RDD on different steps of angiogenesis was evaluated. At concentrations of 2-10 micro g/ml, RDD exhibited inhibitory activities in a variety of in vitro functional assays, including endothelial cell proliferation and adhesion on the integrin substrates fibronectin, vitronectin, and fibrinogen. RDD (10 micro g/ml) totally abrogated endothelial cell migration and blocked most capillary formation in a three-dimensional fibrin gel. To test RDD efficacy in vivo, the RDD gene inserted into pBi vector containing a tetracycline-inducible promoter was electrotransferred into nude mouse muscle. RDD was successfully synthesized by muscle cells in vivo as shown by immunolabeling and Western blotting. In addition, 78% less MDA-MB-231 tumor growth, associated with strong inhibition of tumor angiogenesis, was observed in athymic mice bearing electrotransferred RDD. Moreover, in the presence of RDD, 74% fewer B16F10 melanoma lung metastases were found in C57BL/6 mice. Taken together, these results identified this RDD as a potent intrinsic inhibitor of angiogenesis, tumor growth, and metastasis, making it a promising tool for use in anticancer treatment.
Collapse
MESH Headings
- ADAM Proteins
- Animals
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Cell Adhesion/drug effects
- Cell Division/drug effects
- Cell Movement/drug effects
- Disintegrins/therapeutic use
- Dose-Response Relationship, Drug
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Escherichia coli/genetics
- Female
- Lung Neoplasms/blood supply
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/prevention & control
- Melanoma, Experimental/secondary
- Membrane Proteins/therapeutic use
- Metalloendopeptidases/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Muscle, Skeletal/pathology
- Neovascularization, Pathologic/prevention & control
- Recombinant Proteins/therapeutic use
- Tumor Cells, Cultured
Collapse
|
413
|
Intravenous liposomal delivery of the snake venom disintegrin contortrostatin limits breast cancer progression. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.499.3.4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Despite significant research in this area, metastatic breast cancer remains a disease with a poor prognosis. Until an effective therapy is developed, it is imperative that new treatment modalities be investigated. In this report, we describe an effective method for delivery of a novel snake venom disintegrin, contortrostatin (CN), in an orthotopic, xenograft model of human mammary cancer in immunodeficient mice. CN (Mr 13,500) is a homodimeric disintegrin isolated from venom of the Southern Copperhead snake. The homodimer possesses two Arg-Gly-Asp sites, which modulate its interaction with integrins on tumor cells and angiogenic vascular endothelial cells. Although our laboratory has previously described the antitumor activity of CN in a mouse model of human mammary cancer, the method of delivery, daily intratumor injection, was not translatable to clinical application. We now describe a clinically relevant method of administering CN, liposomal delivery (LCN). A unique liposomal system has been designed for i.v. administration of a biologically active protein with full retention of biological activity. Pharmacokinetics, biodistribution, platelet reactivity, and immunogenicity of LCN were determined and compared with similar characteristics of native, unencapsulated CN. There are several advantages to liposomal delivery of CN: (1) LCN has a significantly prolonged circulatory half-life compared with native CN; (2) LCN is passively accumulated in the tumor; (3) LCN has no platelet reactivity; and (4) LCN is not recognized by the immune system. Finally, antiangiogenic activity is an important component of CN's mechanism of antitumor action. We have demonstrated that i.v. delivery of LCN leads to potent antiangiogenic activity in the orthotopic, xenograft human mammary tumor model.
Collapse
|
414
|
Abstract
The integrin family of cell adhesion proteins promotes the attachment and migration of cells on the surrounding extracellular matrix (ECM). Through signals transduced upon integrin ligation by ECM proteins or immunoglobulin superfamily molecules, this family of proteins plays key roles in regulating tumour growth and metastasis as well as tumour angiogenesis. Several integrins play key roles in promoting tumour angiogenesis and tumour metastasis. Antagonists of several integrins (alpha5beta1, alphavbeta3 and alphavbeta5) are now under evaluation in clinical trials to determine their potential as therapeutics for cancer and other diseases.
Collapse
Affiliation(s)
- H Jin
- John and Rebecca Moores Comprehensive Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA
| | - J Varner
- John and Rebecca Moores Comprehensive Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA
- John and Rebecca Moores Comprehensive Cancer Center, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0912, USA. E-mail:
| |
Collapse
|
415
|
Bouissou C, Potter U, Altroff H, Mardon H, Van Der Walle C. Controlled release of the fibronectin central cell binding domain from polymeric microspheres. J Control Release 2004; 95:557-66. [PMID: 15023466 DOI: 10.1016/j.jconrel.2003.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2003] [Accepted: 12/23/2003] [Indexed: 11/28/2022]
Abstract
Non-ionic surfactants have been employed as alternatives to PVA for the emulsification-encapsulation of a conformationally labile protein (FIII9'-10) into PLGA microspheres. FIII9'-10 was encapsulated using a w/o/w double emulsification-evaporation technique and the microspheres fabricated were characterized by SEM and CLSM. The peptide backbone integrity of FIII9'-10 was assayed by SDS-PAGE and the degree of unfolding of FIII9'-10 following emulsification-encapsulation was assessed using a fibroblast cell-attachment assay. The encapsulation efficiency for FIII9'-10 was 25% when using PVA, compared to 50-60% when using Igepal CA-630 or Triton-X100, with values below for the other surfactants. FIII9'-10 released from microspheres promoted cell attachment in a concentration-dependent manner, only Igepal CA-630 and Triton X-100 maintaining near-maximal cell attachment, indicating that the conformation of the relatively unstable FIII9' domain was preserved. All non-ionic surfactants reduced microsphere surface porosity, compared to PVA, and an increasing surface rugosity (leading to minor 'ridges') could be correlated with decreasing surfactant HLB. Low surface porosities did not effect the diffusion of FIII9'-10 from the microspheres' internal pores in a 'burst release', as may have been imagined. In summary, non-ionic surfactants should be considered over PVA for the maintenance of biological activity of conformationally labile proteins during encapsulation.
Collapse
Affiliation(s)
- Camille Bouissou
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | | | | | | | |
Collapse
|
416
|
Abstract
A variety of factors cooperate to regulate neovessel formation and persistence. Proangiogenic growth factors have remained an area of intense interest due to their capacity to promote endothelial cell (EC) proliferation and to initiate the angiogenic program. These growth factors are associated with increased cell survival, yet paradoxically, angiogenic ECs are more susceptible to apoptosis than quiescent ECs. Survival is regulated by cooperation between growth factor receptors and integrins, which are in turn governed by the composition of the local extracellular matrix (ECM). Integrin-mediated signaling is altered or disrupted by the presence of soluble, rather than matrix-bound ligands, thus providing a means by which ECM remodeling can influence both integrin- and growth factor-mediated events. Ultimately, the collaboration of these factors determines whether ECs survive or die, thereby regulating neovascularization.
