Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Angiogenesis ; 21(3): 425-532, 2018 08.
Article in English | MEDLINE | ID: mdl-29766399

ABSTRACT

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Subject(s)
Biological Assay/methods , Neoplasms , Neovascularization, Pathologic , Animals , Biological Assay/instrumentation , Guidelines as Topic , Humans , Mice , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
2.
Hum Mutat ; 34(11): 1481-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23946118

ABSTRACT

Dyskeratosis congenita (DC) is a telomere-mediated syndrome defined by mucocutaneous features. The X-linked mode of inheritance accounts for half the cases, and is thought to predominantly manifest in childhood as bone marrow failure. We identified two male probands who presented in the fifth decade with idiopathic pulmonary fibrosis and cancer. Their pedigrees displayed consecutively affected generations. Five of six females (83%) manifested mucocutaneous features of DC, and two had wound-healing complications. No mutations in autosomal dominant telomere genes were present, but exome sequencing revealed novel variants in the X-chromosome DKC1 gene that predicted missense mutations in conserved residues, p.Thr49Ser and p.Pro409Arg. Variants segregated with the telomere phenotype, and affected females were heterozygotes, showing skewed X-inactivation. Telomerase RNA levels were compromised in cells from DKC1 mutation carriers, consistent with their pathogenic role. These findings indicate that females with heterozygous DKC1 mutations may be at increased risk for developing penetrant telomere phenotypes that, at times, may be associated with clinical morbidity.


Subject(s)
Cell Cycle Proteins/genetics , Dyskeratosis Congenita/genetics , Heterozygote , Mutation , Nuclear Proteins/genetics , Phenotype , Telomere/genetics , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Dyskeratosis Congenita/diagnosis , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Nuclear Proteins/chemistry , Sequence Alignment
3.
Res Vet Sci ; 81(1): 76-86, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16256156

ABSTRACT

Canine hemangiosarcoma (HSA) is a devastating disease. Investigation of novel therapies has been limited by the limited availability of canine HSA-derived cell lines. We report the development of a canine HSA-derived cell line, DEN-HSA, which recapitulates features of angiogenic endothelium. DEN-HSA cells were derived from a spontaneous HSA arising in the kidney of a dog. DEN-HSA displayed surface molecules distinctive of endothelial histogenesis, including factor VIII-related antigen, ICAM-1 and alpha(v)beta3 integrin. In vitro, DEN-HSA formed microvascular tube-like structures on Matrigel, and proliferated in response to a variety of angiogenic growth factors. The cells expressed mRNA and protein specific for bFGF and its receptors, and VEGF and its receptors, among others. DEN-HSA conditioned medium evoked a marked angiogenic response in a murine corneal pocket assay, indicating potent proangiogenic activity of substances secreted by this cell line. This research confirms the DEN-HSA cell line as endothelial in origin, suggests the presence of angiogenic growth factor autocrine loops, and offers the potential to utilize DEN-HSA cells for the study of novel therapies that modulate endothelial proliferation.


Subject(s)
Hemangiosarcoma/pathology , Hemangiosarcoma/veterinary , Kidney Neoplasms/pathology , Kidney Neoplasms/veterinary , Tumor Cells, Cultured/metabolism , Angiogenic Proteins/analysis , Angiogenic Proteins/biosynthesis , Animals , Cell Proliferation , Dogs , Flow Cytometry , Immunohistochemistry , Neovascularization, Pathologic , RNA, Messenger/biosynthesis
4.
Mol Cancer Res ; 2(12): 663-73, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15634755