Collapse
Affiliation(s)
- Dwayne G Stupack
- Department of Immunology, The Scripps Research Institute, 10550 N Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
417
|
Koth L, Sheppard D. Integrins and Pulmonary Fibrosis. LUNG BIOLOGY IN HEALTH AND DISEASE 2003. [DOI: 10.1201/b14211-15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
418
|
Ritter MR, Moreno SK, Dorrell MI, Rubens J, Ney J, Friedlander DF, Bergman J, Cunningham BB, Eichenfield L, Reinisch J, Cohen S, Veccione T, Holmes R, Friedlander SF, Friedlander M. Identifying Potential Regulators of Infantile Hemangioma Progression through Large-scale Expression Analysis: A Possible Role for the Immune System and Indoleamine 2,3 Dioxygenase (IDO) during Involution. Lymphat Res Biol 2003; 1:291-9. [PMID: 15624557 DOI: 10.1089/153968503322758094] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hemangiomas are benign endothelial tumors. Often referred to as hemangiomas of infancy (HOI), these tumors are the most common tumor of infancy. Most of these lesions proliferate rapidly in the first months of life, and subsequently slowly involute during early childhood without significant complications. However, they often develop on the head or neck, and may pose a significant cosmetic concern for families. In addition, a fraction of these tumors can grow explosively and ulcerate, bleed, or obstruct vision or airway structures. Current treatments for these tumors are associated with significant side effects, and our knowledge of the biology of hemangiomas is limited. The natural evolution of these lesions creates a unique opportunity to study the changes in gene expression that occur as the endothelium of these tumors proliferates and then subsequently regresses. Such information may also increase our understanding of the basic principals of angiogenesis in normal and abnormal tissue. We have performed large-scale genomic analysis of hemangioma gene expression using DNA microarrays. We recently identified insulin-like growth factor 2 as a potentially important regulator of hemangioma growth using this approach. However, little is known about the mechanisms involved in hemangioma involution. Here we explore the idea that hemangioma involution might be an immune-mediated process and present data to support this concept. We also demonstrate that proliferating hemangiomas express indoleamine 2,3 dioxygenase (IDO) and discuss a possible mechanism that accounts for the often slow regression of these lesions.
Collapse
Affiliation(s)
- Matthew R Ritter
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
419
|
Boudreau NJ, Varner JA. The homeobox transcription factor Hox D3 promotes integrin alpha5beta1 expression and function during angiogenesis. J Biol Chem 2003; 279:4862-8. [PMID: 14610084 DOI: 10.1074/jbc.m305190200] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neovascularization promotes wound healing, tumor growth, and arthritis. Endothelial cell migration and survival during neovascularization are regulated by adhesion proteins, including integrin alpha5beta1. Integrin alpha5beta1 is poorly expressed on normal quiescent blood vessels, but its expression is induced on tumor blood vessels and in response to angiogenic factors such as basic fibroblast growth factor, interleukin-8, tumor necrosis factor-alpha, and the angiomatrix protein Del-1. We show here that alpha5beta1 expression, and hence function, during angiogenesis is regulated by the transcription factor Hox D3, a homeobox gene that also controls the expression of endothelial cell integrin alphavbeta3 and urokinase-type plasminogen activator. Hox D3 expression in endothelial cells enhances integrin alpha5 protein and message expression, whereas Hox D3 antisense inhibits its expression. Hox D3 promotes alpha5 expression during angiogenesis in vivo, whereas inhibition of alpha5 expression by Hox D3 antisense suppresses angiogenesis. Hox D3 binds directly to the promoters of the integrin alpha5 and beta3 subunits, inducing subunit expression. As Hox D3, integrin alphavbeta3, and integrin alpha5beta1 are expressed on tumor blood vessels but not on normal quiescent vessels, these studies suggest that Hox D3 coordinately regulates the expression of integrin alpha5beta1 and integrin alphavbeta3 during angiogenesis in vivo. These studies also suggest that Hox D3 inhibition could be a useful approach to inhibit tumor angiogenesis.
Collapse
Affiliation(s)
- Nancy J Boudreau
- Department of Surgery, University of California, San Francisco, California 94143, USA
| | | |
Collapse
|
420
|
Baluk P, Morikawa S, Haskell A, Mancuso M, McDonald DM. Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1801-15. [PMID: 14578181 PMCID: PMC1892429 DOI: 10.1016/s0002-9440(10)63540-7] [Citation(s) in RCA: 375] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/16/2003] [Indexed: 01/17/2023]
Abstract
Often described as incomplete or absent, the basement membrane of blood vessels in tumors has attracted renewed attention as a source of angiogenic and anti-angiogenic molecules, site of growth factor binding, participant in angiogenesis, and potential target in cancer therapy. This study evaluated the composition, extent, and structural integrity of the basement membrane on blood vessels in three mouse tumor models: spontaneous RIP-Tag2 pancreatic islet tumors, MCa-IV mammary carcinomas, and Lewis lung carcinomas. Tumor vessels were identified by immunohistochemical staining for the endothelial cell markers CD31, endoglin (CD105), vascular endothelial growth factor receptor-2, and integrin alpha5 (CD49e). Confocal microscopic studies revealed that basement membrane identified by type IV collagen immunoreactivity covered >99.9% of the surface of blood vessels in the three tumors, just as in normal pancreatic islets. Laminin, entactin/nidogen, and fibronectin immunoreactivities were similarly ubiquitous on tumor vessels. Holes in the basement membrane, found by analyzing 1- micro m confocal optical sections, were <2.5 micro m in diameter and involved only 0.03% of the vessel surface. Despite the extensive vessel coverage, the basement membrane had conspicuous structural abnormalities, including a loose association with endothelial cells and pericytes, broad extensions away from the vessel wall, and multiple layers visible by electron microscopy. Type IV collagen-immunoreactive sleeves were also present on endothelial sprouts, supporting the idea that basement membrane is present where sprouts grow and regress. These findings indicate that basement membrane covers most tumor vessels but has profound structural abnormalities, consistent with the dynamic nature of endothelial cells and pericytes in tumors.
Collapse
Affiliation(s)
- Peter Baluk
- Cardiovascular Research Institute, Comprehensive Cancer Center, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0452, USA
| | | | | | | | | |
Collapse
|
421
|
Hassanieh L, Rodriguez D, Xu J, Brooks PC, Broek D. Generation of a Monoclonal Antibody to a Cryptic Site Common to Both Integrin β1 as Well as Gelatinase MMP9. ACTA ACUST UNITED AC 2003; 22:285-92. [PMID: 14678645 DOI: 10.1089/153685903322538809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Integrins are one class of cell surface receptors that have been implicated in the regulation of a diverse set of cellular processes, including cell adhesion, migration, and invasion as well as gene expression, differentiation, and signal transduction. Cellular invasion not only requires the adhesive properties of integrins but also the proteolytic properties of matrix-degrading enzymes, such as the metalloproteinases (MMPs). Previous studies have shown that integrin alphavbeta3 is a receptor for MMP2, localizing its proteinase activity to the cell surface, ultimately leading to site-specific extracellular matrix (ECM) degradation. Here we develop reagents to investigate the possibility of an interplay between MMP9 and integrin alpha5beta1. With the use of EV22 viral studies, the tetrapeptide sequence, LRSG, was shown to be a dimerizing sequence mediating beta1 integrin binding to EV22. The same study also showed that cellular infection could be halted with the use of LRSG-containing peptides. In a later study, in an effort to isolate inhibitors of the MMP family, LRSG sequence was identified as one capable of binding MMP9. Interestingly, MMP9 contains an LRSG sequence, raising the possibility that MMP9 binds the cell surface via beta1 integrins through the dimerizing LRSG motif. We used the LRSG-containing sequence from beta1 integrins as an antigen to generate the monoclonal antibody (MAB) FM155 in the mouse model. MAB FM155 will help identify a cryptic epitope, LRSG, and its role in matrix remodeling as well as tumor growth, cancer cell migration, and angiogenesis.
Collapse
Affiliation(s)
- Loubna Hassanieh
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Center, Keck School of Medicine at the University of Southern California, Los Angeles, California 90089, USA
| | | | | | | | | |
Collapse
|
422
|
Yi M, Sakai T, Fassler R, Ruoslahti E. Antiangiogenic proteins require plasma fibronectin or vitronectin for in vivo activity. Proc Natl Acad Sci U S A 2003; 100:11435-11438. [PMID: 13679585 PMCID: PMC208775 DOI: 10.1073/pnas.1635112100] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fragmentation of various extracellular matrix and blood proteins generates antiangiogenic substances that are physiological regulators of angiogenesis. Some of these compounds are in clinical trials as inhibitors of tumor angiogenesis. Anastellin, an antiangiogenic protein fragment derived from fibronectin, was unable to inhibit matrigel plug angiogenesis in mice that lack plasma fibronectin. Anastellin was fully active in mice that are null for vitronectin, which, like fibronectin, is a major adhesion protein in the blood. An antiangiogenic form of antithrombin showed the opposite pattern. The activity of endostatin was impaired in both fibronectin- and vitronectin-deficient mice. These results suggest a shared mechanism of action for antiangiogenic factors derived from extracellular matrix and plasma proteins: these factors form complexes with adhesion proteins in plasma to create an active antiangiogenic substance.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Research Center, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
423
|
Hiran TS, Mazurkiewicz JE, Kreienberg P, Rice FL, LaFlamme SE. Endothelial expression of the alpha6beta4 integrin is negatively regulated during angiogenesis. J Cell Sci 2003; 116:3771-81. [PMID: 12902402 DOI: 10.1242/jcs.00681] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Development and homeostasis of the vascular system requires integrin-facilitated cellular adhesion, migration, proliferation and survival. A specific role for the alpha6beta4 integrin in the vasculature, however, has not been identified. Using immunohistochemistry, we observed alpha6beta4 expression on the dermal microvasculature of human foreskin. Analysis of individual cells isolated from trypsin-disrupted foreskin tissue indicated that alpha6beta4 was expressed by a subset of epithelial and endothelial cells, and not by smooth muscle cells. Expression of alpha6beta4 was also analyzed during new vessel growth using explants of human saphenous vein cultured in fibrinogen gels. The results indicate that alpha6beta4 is not expressed by outgrowing endothelial cells, and is downregulated by the original alpha6beta4-positive endothelial cells of the explant. To determine whether alpha6beta4 is expressed during angiogenesis in vivo, the expression of the beta4 subunit was analyzed during the development of the mouse mystacial (whisker) pad. Immunohistochemical staining of the whisker pad indicates that beta4 is expressed by the adult vasculature. To identify when and where beta4 is turned on in the vasculature, we examined the whisker pads from the developing embryo (E19.5 pc), and from postnatal days zero (P0), three (P3) and seven (P7) pups. The expression of alpha6beta4 was found to be turned on spatially and temporally from caudal to rostral regions and from the deep to superficial vasculature, correlating with the maturation of the whisker pad and its corresponding vasculature. Together, these findings suggest a potential role for alpha6beta4 as a negative component of the angiogenic switch, whereas expression of alpha6beta4 on the adult vasculature may indicate regions requiring additional adhesive mechanisms.
Collapse
Affiliation(s)
- Tejindervir S Hiran
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | |
Collapse
|
424
|
Hood JD, Cheresh DA. Targeted delivery of mutant Raf kinase to neovessels causes tumor regression. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:285-91. [PMID: 12858551 DOI: 10.1101/sqb.2002.67.285] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- J D Hood
- Scripps Research Institute, Department of Immunology, La Jolla, California 92037, USA
| | | |
Collapse
|
425
|
Hynes RO, Lively JC, McCarty JH, Taverna D, Francis SE, Hodivala-Dilke K, Xiao Q. The diverse roles of integrins and their ligands in angiogenesis. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:143-53. [PMID: 12858535 DOI: 10.1101/sqb.2002.67.143] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- R O Hynes
- Howard Hughes Medical Institute, Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | |
Collapse
|
426
|
Orecchia A, Lacal PM, Schietroma C, Morea V, Zambruno G, Failla CM. Vascular endothelial growth factor receptor-1 is deposited in the extracellular matrix by endothelial cells and is a ligand for the alpha 5 beta 1 integrin. J Cell Sci 2003; 116:3479-89. [PMID: 12865438 DOI: 10.1242/jcs.00673] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Vascular endothelial growth factor receptor-1 (VEGFR-1) is a tyrosine kinase receptor for several growth factors of the VEGF family. Endothelial cells express a membrane-spanning form of VEGFR-1 and secrete a soluble variant of the receptor comprising only the extracellular region. The role of this variant has not yet been completely defined. In this study, we report that the secreted VEGFR-1 is present within the extracellular matrix deposited by endothelial cells in culture, suggesting a possible involvement in endothelial cell adhesion and migration. In adhesion assays, VEGFR-1 extracellular region specifically promoted endothelial cell attachment. VEGFR-1-mediated cell adhesion was divalent cation-dependent, and inhibited by antibodies directed against the alpha 5 beta 1 integrin. Moreover, VEGFR-1 promoted endothelial cell migration, and this effect was inhibited by anti-alpha 5 beta 1 antibodies. Direct binding of VEGFR-1 to the alpha 5 beta 1 integrin was also detected. Finally, binding to VEGFR-1 initiated endothelial cell spreading. Altogether these results indicate that the soluble VEGFR-1 secreted by endothelial cells becomes a matrix-associated protein that is able to interact with the alpha 5 beta 1 integrin, suggesting a new role of VEGFR-1 in angiogenesis, in addition to growth factor binding.
Collapse
Affiliation(s)
- Angela Orecchia
- Molecular and Cell Biology Laboratory, IDI-IRCCS, via Monti di Creta 104, 00167 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
427
|
Wu J, Sheibani N. Modulation of VE-cadherin and PECAM-1 mediated cell-cell adhesions by mitogen-activated protein kinases. J Cell Biochem 2003; 90:121-37. [PMID: 12938162 DOI: 10.1002/jcb.10600] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Endothelial cell transition from a differentiated, quiescent phenotype to a migratory, proliferative phenotype is essential during angiogenesis. This transition is dependent on alterations in the balanced production of stimulatory and inhibitory factors, which normally keep angiogenesis in check. Activation of MAPK/ERKs is essential for endothelial cell migration and proliferation. However, its role in regulation of endothelial cell adhesive mechanisms requires further delineation. Here, we show that sustained activation of MAPK/ERKs results in disruption of cadherin-mediated cell-cell adhesion, down-regulation of PECAM-1 expression, and enhanced cell migration in microvascular endothelial cells. Expression of a constitutively active MEK-1 in mouse brain endothelial (bEND) cells resulted in down-regulation of VE-cadherin and catenins expression concomitant with down-regulation of PECAM-1 expression. In contrast, inhibition of MEK-1 restored parental morphology, cadherin/catenins expression and localization. These data are further supported by our observation that sustained activation of MAPK/ERKs in phorbol myristate acetate incubated HUVEC lead to disruption of cadherin-mediate cell-cell interactions and enhanced capillary formation on Matrigel. Thus, sustained activation of MAPK/ERKs plays an important role in disruption of cell-cell adhesion and migration of endothelial cells.
Collapse
Affiliation(s)
- Jianqiang Wu
- Department of Ophthalmology, University of Wisconsin Medical School, Madison, Wisconsin 53792, USA
| | | |
Collapse
|
428
|
Horiuchi K, Weskamp G, Lum L, Hammes HP, Cai H, Brodie TA, Ludwig T, Chiusaroli R, Baron R, Preissner KT, Manova K, Blobel CP. Potential role for ADAM15 in pathological neovascularization in mice. Mol Cell Biol 2003; 23:5614-24. [PMID: 12897135 PMCID: PMC166329 DOI: 10.1128/mcb.23.16.5614-5624.2003] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ADAM15 (named for a disintegrin and metalloprotease 15, metargidin) is a membrane-anchored glycoprotein that has been implicated in cell-cell or cell-matrix interactions and in the proteolysis of molecules on the cell surface or extracellular matrix. To characterize the potential roles of ADAM15 during development and in adult mice, we analyzed its expression pattern by mRNA in situ hybridization and generated mice carrying a targeted deletion of ADAM15 (adam15(-/-) mice). A high level of expression of ADAM15 was found in vascular cells, the endocardium, hypertrophic cells in developing bone, and specific areas of the hippocampus and cerebellum. However, despite the pronounced expression of ADAM15 in these tissues, no major developmental defects or pathological phenotypes were evident in adam15(-/-) mice. The elevated levels of ADAM15 in endothelial cells prompted an evaluation of its role in neovascularization. In a mouse model for retinopathy of prematurity, adam15(-/-) mice had a major reduction in neovascularization compared to wild-type controls. Furthermore, the size of tumors resulting from implanted B16F0 mouse melanoma cells was significantly smaller in adam15(-/-) mice than in wild-type controls. Since ADAM15 does not appear to be required for developmental angiogenesis or for adult homeostasis, it may represent a novel target for the design of inhibitors of pathological neovascularization.
Collapse
MESH Headings
- ADAM Proteins
- Alleles
- Animals
- Blotting, Western
- Brain/metabolism
- Cell Membrane/metabolism
- Cells, Cultured
- DNA, Complementary/metabolism
- Endothelium, Vascular/cytology
- Flow Cytometry
- Gene Deletion
- Humans
- Immunohistochemistry
- In Situ Hybridization
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Metalloendopeptidases/genetics
- Metalloendopeptidases/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Models, Genetic
- Neovascularization, Pathologic
- Phenotype
- Protein Structure, Tertiary
- RNA, Messenger/metabolism
- Retinal Diseases/pathology
- Time Factors
- Tissue Distribution
- Tumor Cells, Cultured
- Umbilical Veins/cytology
Collapse
Affiliation(s)
- Keisuke Horiuchi
- Cellular Biochemistry and Biophysics Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
429
|
Gratzinger D, Canosa S, Engelhardt B, Madri JA. Platelet endothelial cell adhesion molecule-1 modulates endothelial cell motility through the small G-protein Rho. FASEB J 2003; 17:1458-69. [PMID: 12890700 DOI: 10.1096/fj.02-1040com] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Platelet endothelial cell adhesion molecule-1 (PECAM-1), an immunoglobulin family vascular adhesion molecule, is involved in endothelial cell migration and angiogenesis (1, 2). We found that endothelial cells lacking PECAM-1 exhibit increased single cell motility and extension formation but poor wound healing migration, reminiscent of cells in which Rho activity has been suppressed by overexpressing a GTPase-activating protein (3). The ability of PECAM-1 to restore wound healing migration to PECAM-1-deficient cells was independent of its extracellular domain or signaling via its immunoreceptor tyrosine-based inhibitory motif. PECAM-1-deficient endothelial cells had a selective defect in RhoGTP loading, and inhibition of Rho activity mimicked the PECAM-1-deficient phenotype of increased chemokinetic single cell motility at the expense of coordinated wound healing migration. The wound healing advantage of PECAM-1-positive endothelial cells was not only Rho mediated but pertussis toxin inhibitable, characteristic of migration mediated by heterotrimeric G-protein-linked seven-transmembrane receptor signaling such as signaling in response to the serum sphingolipid sphingosine-1-phosphate (S1P) (4, 5). Indeed, we found that the wound healing defect of PECAM-1 null endothelial cells is minimized in sphingolipid-depleted media; moreover, PECAM-1 null endothelial cells fail to increase their migration in response to S1P. We have also found that PECAM-1 localizes to rafts and that in its absence heterotrimeric G-protein components are differentially recruited to rafts, providing a potential mechanism for PECAM-1-mediated coordination of S1P signaling. PECAM-1 may thus support the effective S1P/RhoGTP signaling required for wound healing endothelial migration by allowing for the spatially directed, coordinated activation of Galpha signaling pathways.
Collapse
Affiliation(s)
- Dita Gratzinger
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | |
Collapse
|
430
|
Serini G, Valdembri D, Zanivan S, Morterra G, Burkhardt C, Caccavari F, Zammataro L, Primo L, Tamagnone L, Logan M, Tessier-Lavigne M, Taniguchi M, Püschel AW, Bussolino F. Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature 2003; 424:391-7. [PMID: 12879061 DOI: 10.1038/nature01784] [Citation(s) in RCA: 446] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2003] [Accepted: 05/28/2003] [Indexed: 12/15/2022]
Abstract
The motility and morphogenesis of endothelial cells is controlled by spatio-temporally regulated activation of integrin adhesion receptors, and integrin activation is stimulated by major determinants of vascular remodelling. In order for endothelial cells to be responsive to changes in activator gradients, the adhesiveness of these cells to the extracellular matrix must be dynamic, and negative regulators of integrins could be required. Here we show that during vascular development and experimental angiogenesis, endothelial cells generate autocrine chemorepulsive signals of class 3 semaphorins (SEMA3 proteins) that localize at nascent adhesive sites in spreading endothelial cells. Disrupting endogenous SEMA3 function in endothelial cells stimulates integrin-mediated adhesion and migration to extracellular matrices, whereas exogenous SEMA3 proteins antagonize integrin activation. Misexpression of dominant negative SEMA3 receptors in chick embryo endothelial cells locks integrins in an active conformation, and severely impairs vascular remodelling. Sema3a null mice show vascular defects as well. Thus during angiogenesis endothelial SEMA3 proteins endow the vascular system with the plasticity required for its reshaping by controlling integrin function.
Collapse
Affiliation(s)
- Guido Serini
- Division of Molecular Angiogenesis, IRCC, Institute for Cancer Research and Treatment, and Department of Oncological Sciences, University of Torino School of Medicine, 10060 Candiolo, TO, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
431
|
Zhong J, Eliceiri B, Stupack D, Penta K, Sakamoto G, Quertermous T, Coleman M, Boudreau N, Varner JA. Neovascularization of ischemic tissues by gene delivery of the extracellular matrix protein Del-1. J Clin Invest 2003; 112:30-41. [PMID: 12840057 PMCID: PMC162283 DOI: 10.1172/jci17034] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The ECM protein Del-1 is one of several novel ECM proteins that accumulate around angiogenic blood vessels in embryonic and tumor tissue and promote angiogenesis in the absence of exogenous growth factors. Del-1 expressed in mouse or rabbit ischemic hind-limb muscle by gene transfer rapidly promotes new blood vessel formation and restores muscle function. This angiogenic ECM protein initiates angiogenesis by binding to integrin alphavbeta5 on resting endothelium, thereby resulting in expression of the transcription factor Hox D3 and integrin alphavbeta3. Hox D3 converts resting endothelium to angiogenic endothelium by inducing expression of proangiogenic molecules such as integrin alphavbeta3. These findings provide evidence for an angiogenic switch that can be initiated in the absence of exogenous growth factors and indicate that the angiogenic matrix protein Del-1 may be a useful tool for the therapy of ischemic disease.
Collapse
Affiliation(s)
- Jingping Zhong
- Comprehensive Cancer Center, University of California, San Diego, La Jolla, California 92093-0912, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
432
|
Abstract
Advances in imaging are transforming our understanding of angiogenesis and the evaluation of drugs that stimulate or inhibit angiogenesis in preclinical models and human disease. Vascular imaging makes it possible to quantify the number and spacing of blood vessels, measure blood flow and vascular permeability, and analyze cellular and molecular abnormalities in blood vessel walls. Microscopic methods ranging from fluorescence, confocal and multiphoton microscopy to electron microscopic imaging are particularly useful for elucidating structural and functional abnormalities of angiogenic blood vessels. Magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), ultrasonography and optical imaging provide noninvasive, functionally relevant images of angiogenesis in animals and humans. An ongoing dilemma is, however, that microscopic methods provide their highest resolution on preserved tissue specimens, whereas clinical methods give images of living tissues deep within the body but at much lower resolution and specificity and generally cannot resolve vessels of the microcirculation. Future challenges include developing new imaging methods that can bridge this resolution gap and specifically identify angiogenic vessels. Another goal is to determine which microscopic techniques are the best benchmarks for interpreting clinical images. The importance of angiogenesis in cancer, chronic inflammatory diseases, age-related macular degeneration and reversal of ischemic heart and limb disease provides incentive for meeting these challenges.
Collapse
Affiliation(s)
- Donald M McDonald
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA.
| | | |
Collapse
|
433
|
Perruzzi CA, de Fougerolles AR, Koteliansky VE, Whelan MC, Westlin WF, Senger DR. Functional overlap and cooperativity among alphav and beta1 integrin subfamilies during skin angiogenesis. J Invest Dermatol 2003; 120:1100-9. [PMID: 12787141 DOI: 10.1046/j.1523-1747.2003.12236.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Angiogenesis requires endothelial cell survival and proliferation, which depend upon cytokine stimulation together with integrin-mediated cell adhesion to extracellular matrix; however, the question of which specific integrins are the best targets for suppressing neovascularization is controversial and unresolved. Therefore, we designed experiments to compare contributions of individual integrins from both the alphav and beta1 integrin subfamilies. With immobilized antibodies, we determined that adhesion through integrins alpha1beta1, alpha2beta1, alphavbeta3, and alphavbeta5 each individually supported dermal microvascular endothelial cell survival. Also, substratum coated with collagen I (which binds alpha1beta1 and alpha2beta1) and vitronectin (which binds alphavbeta3 and alphavbeta5) each supported survival. Importantly, substratum coated with combinations of collagen I and vitronectin were most effective at promoting survival, and survival on three-dimensional collagen I gels was strongly enhanced by vitronectin. Vascular endothelial growth factor activation of the p44/p42 mitogen-activated protein kinase pathway, which is required for angiogenesis, was supported by adhesion through either alpha1beta1, alpha2beta1, alphavbeta3, or alphavbeta5, and pharmacologic inhibition of this pathway blocked proliferation and suppressed survival. Therefore, these studies establish that the alpha1beta1, alpha2beta1, alphavbeta3, and alphavbeta5 integrins each support dermal microvascular endothelial cell viability, and that each collaborate with vascular endothelial growth factor to support robust activation of the mitogen-activated protein kinase pathway which mediates both proliferation and survival. Moreover, survival is supported most significantly by extracellular matrices, which engage all of these integrins in combination. Consistent with important complementary and overlapping functions, combined antagonism of these integrins provided superior inhibition of angiogenesis in skin, indicating that multiplicity of integrin involvement should be considered in designing strategies for controlling neovascularization.
Collapse
Affiliation(s)
- Carole A Perruzzi
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
434
|
Abstract
Proteolytic fragments of various components of the extracellular matrix exhibit antiangiogenic activity via interaction with endothelial cell surface integrins. Kalluri and coworkers (this issue of Cancer Cell) use gene-targeted mice to show a physiological role for a carboxy-terminal fragment of collagen IV in the regulation of tumor angiogenesis.
Collapse
|
435
|
Hotchkiss KA, Ashton AW, Schwartz EL. Thymidine phosphorylase and 2-deoxyribose stimulate human endothelial cell migration by specific activation of the integrins alpha 5 beta 1 and alpha V beta 3. J Biol Chem 2003; 278:19272-9. [PMID: 12639965 DOI: 10.1074/jbc.m212670200] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Thymidine phosphorylase is an angiogenic factor that is frequently overexpressed in solid tumors, in rheumatoid arthritis, and in response to inflammatory cytokines. Our previous studies showed that cells expressing thymidine phosphorylase stimulated endothelial cell migration in vitro. This was a consequence of the intracellular metabolism of thymidine by thymidine phosphorylase and subsequent extracellular release of 2-deoxyribose. The mechanisms by which 2-deoxyribose might mediate thymidine phosphorylase-induced cell migration in vitro, however, are obscure. Here we show that both thymidine phosphorylase and 2-deoxyribose stimulated the formation of focal adhesions and the tyrosine 397 phosphorylation of focal adhesion kinase in human umbilical vein endothelial cells. Although similar actions occurred upon treatment with the angiogenic factor vascular endothelial growth factor (VEGF), thymidine phosphorylase differed from VEGF in that its effect on endothelial cell migration was blocked by antibodies to either integrin alpha 5 beta 1 or alpha v beta 3, whereas VEGF-induced endothelial cell migration was only blocked by the alpha v beta 3 antibody. Further, thymidine phosphorylase and 2-deoxyribose, but not VEGF, increased the association of both focal adhesion kinase and the focal adhesion-associated protein vinculin with integrin alpha 5 beta 1 and, in intact cells, increased the co-localization of focal adhesion kinase with alpha 5 beta 1. Thymidine phosphorylase and 2-deoxyribose-induced focal adhesion kinase phosphorylation was blocked by the antibodies to alpha 5 beta 1 and alpha v beta 3, directly linking the migration and signaling components of thymidine phosphorylase and 2-deoxyribose action. Cell surface expression of alpha 5 beta 1 was also increased by thymidine phosphorylase and 2-deoxyribose. These experiments are the first to demonstrate a direct effect of thymidine phosphorylase and 2-deoxyribose on signaling pathways associated with endothelial cell migration.
Collapse
Affiliation(s)
- Kylie A Hotchkiss
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York 10467, USA
| | | | | |
Collapse
|
436
|
Bourguet E, Banères JL, Parello J, Lusinchi X, Girard JP, Vidal JP. Nonpeptide RGD antagonists: a novel class of mimetics, the 5,8-disubstituted 1-azabicyclo[5.2.0]nonan-2-one lactam. Bioorg Med Chem Lett 2003; 13:1561-4. [PMID: 12699755 DOI: 10.1016/s0960-894x(03)00181-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The 1-azabicyclo[5.2.0]nonan-2-one lactam 1 adequately substituted on both cycles A and B as scaffolds mimics the conformationally constrained beta-turn of the tripeptide RGD signaling motif of fibronectin. Using an in vitro assay, we establish that trans diastereoisomer 1b dissociates a soluble fibronectin-integrin alpha(5)beta(1) complex at concentrations comparable to those of a linear RGDS peptide as a competitor.
Collapse
Affiliation(s)
- Erika Bourguet
- Laboratoire de Chimie Biomoléculaire et Interactions Biologiques, Unité Mixte de Recherche CNRS 5074, Université Montpellier I, Faculté de Pharmacie, 15 Av. C. Flahault, BP 14491, 34093 Montpellier Cedex 5, France
| | | | | | | | | | | |
Collapse
|
437
|
Stoeltzing O, Liu W, Reinmuth N, Fan F, Parry GC, Parikh AA, McCarty MF, Bucana CD, Mazar AP, Ellis LM. Inhibition of integrin alpha5beta1 function with a small peptide (ATN-161) plus continuous 5-FU infusion reduces colorectal liver metastases and improves survival in mice. Int J Cancer 2003; 104:496-503. [PMID: 12584749 DOI: 10.1002/ijc.10958] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Integrin alpha(5)beta(1) is expressed on activated endothelial cells and plays a critical role in tumor angiogenesis. We hypothesized that a novel integrin alpha(5)beta(1) antagonist, ATN-161, would inhibit angiogenesis and growth of liver metastases in a murine model. We further hypothesized that combining ATN-161 with 5-fluorouracil (5-FU) chemotherapy would enhance the antineoplastic effect. Murine colon cancer cells (CT26) were injected into spleens of BALB/c mice to produce liver metastases. Four days thereafter, mice were given either ATN-161 (100 mg/kg, every 3rd day) or saline by intraperitoneal injection, with or without combination of continuous-infusion 5-FU (100 mg/kg/2 weeks), which was started on day 7. On day 20 after tumor cell inoculation, mice were killed and liver weights and number of liver metastases were determined. A follow-up study on survival was also conducted in which mice were randomized to receive ATN-161, 5-FU or ATN-161+5-FU. Combination therapy with ATN-161+5-FU significantly reduced tumor burden (liver weight) and number of liver metastases (p<0.02). Liver tumors in the ATN-161 and ATN-161+5-FU groups had significantly fewer microvessels (p<0.05) than tumors in the control or 5-FU-treated groups. Unlike treatment with either agent alone, ATN-161+5-FU significantly increased tumor cell apoptosis and decreased tumor cell proliferation (p<0.03) and improved overall survival (p<0.03, log-rank test). Targeting integrin alpha(5)beta(1) in combination with 5-FU infusion reduced liver metastases formation and improved survival in this colon cancer model. The enhancement of antineoplastic activity from the combination of anti-angiogenic therapy and chemotherapy may be a promising approach for treating metastatic colorectal cancer.
Collapse
Affiliation(s)
- Oliver Stoeltzing
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston 77030-4009, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
438
|
Rüegg C, Dormond O, Foletti A. Suppression of tumor angiogenesis through the inhibition of integrin function and signaling in endothelial cells: which side to target? ENDOTHELIUM : JOURNAL OF ENDOTHELIAL CELL RESEARCH 2003; 9:151-60. [PMID: 12380640 DOI: 10.1080/10623320213635] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor angiogenesis is an essential step in tumor progression and metastasis formation. Suppression of tumor angiogenesis results in the inhibition of tumor growth. Recent evidence indicates that vascular integrins, in particular alpha V beta 3, are important regulators of angiogenesis, including tumor angiogenesis. Integrin alpha V beta 3 antagonists, such as blocking antibodies or peptides, suppress tumor angiogenesis and tumor progression in many preclinical tumor models. The potential therapeutic efficacy of extracellular integrin antagonists in human cancer is currently being tested in clinical trials. Selective disruption of the tumor vasculature by high doses of tumor necrosis factor (TNF) and interferon gamma (IFN-gamma), and the antiangiogenic activity of nonsteroidal anti-inflammatory drugs are associated with the suppression of integrin alpha V beta 3 function and signaling in endothelial cells. Furthermore, expression of isolated integrin cytoplasmic domains disrupts integrin-dependent adhesion, resulting in endothelial cell detachment and apoptosis. These results confirm the critical role of vascular integrins in promoting endothelial cell survival and angiogenesis and suggest that intracellular targeting of integrin function and signaling may be an alternative strategy to extracellular integrin antagonists for the therapeutic inhibition of tumor angiogenesis.
Collapse
Affiliation(s)
- C Rüegg
- Centre Pluridisciplinaire d'Oncologie, University of Lausanne Medical School, Switzerland.
| | | | | |
Collapse
|
439
|
Alahari SK, Reddig PJ, Juliano RL. Biological aspects of signal transduction by cell adhesion receptors. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 220:145-84. [PMID: 12224548 DOI: 10.1016/s0074-7696(02)20005-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell adhesion receptors such as integrins, cadherins, selectins, and immunoglobulin family receptors profoundly modulate many signal transduction cascades. In this review we examine aspects of adhesion receptor signaling and how this impinges on key biological processes. We have chosen to focus on cell migration and on programmed cell death. We examine many of the cytoplasmic signaling molecules that interface with adhesion receptors, including focal adhesion kinase (FAK), phosphatidylinositol-3-kinase (PI3K), and elements of the Erk/MAP kinase pathway. In many cases these molecules impinge on both the regulation of cell movement and on control of apoptosis.
Collapse
Affiliation(s)
- Suresh K Alahari
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill 27599, USA
| | | | | |
Collapse
|
440
|
Affiliation(s)
- Roy R Lobb
- Biogen, Inc., Cambridge, Massachusetts 02142, USA
| |
Collapse
|
441
|
Bubenik M, Meerovitch K, Bergeron F, Attardo G, Chan L. Thiophene-based vitronectin receptor antagonists. Bioorg Med Chem Lett 2003; 13:503-6. [PMID: 12565960 DOI: 10.1016/s0960-894x(02)00942-3] [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: 11/19/2022]
Abstract
A series of alpha(v)beta(3) antagonists based on a thiophene scaffold were synthesized via two routes and evaluated for in vitro biological activity. We have identified several structurally similar antagonists with different selectivities towards alpha(IIb)beta(3), alpha(v)beta(5) and alpha(5)beta(1) at the cellular level. In addition, these antagonists exerted an antiangiogenic effect in the chick chorioallantoic membrane (CAM) assay.
Collapse
Affiliation(s)
- Monica Bubenik
- Shire BioChem Inc., 275 Armand-Frappier Blvd., Laval, Québec, Canada H7V 4A7.
| | | | | | | | | |
Collapse
|
442
|
Meerovitch K, Bergeron F, Leblond L, Grouix B, Poirier C, Bubenik M, Chan L, Gourdeau H, Bowlin T, Attardo G. A novel RGD antagonist that targets both alphavbeta3 and alpha5beta1 induces apoptosis of angiogenic endothelial cells on type I collagen. Vascul Pharmacol 2003; 40:77-89. [PMID: 12646396 DOI: 10.1016/s1537-1891(02)00339-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Integrin-mediated cell adhesion is necessary for endothelial cell proliferation and apoptosis, which is a major determinant in tumor-induced angiogenesis. In this study, we compared two novel, structurally similar, Arg-Gly-Asp (RGD) peptidomimetic compounds having different integrin selectivities, for their inhibition of endothelial cell proliferation and induction of apoptosis on functionally relevant extracellular matrices (ECM) for angiogenesis. BCH-14661 was specific for integrin alphavbeta3, whereas BCH-15046 nonselectively antagonized integrins alphavbeta3, alphavbeta5, and alpha5beta1. Both compounds were potent inducers of endothelial cell apoptosis when plated on RGD-dependent ECM (vitronectin, VN), which was dependent on the ability to induce cell detachment. However, with endothelial cells plated on RGD-independent ECM (type I collagen, COL), only BCH-15046 was able to significantly prevent growth and induce apoptosis. This effect was not dependent on the induction of detachment. Experiments using the matrix metalloproteinase (MMP) inhibitor GM 6001 revealed that cleavage of COL was not required for the ability of BCH-15046 to induce apoptosis. However, the inhibition of growth factor-stimulated endothelial cell proliferation, required MMPs, and correlated with BCH-15046s' potent inhibition of endothelial cell attachment to denatured collagen. Antibody inhibition experiments showed that adhesion to denatured collagen required integrins alphavbeta3 and beta1, but not alphavbeta5. In addition, BCH-15046 exerted a significant inhibition of VEGF-stimulated angiogenesis in the chick chorioallontoic membrane in vivo. These results suggest that integrin antagonism of both alphavbeta3 and alpha5beta1 are important for MMP-independent induction of apoptosis on COL and MMP-dependent inhibition of endothelial cell-denatured collagen interactions required for proliferation.
Collapse
Affiliation(s)
- Karen Meerovitch
- Department of Cancer Biology, Shire-BioChem Inc., 275 Armand Frappier Boulevard, Laval, Quebec, Canada H7V 4A7.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
443
|
Boudreau N, Myers C. Breast cancer-induced angiogenesis: multiple mechanisms and the role of the microenvironment. Breast Cancer Res 2003; 5:140-6. [PMID: 12793895 PMCID: PMC165004 DOI: 10.1186/bcr589] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Growth and progression of breast cancers are accompanied by increased neovascularization (angiogenesis). A variety of factors, including hypoxia and genetic changes in the tumor cells, contribute to increased production of angiogenic factors. Furthermore, cells within the activated tumor stroma also contribute to the increase in production of vascular endothelial growth factor and other angiogenic factors, including basic fibroblast growth factor and platelet-derived growth factor. The contribution of the microenvironment to tumor-induced angiogenesis is underscored by findings that breast tumors implanted into different tissue sites show marked differences in the extent and nature of the angiogenic response. These findings have important implications for designing anti-angiogenic therapies.
Collapse
Affiliation(s)
- Nancy Boudreau
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA.
| | | |
Collapse
|
444
|
Abstract
Angiogenesis is a critical step in the metastatic cascade of colorectal cancer. Several angiogenesis inhibitors have been evaluated in animal models and have shown efficacy, but challenges remain in using these drugs effectively in the clinical setting. Although several of these angiogenesis inhibitors are currently being evaluated in clinical trials, alone or in combination with cytotoxic chemotherapy, early results suggest that angiogenesis inhibitors alone, when used for advanced disease, have minimal activity. It is likely that this class of drugs will prove more efficacious when used either in the setting of minimal disease as agents that may promote tumor dormancy or in combination with other conventional forms of therapy. In addition, strategies such as metronomic therapy have been proposed whereby lower doses of cytotoxic chemotherapy, given more frequently, may act via an antiangiogenic mechanism [67,68]. Another challenge is identifying methods of assessing response to antiangiogenic therapy. To date, traditional methods of identifying response to treatment have not proven effective. Several investigators are working toward identifying circulating endothelial or tumor-associated factors that may be useful in following treatment. Novel imaging techniques are also being evaluated with enhanced CT and MRI, and newer modalities. Hepatic colorectal metastases provide an opportune setting in which to accomplish these challenges because the high incidence of disease and the ability to measure tumor with a variety of techniques lend themselves to evaluation of antiangiogenic therapy.
Collapse
Affiliation(s)
- Michael O Meyers
- Department of Surgical Oncology, Fox Chase Cancer Center, 7701 Burholme Avenue, Philadelphia, PA 19111, USA
| | | |
Collapse
|
445
|
Abstract
The blood vessels of individual tissues are biochemically distinct, and pathological lesions put their own signature on the vasculature. In tumors, both blood and lymphatic vessels differ from normal vessels. New methods, such as in vivo screening of phage libraries, have provided peptides and antibodies that recognize these vascular signatures and can be used in targeted delivery of therapeutic agents. Targeting a therapy to the diseased tissue enhances the efficacy of the treatment while reducing the side effects in mouse experiments. Results from drug delivery to tumor vessels have been particularly encouraging.
Collapse
Affiliation(s)
- Erkki Ruoslahti
- Cancer Research Center, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
446
|
Davis GE, Bayless KJ, Mavila A. Molecular basis of endothelial cell morphogenesis in three-dimensional extracellular matrices. THE ANATOMICAL RECORD 2002; 268:252-75. [PMID: 12382323 DOI: 10.1002/ar.10159] [Citation(s) in RCA: 180] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although many studies have focused on blood vessel development and new blood vessel formation associated with disease processes, the question of how endothelial cells (ECs) assemble into tubes in three dimensions (i.e., EC morphogenesis) remains unanswered. EC morphogenesis is particularly dependent on a signaling axis involving the extracellular matrix (ECM), integrins, and the cytoskeleton, which regulates EC shape changes and signals the pathways necessary for tube formation. Recent studies reveal that genes regulating this matrix-integrin-cytoskeletal (MIC) signaling axis are differentially expressed during EC morphogenesis. The Rho GTPases represent an important class of molecules involved in these events. Cdc42 and Rac1 are required for the process of EC intracellular vacuole formation and coalescence that regulates EC lumen formation in three-dimensional (3D) extracellular matrices, while RhoA appears to stabilize capillary tube networks. Once EC tube networks are established, supporting cells, such as pericytes, are recruited to further stabilize these networks, perhaps by regulating EC basement membrane matrix assembly. Furthermore, we consider recent work showing that EC morphogenesis is balanced by a tendency for newly formed tubes to regress. This morphogenesis-regression balance is controlled by differential gene expression of such molecules as VEGF, angiopoietin-2, and PAI-1, as well as a plasmin- and matrix metalloproteinase-dependent mechanism that induces tube regression through degradation of ECM scaffolds that support EC-lined tubes. It is our hope that this review will stimulate increased interest and effort focused on the basic mechanisms regulating capillary tube formation and regression in 3D extracellular matrices.
Collapse
Affiliation(s)
- George E Davis
- Department of Pathology, Texas A&M University System Health Science Center, College Station 77843, USA.
| | | | | |
Collapse
|
447
|
Fathallah-Shaykh HM, Rigen M, Zhao LJ, Bansal K, He B, Engelhard HH, Cerullo L, Roenn KV, Byrne R, Munoz L, Rosseau GL, Glick R, Lichtor T, DiSavino E. Mathematical modeling of noise and discovery of genetic expression classes in gliomas. Oncogene 2002; 21:7164-74. [PMID: 12370806 DOI: 10.1038/sj.onc.1205654] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2002] [Revised: 05/30/2002] [Accepted: 05/31/2002] [Indexed: 12/18/2022]
Abstract
The microarray array experimental system generates noisy data that require validation by other experimental methods for measuring gene expression. Here we present an algebraic modeling of noise that extracts expression measurements true to a high degree of confidence. This work profiles the expression of 19 200 cDNAs in 35 human gliomas; the experiments are designed to generate four replicate spots/gene with switching of probes. The validity of the extracted measurements is confirmed by: (1) cluster analysis that generates a molecular classification differentiating glioblastoma from lower-grade tumors and radiation necrosis; (2) By what other investigators have reported in gliomas using paradigms for assaying molecular expression other than gene profiling; and (3) Real-time RT-PCR. The results yield a genetic analysis of gliomas and identify classes of genetic expression that link novel genes to the biology of gliomas.
Collapse
Affiliation(s)
- Hassan M Fathallah-Shaykh
- Department of Neurological Sciences, Rush Presbyterian-St. Lukes Medical Center, Chicago, Illinois, IL 60612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
448
|
Sheppard D. Endothelial integrins and angiogenesis: not so simple anymore. J Clin Invest 2002; 110:913-4. [PMID: 12370267 PMCID: PMC151161 DOI: 10.1172/jci16713] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Dean Sheppard
- Department of Medicine, Sandler Center for Basic Research in Asthma and the Lung Biology Center, University of California, San Francisco, San Francisco, California 94110, USA.
| |
Collapse
|
449
|
|
450
|
Kim S, Bakre M, Yin H, Varner JA. Inhibition of endothelial cell survival and angiogenesis by protein kinase A. J Clin Invest 2002; 110:933-41. [PMID: 12370271 PMCID: PMC151143 DOI: 10.1172/jci14268] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2001] [Accepted: 07/30/2002] [Indexed: 12/29/2022] Open
Abstract
Receptors for the provisional ECM are important regulators of angiogenesis. One of these receptors, integrin alpha5beta1, plays a critical role in tumor- and growth factor-induced angiogenesis, because antagonists of this integrin potently inhibit angiogenesis and tumor growth. Here we show that the integrin alpha5beta1 promotes endothelial cell survival during angiogenesis in vivo by suppressing the activity of protein kinase A (PKA). Antagonists of integrin alpha5beta1 activate PKA, which then leads to the activation of caspase-8 and induction of apoptosis. Direct activation of PKA by cAMP or by expression of the PKA catalytic subunit also induces endothelial cell apoptosis, resulting in angiogenesis inhibition in vivo. Our studies indicate that ligation of integrin alpha5beta1 during angiogenesis suppresses an apoptotic program that is dependent on PKA. These studies also indicate that induction of endothelial cell apoptosis in vivo by genetic or pharmacological activation of PKA may be a useful strategy to inhibit angiogenesis.
Collapse
Affiliation(s)
- Semi Kim
- University of California, San Diego Comprehensive Cancer Center, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|