ABSTRACT

We engineered a fusion protein, mrIL-12vp [mouse recombinant interleukin (IL)-12 linked to vascular peptide], linking the vascular homing peptide CDCRGDCFC (RGD-4C), a ligand for alphavbeta3 integrin, to mrIL-12 to target IL-12 directly to tumor neovasculature. The fusion protein stimulated IFN-gamma production in vitro and in vivo, indicating its biological activity was consistent with mrIL-12. Immunofluorescence techniques showed mrIL-12vp specifically bound to alphavbeta3 integrin-positive cells but not to alphavbeta3 integrin-negative cells. In corneal angiogenesis assays using BALB/c mice treated with either 0.5 microg/mouse/d of mrIL-12vp or mrIL-12 delivered by subcutaneous continuous infusion, mrIL-12vp inhibited corneal neovascularization by 67% compared with only a slight reduction (13%) in angiogenesis in the mrIL-12-treated animals (P = 0.008). IL-12 receptor knockout mice given mrIL-12vp showed a marked decrease in the area of corneal neovascularization compared with mice treated with mrIL-12. These results indicate that mrIL-12vp inhibits angiogenesis through IL-12-dependent and IL-12-independent mechanisms, and its augmented antiangiogenic activity may be due to suppression of endothelial cell signaling pathways by the RGD-4C portion of the fusion protein. Mice injected with NXS2 neuroblastoma cells and treated with mrIL-12vp showed significant suppression of tumor growth compared with mice treated with mrIL-12 (P = 0.03). Mice did not show signs of IL-12 toxicity when treated with mrIL-12vp, although hepatic necrosis was present in mrIL-12-treated mice. Localization of IL-12 to neovasculature significantly enhances the antiangiogenic effect, augments antitumor activity, and decreases toxicity of IL-12, offering a promising strategy for expanding development of IL-12 for treatment of cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Integrin alphaVbeta3/metabolism , Interleukin-12/pharmacology , Peptides/chemistry , Animals , CHO Cells , Cell Line, Tumor , Cloning, Molecular , Cricetinae , Flow Cytometry , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Necrosis/pathology , Neoplasm Transplantation , Neovascularization, Pathologic , Oligopeptides/chemistry , Plasmids/metabolism , Protein Engineering , Protein Isoforms , Receptors, Interleukin/genetics , Receptors, Interleukin-12 , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Signal Transduction , Time Factors , Transfection
6.
Neoplasia ; 6(2): 106-16, 2004.
Article in English | MEDLINE | ID: mdl-15140399

ABSTRACT

We established a canine hemangiosarcoma cell line derived from malignant endothelial cells comprising a spontaneous tumor in a dog to provide a renewable source of endothelial cells for studies of angiogenesis in malignancy. Pieces of the hemangiosarcoma biopsy were engrafted subcutaneously in a bg/nu/XID mouse allowing the tumor cells to expand in vivo. A cell line, SB-HSA, was derived from the xenograft. SB-HSA cells expressed vascular endothelial growth factor (VEGF) receptors 1 and 2, CD31, CD146, and alpha(v)beta(3) integrin, and produced several growth factors and cytokines, including VEGF, basic fibroblast growth factor, and interleukin (IL)-8 that are stimulatory to endothelial cell growth. These results indicated that the cells recapitulated features of mitotically activated endothelia. In vivo, SB-HSA cells stimulated robust angiogenic responses in mice and formed tumor masses composed of aberrant vascular channels in immunocompromised mice providing novel opportunities for investigating the effectiveness of antiangiogenic agents. Using this model, we determined that IL-12, a cytokine with both immunostimulatory and antiangiogenic effects, suppressed angiogenesis induced by, and tumor growth of, SB-HSA cells. The endothelial cell model we have described offers unique opportunities to pursue further investigations with IL-12, as well as other antiangiogenic approaches in cancer therapy.


Subject(s)
Corneal Neovascularization/drug therapy , Hemangiosarcoma/blood supply , Interleukin-12/therapeutic use , Neovascularization, Pathologic/drug therapy , Soft Tissue Neoplasms/blood supply , Animals , Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Division , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Cytokines/metabolism , Dogs , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Flow Cytometry , Growth Substances/metabolism , Hemangiosarcoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Reverse Transcriptase Polymerase Chain Reaction , Soft Tissue Neoplasms/pathology , Transplantation, Heterologous , Tumor Cells, Cultured
7.
In Vitro Cell Dev Biol Anim ; 38(6): 334-42, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12513121

ABSTRACT

Endothelial cell lines have been established from cells that were isolated from porcine yolk sacs from day 18 and day 22 embryos and propagated in vitro under various growth conditions. After expansion in vitro, the general properties of the cells proved similar for the different media used. The endothelial cells expressed cell surface receptors for acetylated low-density lipoprotein and also expressed cell surface-associated angiotensin-converting enzyme. The cells showed a characteristically high level of binding for Bandeiraea simplicifolia lectin I and Dolichos biflorus agglutinin but did not bind significant amounts of Ulex europaeus lectin I. The cells expressed low but serologically detectable levels of Class I major histocompatibility complex (MHC) antigens but failed to bind antibodies directed against Class II MHC antigens. Alpha5beta1 integrins were weakly expressed, whereas vascular cell adhesion molecule-1 (CD106) and alphavbeta3 integrins were not detected. Three-dimensional tube formation was readily observed in cultures grown on Matrigel and occurred even in uncoated plastic dishes in the absence of Matrigel. In contrast to most of the adult porcine endothelial cells, yolk sac-derived endothelial cells did not possess serologically detectable receptors for porcine growth hormone (GH), an observation consistent with the finding that GH did not increase the proliferative rate of these cells. Electron microscopic examination demonstrated the presence of Weibel-Palade bodies, tight endothelial cell junctions, and typical rough endoplasmic reticulum. Exposure of the cells to either concanavalin-A-stimulated porcine splenocyte culture supernatants or to human tumor necrosis factor alpha did not cause upregulation of VCAM-1 or Class II MHC antigens. Addition of porcine interferon-gamma led to an increase in the level of expression of Class I MHC. Yolk sac endothelial cells from day 22 embryos showed a low but detectable level of expression of Class II MHC antigens, whereas the endothelial cells from day 18 embryos showed no expression of Class II antigens after interferon-gamma stimulation. The cells maintained competence to develop vascular structures in vitro and could do so after coinjection with murine tumor cells into adult, immunocompromised mice.


Subject(s)
Cell Line , Endothelium, Vascular/cytology , Yolk Sac/cytology , Animals , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Growth Hormone/pharmacology , Histocompatibility Antigens/immunology , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Swine , Transplantation, Heterologous
9.
Biomaterials ; 31(20): 5418-26, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20400175

ABSTRACT

Basement membranes possess a complex three-dimensional topography in the nanoscale and submicron range which have been shown to profoundly modulate a large menu of fundamental cell behaviors. Using the topographic features found in native vascular endothelial basement membranes as a guide, polyurethane substrates were fabricated containing anisotropically ordered ridge and groove structures and isotropically ordered pores from 200 nm to 2000 nm in size. We investigated the impact of biomimetic length-scale topographic cues on orientation/elongation, proliferation and migration on four human vascular endothelial cell-types from large and small diameter vessels. We found that all cell-types exhibited orientation and alignment with the most pronounced response on anisotropically ordered ridges > or =800 nm. HUVEC cells were the only cell-type examined to demonstrate a decrease in proliferation in response to the smallest topographic features regardless of surface order. On anisotropically ordered surfaces all cell-types migrated preferentially parallel to the long axis of the ridges, with the greatest increase in cell migration being observed on the 1200 nm pitch. In contrast, cells did not exhibit any preference in direction or increase in migration speed on isotropically ordered surfaces. Overall, our data demonstrate that surface topographic features impact vascular endothelial cell behavior and that the impact of features varies with the cell behavior being considered, topographic feature scale, surface order, and the anatomic origin of the cell being investigated.


Subject(s)
Endothelial Cells/cytology , Nanostructures/chemistry , Cell Movement , Cell Proliferation , Humans , Particle Size
10.
11.
Angiogenesis ; 5(1-2): 75-80, 2002.
Article in English | MEDLINE | ID: mdl-12549862

ABSTRACT

It has become increasingly clear that definitive tests for angiogenesis require in vivo assays. Recently, the Matrigel plug assay has become the method of choice for many studies involving in vivo testing for angiogenesis. In this assay, test angiogenesis-inducing compounds such as bFGF or tumor cells are introduced into cold liquid Matrigel which, after subcutaneous injection, solidifies and permits penetration by host cells and the formation of new blood vessels. Assessment of angiogenesis in the Matrigel plug is achieved either by measuring hemoglobin or by scoring selected regions of histological sections for vascular density. We now describe a modification of the Matrigel plug assay which permits a more precise visualization of the angiogenic reaction, provides directional information, requires no histological analysis, and lends itself to photographic documentation and image analysis protocols. We illustrate the assay by describing dose- and time-dependent responses to tumors of murine and human origin, to angiogenesis-inducing factors such as bFGF (FGF-2) and VEGF and to anti-angiogenic agents such as endostatin. The method has been used as well to demonstrate blood vessel recruitment by embryonic chick aortic arch rudiments. Additionally it has been able to detect strain-dependent differences in susceptibility to angiogenic stimulation.


Subject(s)
Biological Assay/methods , Collagen/physiology , Laminin/physiology , Neovascularization, Pathologic/diagnosis , Neovascularization, Physiologic/physiology , Proteoglycans/physiology , Angiogenesis Inhibitors/pharmacology , Animals , Collagen/pharmacology , Dogs , Drug Combinations , Endostatins , Endothelial Growth Factors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Growth Substances/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/pharmacology , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Clin Chem ; 49(1): 32-40, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12507958

ABSTRACT

BACKGROUND: Angiogenesis, the formation of new blood vessels, is an integral part of both normal developmental processes and numerous pathologies, ranging from tumor growth and metastasis to inflammation and ocular disease. Angiogenesis assays are used to test efficacy of both pro- and antiangiogenic agents. METHODS: Most studies of angiogenesis inducers and inhibitors rely on various models, both in vitro and in vivo, as indicators of efficacy. In this report we describe the principal methods now in use: the in vivo Matrigel plug and corneal neovascularization assays, the in vivo/in vitro chick chorioallantoic membrane (CAM) assay, and the in vitro cellular (proliferation, migration, tube formation) and organotypic (aortic ring) assays. We include description of two new methods, the chick aortic arch and the Matrigel sponge assays. CONCLUSIONS: In vitro tests are valuable, can be carried out expeditiously, and lend themselves to quantification, but must be interpreted with extreme caution. In vitro tests are best viewed as providing initial information, subject to confirmation by in vivo assays. Multiple tests should be used to obtain maximum benefit from in vitro tests. In vivo tests are more difficult and time-consuming to perform, thereby limiting the number of tests that can run at any one time. Quantification is generally more difficult as well. However, in vivo assays are essential because of the complex nature of vascular responses to test reagents, responses that no in vitro model can fully achieve.


Subject(s)
Clinical Laboratory Techniques , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Cell Culture Techniques , Cell Division , Cell Movement , Chick Embryo , Cornea/blood supply , Endothelium, Vascular/cytology , Humans , Organ Culture Techniques
13.
Blood ; 103(4): 1325-32, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14563647

ABSTRACT

We have used rhesus monkey embryonic stem (ES) cells to study endothelial cell development. Rhesus ES cells (R366.4 cell line) exposed to medium containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF), and epidermal growth factor (EGF) assumed a relatively uniform endothelial cell morphology and could be propagated and expanded with a consistent phenotype and normal karyotype. When placed in Matrigel, these rhesus ES cell-derived endothelial cells (RESDECs) formed capillary-like structures characteristic of endothelial cells. Immunohistochemical and flow cytometric analysis of RESDECs showed that they take up acetylated low-density lipoprotein (LDL), express CD146, von Willebrand factor, and the integrin alpha v beta 3, and bind the lectin ulex europaeus agglutinin-1. These cells also express the VEGF receptor Flk-1 and secrete VEGF. When introduced in a Matrigel plug implanted subcutaneously in mice, RESDECs formed intact vessels and recruited new endothelial cell growth. In vivo function was demonstrated by coinjection of RESDECs with murine tumor cells subcutaneously into immunocompromised adult mice. RESDECs injected alone did not form measurable tumors. Tumor cells grew more rapidly and had increased vascularization when coinjected with the RESDECs. Immunohistochemical staining demonstrated that the RESDECs participated in forming the tumor neovasculature. RESDECs provide a novel means to examine the mechanisms of endothelial cell development, and may open up new therapeutic strategies.


Subject(s)
Endothelium, Vascular/cytology , Stem Cells/cytology , Animals , Biocompatible Materials , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Collagen , Drug Combinations , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Immunophenotyping , Insulin-Like Growth Factor I/pharmacology , Laminin , Macaca mulatta , Mice , Neovascularization, Physiologic/physiology , Proteoglycans , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology
14.
J Immunother ; 27(2): 161-75, 2004.
Article in English | MEDLINE | ID: mdl-14770088

ABSTRACT

The field of tumor angiogenesis has seen explosive growth over the last 5 years. Preclinical as well as early clinical evaluation of novel compounds is progressing at a rapid pace. To gain a perspective on the field and to take stock of advances in the understanding of molecular mechanisms underlying the process of tumor angiogenesis as well as ways of monitoring the activity of agents, the Society for Biologic Therapy and the National Cancer Institute's Vascular Biology Faculty convened a Workshop on Angiogenesis Monitoring in November 2002. The Workshop was composed of invited speakers and participants from academia, industry, and government. It was divided into 3 sessions, each chaired by leaders in the field. The first focused on advances in the understanding of the cellular and molecular mechanisms of angiogenesis in tumors. The second examined preclinical assay systems that are useful in vascular biology. The third addressed the translation to the clinic and monitoring of antiangiogenic activity of agents in patients and novel trial designs. What follows is a summary of the discussions and findings of each session.


Subject(s)
Immunotherapy/methods , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic , Animals , Biomarkers, Tumor , Clinical Trials as Topic , Endothelium/cytology , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL