Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Angiogenesis ; 25(3): 397-410, 2022 08.
Article in English | MEDLINE | ID: mdl-35212873

ABSTRACT

Anthrax protective antigen (PA) is a potent inhibitor of pathological angiogenesis with an unknown mechanism. In anthrax intoxication, PA interacts with capillary morphogenesis gene 2 (CMG2) and tumor endothelial marker 8 (TEM8). Here, we show that CMG2 mediates the antiangiogenic effects of PA and is required for growth-factor-induced chemotaxis. Using specific inhibitors of CMG2 and TEM8 interaction with natural ligand, as well as mice with the CMG2 or TEM8 transmembrane and intracellular domains disrupted, we demonstrate that inhibiting CMG2, but not TEM8 reduces growth-factor-induced angiogenesis in the cornea. Furthermore, the antiangiogenic effect of PA was abolished when the CMG2, but not the TEM8, gene was disrupted. Binding experiments demonstrated a broad ligand specificity for CMG2 among extracellular matrix (ECM) proteins. Ex vivo experiments demonstrated that CMG2 (but not TEM8) is required for PA activity in human dermal microvascular endothelial cell (HMVEC-d) network formation assays. Remarkably, blocking CMG2-ligand binding with PA or CRISPR knockout abolishes endothelial cell chemotaxis but not chemokinesis in microfluidic migration assays. These effects are phenocopied by Rho inhibition. Because CMG2 mediates the chemotactic response of endothelial cells to peptide growth factors in an ECM-dependent fashion, CMG2 is well-placed to integrate growth factor and ECM signals. Thus, CMG2 targeting is a novel way to inhibit angiogenesis.


Subject(s)
Chemotaxis , Endothelial Cells , Neovascularization, Pathologic , Receptors, Peptide , Animals , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Ligands , Mice , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
2.
FASEB J ; 35(5): e21404, 2021 05.
Article in English | MEDLINE | ID: mdl-33899275

ABSTRACT

We have previously used the genetic diversity available in common inbred mouse strains to identify quantitative trait loci (QTLs) responsible for the differences in angiogenic response using the corneal micropocket neovascularization (CoNV) assay. Employing a mouse genome-wide association study (GWAS) approach, the region on chromosome 15 containing Basp1 was identified as being significantly associated with angiogenesis in inbred strains. Here, we developed a unique strategy to determine and verify the role of BASP1 in angiogenic pathways. Basp1 expression in cornea had a strong correlation with a haplotype shared by mouse strains with varied angiogenic phenotypes. In addition, inhibition of BASP1 demonstrated a dosage-dependent effect in both primary mouse brain endothelial and human microvascular endothelial cell (HMVEC) migration. To investigate its role in vivo, we knocked out basp1 in transgenic kdrl:zsGreen zebrafish embryos using a widely adopted CRISPR-Cas9 system. These embryos had severely disrupted vessel formation compared to control siblings. We further show that basp1 promotes angiogenesis by upregulating ß-catenin gene and the Dll4/Notch1 signaling pathway. These results, to the best of our knowledge, provide the first in vivo evidence to indicate the role of Basp1 as an angiogenesis-regulating gene and opens the potential therapeutic avenues for a wide variety of systemic angiogenesis-dependent diseases.


Subject(s)
Corneal Neovascularization/pathology , Membrane Proteins/metabolism , Models, Biological , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Movement , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Genome-Wide Association Study , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Morphogenesis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics , Wnt Signaling Pathway , Zebrafish
3.
Angiogenesis ; 24(1): 111-127, 2021 02.
Article in English | MEDLINE | ID: mdl-32955682

ABSTRACT

Angiogenesis plays a key role in the pathology of diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. Understanding the driving forces of endothelial cell migration and organization, as well as the time frame of these processes, can elucidate mechanisms of action of important pathological pathways. Herein, we have developed an organ-specific microfluidic platform recapitulating the in vivo angiogenic microenvironment by co-culturing mouse primary brain endothelial cells with brain pericytes in a three-dimensional (3D) collagen scaffold. As a proof of concept, we show that this model can be used for studying the angiogenic process and further comparing the angiogenic properties between two different common inbred mouse strains, C57BL/6J and 129S1/SvlmJ. We further show that the newly discovered angiogenesis-regulating gene Padi2 promotes angiogenesis through Dll4/Notch1 signaling by an on-chip mechanistic study. Analysis of the interplay between primary endothelial cells and pericytes in a 3D microfluidic environment assists in the elucidation of the angiogenic response.


Subject(s)
Cell Engineering , Cellular Microenvironment , Endothelial Cells/pathology , Imaging, Three-Dimensional , Microfluidics , Pericytes/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Separation , Cells, Cultured , Down-Regulation , Endothelial Cells/metabolism , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Pericytes/metabolism , Protein-Arginine Deiminase Type 2/antagonists & inhibitors , Protein-Arginine Deiminase Type 2/metabolism , Receptors, Notch/metabolism , Signal Transduction
4.
PLoS Genet ; 13(6): e1006848, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28617813

ABSTRACT

Recent findings indicate that growth factor-driven angiogenesis is markedly influenced by genetic variation. This variation in angiogenic responsiveness may alter the susceptibility to a number of angiogenesis-dependent diseases. Here, we utilized the genetic diversity available in common inbred mouse strains to identify the loci and candidate genes responsible for differences in angiogenic response. The corneal micropocket neovascularization assay was performed on 42 different inbred mouse strains using basic fibroblast growth factor (bFGF) pellets. We performed a genome-wide association study utilizing efficient mixed-model association (EMMA) mapping using the induced vessel area from all strains. Our analysis yielded five loci with genome-wide significance on chromosomes 4, 8, 11, 15 and 16. We further refined the mapping on chromosome 4 within a haplotype block containing multiple candidate genes. These genes were evaluated by expression analysis in corneas of various inbred strains and in vitro functional assays in human microvascular endothelial cells (HMVECs). Of these, we found the expression of peptidyl arginine deiminase type II (Padi2), known to be involved in metabolic pathways, to have a strong correlation with a haplotype shared by multiple high angiogenic strains. In addition, inhibition of Padi2 demonstrated a dosage-dependent effect in HMVECs. To investigate its role in vivo, we knocked down Padi2 in transgenic kdrl:zsGreen zebrafish embryos using morpholinos. These embryos had disrupted vessel formation compared to control siblings. The impaired vascular pattern was partially rescued by human PADI2 mRNA, providing evidence for the specificity of the morphant phenotype. Taken together, our study is the first to indicate the potential role of Padi2 as an angiogenesis-regulating gene. The characterization of Padi2 and other genes in associated pathways may provide new understanding of angiogenesis regulation and novel targets for diagnosis and treatment of a wide variety of angiogenesis-dependent diseases.


Subject(s)
Genome-Wide Association Study , Hydrolases/genetics , Neovascularization, Pathologic/genetics , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblast Growth Factor 2/genetics , Genetic Variation , Haplotypes , Humans , Hydrolases/biosynthesis , Mice , Mice, Inbred Strains , Phenotype , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases , Zebrafish
5.
FASEB J ; 29(2): 662-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406462

ABSTRACT

The incidence of certain angiogenesis-dependent diseases is higher in Caucasians than in African Americans. Angiogenesis is amplified in wound healing and cornea models in albino C57 mice compared with black C57 mice. Moreover, mouse and human melanocytes with low pigmentation stimulate endothelial cell (EC) proliferation and migration in vitro more than melanocytes with high pigmentation. This effect is due, in part, to the secretion of an angiogenic protein called fibromodulin (FMOD) from lowly pigmented melanocytes. Herein, we expand upon the mechanism contributing to increased angiogenesis in lighter skin and report that monocyte chemotactic protein-1 (MCP-1) is secreted by nonpigmented mouse melanocytes by 5- to 10-fold more than pigmented melanocytes. MCP-1 protein stimulates EC proliferation and migration in vitro and angiogenesis in vivo. Mechanistic studies determine that FMOD is upstream of MCP-1 and promotes its secretion from both melanocytes and activated ECs via stimulation of NF-κB activity. Mice injected with FMOD-neutralizing antibodies show 2.3-fold decreased levels of circulating MCP-1. Human studies confirmed that, on average, Caucasians have 2-fold higher serum levels of MCP-1 than African Americans. Taken together, this study implicates the FMOD/MCP-1 pathway in the regulation of angiogenesis by local melanocytes and suggests that melanogenic activity may protect against aberrant angiogenic diseases.


Subject(s)
Chemokine CCL2/metabolism , Melanocytes/cytology , Neovascularization, Pathologic , Skin Pigmentation , Black or African American , Angiogenesis Inducing Agents/metabolism , Animals , Cells, Cultured , Endothelial Cells/cytology , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix Proteins/metabolism , Fibromodulin , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microcirculation , NF-kappa B/metabolism , Pigmentation , Proteoglycans/metabolism
6.
Proc Natl Acad Sci U S A ; 109(28): 11306-11, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733742

ABSTRACT

Antiangiogenesis therapy has become a vital part of the armamentarium against cancer. Hypertension is a dose-limiting toxicity for VEGF inhibitors. Thus, there is a pressing need to address the associated adverse events so these agents can be better used. The hypertension may be mediated by reduced NO bioavailability resulting from VEGF inhibition. We proposed that the hypertension may be prevented by coadministration with endostatin (ES), an endogenous angiogenesis inhibitor with antitumor effects shown to increase endothelial NO production in vitro. We determined that Fc-conjugated ES promoted NO production in endothelial and smooth muscle cells. ES also lowered blood pressure in normotensive mice and prevented hypertension induced by anti-VEGF antibodies. This effect was associated with higher circulating nitrate levels and was absent in eNOS-knockout mice, implicating a NO-mediated mechanism. Retrospective study of patients treated with ES in a clinical trial revealed a small but significant reduction in blood pressure, suggesting that the findings may translate to the clinic. Coadministration of ES with VEGF inhibitors may offer a unique strategy to prevent drug-related hypertension and enhance antiangiogenic tumor suppression.


Subject(s)
Blood Pressure/physiology , Endostatins/metabolism , Hypertension/metabolism , Hypertension/prevention & control , Nitric Oxide/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies/chemistry , Clinical Trials, Phase II as Topic , Female , Heart/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/prevention & control
7.
Front Pharmacol ; 11: 453, 2020.
Article in English | MEDLINE | ID: mdl-32410987

ABSTRACT

Microfluidic-based organ-on-a-chip assays with simultaneous coculture of multi-cell types have been widely utilized for basic research and drug development. Here we describe a novel method for a primary cell-based corneal microphysiological system which aims to recapitulate the basic functions of the in vivo cornea and to study topically applied ocular drug permeation. In this study, the protocols for isolating and cultivating primary corneal epithelial cells and endothelial cells from mouse inbred strain C57BL/6J were optimized, to allow for the development of a primary-cell based microfluidic 3D micro-engineered cornea. This tissue unit, by overcoming the limitations of 2D conventional cell culture, supports new investigations on cornea function and facilitates drug delivery testing.

8.
FASEB J ; 22(10): 3728-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18606869

ABSTRACT

The endothelium lining blood vessels serves as a barrier against vascular hyperpermeability, and its maintenance is critical to organ health. Inflammatory mediators evoke tissue edema by disrupting the expression of membrane junctional proteins, which mediate binding between endothelial cell membranes. Endothelial cell-cell junctions form a diffusion barrier between the intravascular and interstitial space. To prevent the morbidity and mortality caused by exaggerated vascular permeability associated with pathological states (e.g., inflammatory and hypersensitivity disorders, pulmonary edema, traumatic lung injury, cerebral edema resulting from stroke, and others), it is important to develop therapeutic approaches to stabilize these interendothelial junctions. Vascular endothelial growth factor (VEGF), a potent proangiogenic cytokine, was first described as vascular permeability factor (VPF). Doxycycline, a tetracycline derivative, has been shown to inhibit angiogenesis in both humans and animal models. We now report that oral doxycycline prevents VPF/VEGF-induced vascular permeability, interleukin-2-induced pulmonary edema, and delayed-type hypersensitivity (DTH) in mice. Remarkably, doxycycline also inhibits tumor growth and tumor-associated vascular hyperpermeability. Finally, we show that doxycycline targets the adherens junction in vascular endothelial cells by inducing the total amount of VE-cadherin expression while decreasing the degree of its phosphorylation. The potential of doxycyline as a therapeutic inhibitor of vascular hyperpermeability in human clinical conditions is promising and warrants further studies.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Capillary Permeability/drug effects , Cell Membrane/drug effects , Doxycycline/administration & dosage , Endothelium, Vascular/drug effects , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Administration, Oral , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Cell Membrane/metabolism , Cell Proliferation/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Hypersensitivity, Delayed/pathology , Hypersensitivity, Delayed/prevention & control , Interleukin-2/toxicity , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control
9.
FASEB J ; 22(2): 522-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17873101

ABSTRACT

Endometriosis affects 10-15% of women and is associated with pelvic pain and infertility. Angiogenesis plays an essential role in its pathogenesis. Dendritic cells (DCs) were recently implicated in supporting tumor angiogenesis. As both tumors and endometriosis lesions depend on angiogenesis, we investigated the possibility that DCs may also play a role in endometriosis. We induced endometriosis in 8-wk-old female C57BL/6 mice by implantation of autologous endometrium into the peritoneal cavity. We observed an abundance of CD11c(+) DCs infiltrating sites of angiogenesis in endometriosis lesions. We noticed a similar pattern of infiltrating DCs at sites of angiogenesis in the peritoneal Lewis lung carcinoma tumor model. These DCs were immature (major histocompatability complex class II(low)) and expressed vascular endothelial growth factor receptor 2. Peritoneal implanted bone marrow-derived DCs (BMDCs) incorporated into both endometriosis lesions and into B16 melanoma tumors and enhanced their growth at 8 days compared with controls (5.1+/-2.5 vs. 1.5+/-0.5 mm(2), n=4 and 4, P<0.0001 for endometriosis; 67.6+/-15.1 vs. 22.7+/-14.6 mm(2), n=5 and 7, P=0.0004 for mouse melanoma). Finally, immature BMDCs but not mature BMDCs enhanced microvascular endothelial cell migration in vitro (219+/-51 vs. 93+/-32 cells, P=0.02). Based on these findings, we suggest a novel role for DCs in supporting angiogenesis and promoting lesion growth both in endometriosis and in tumors.


Subject(s)
Dendritic Cells , Endometriosis/pathology , Neovascularization, Pathologic/pathology , Animals , Cell Differentiation , Cell Movement , Cell Transplantation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Disease Models, Animal , Endothelial Cells/pathology , Female , Lung Neoplasms/pathology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Neuropharmacology ; 148: 96-106, 2019 04.
Article in English | MEDLINE | ID: mdl-30594697

ABSTRACT

Neurodegenerative diseases affect millions of people worldwide. Optic neuropathies are the most commonly occurring neurodegenerative diseases, characterized by progressive retinal ganglion cell (RGC) degeneration. We recently reported that Prominin-1, a protein found on the surface of stem cells, interacts with VEGF and enhances its activity. VEGF is known to have various protective roles in the nervous system. Subsequently, we have developed a 12-mer peptide derived from Prominin-1, named PR1P, and investigated its effects on neuronal survival of damaged RGCs in a rat model of optic nerve crush (ONC). PR1P prevented RGC apoptosis resulting in improvement of retinal function in the rat ONC model. PR1P treatment significantly increased phosphorylation of ERK and AKT and expression its downstream proteins c-fos and Egr-1 in the retina. Additionally, PR1P beneficially increased the MMP-9/TIMP-1 ratio and promoted glial activation in the retina of ONC rats. Thus, PR1P displayed neuroprotective effects through enhanced VEGF-driven neuronal survival and reconstruction of the extracellular environment in ONC model. Our data indicate that PR1P may be a promising new clinical candidate for the treatment of neurodegenerative diseases.


Subject(s)
Extracellular Matrix/drug effects , Nerve Degeneration/prevention & control , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Early Growth Response Protein 1/biosynthesis , Humans , Male , Matrix Metalloproteinase 9/biosynthesis , Nerve Crush , Neuroglia/metabolism , Neuroprotective Agents/pharmacology , Optic Nerve Injuries/prevention & control , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Retina/metabolism , Retinal Ganglion Cells/drug effects , Tissue Inhibitor of Metalloproteinase-1/biosynthesis
11.
J Clin Invest ; 128(9): 4025-4043, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30102256

ABSTRACT

Impaired lymphangiogenesis is a complication of chronic complex diseases, including diabetes. VEGF-C/VEGFR3 signaling promotes lymphangiogenesis, but how this pathway is affected in diabetes remains poorly understood. We previously demonstrated that loss of epsins 1 and 2 in lymphatic endothelial cells (LECs) prevented VEGF-C-induced VEGFR3 from endocytosis and degradation. Here, we report that diabetes attenuated VEGF-C-induced lymphangiogenesis in corneal micropocket and Matrigel plug assays in WT mice but not in mice with inducible lymphatic-specific deficiency of epsins 1 and 2 (LEC-iDKO). Consistently, LECs isolated from diabetic LEC-iDKO mice elevated in vitro proliferation, migration, and tube formation in response to VEGF-C over diabetic WT mice. Mechanistically, ROS produced in diabetes induced c-Src-dependent but VEGF-C-independent VEGFR3 phosphorylation, and upregulated epsins through the activation of transcription factor AP-1. Augmented epsins bound to and promoted degradation of newly synthesized VEGFR3 in the Golgi, resulting in reduced availability of VEGFR3 at the cell surface. Preclinically, the loss of lymphatic-specific epsins alleviated insufficient lymphangiogenesis and accelerated the resolution of tail edema in diabetic mice. Collectively, our studies indicate that inhibiting expression of epsins in diabetes protects VEGFR3 against degradation and ameliorates diabetes-triggered inhibition of lymphangiogenesis, thereby providing a novel potential therapeutic strategy to treat diabetic complications.


Subject(s)
Adaptor Proteins, Vesicular Transport/deficiency , Diabetes Mellitus, Experimental/metabolism , Lymphangiogenesis/physiology , Vascular Endothelial Growth Factor Receptor-3/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , CSK Tyrosine-Protein Kinase , Diabetes Mellitus, Experimental/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Mice , Mice, Knockout , Models, Biological , Proteolysis , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Vascular Endothelial Growth Factor C/metabolism , src-Family Kinases/metabolism
12.
J Clin Invest ; 124(1): 425-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24355922

ABSTRACT

Studies have established that pigmentation can provide strong, protective effects against certain human diseases. For example, angiogenesis-dependent diseases such as wet age-related macular degeneration and infantile hemangioma are more common in light-skinned individuals of mixed European descent than in African-Americans. Here we found that melanocytes from light-skinned humans and albino mice secrete high levels of fibromodulin (FMOD), which we determined to be a potent angiogenic factor. FMOD treatment stimulated angiogenesis in numerous in vivo systems, including laser-induced choroidal neovascularization, growth factor-induced corneal neovascularization, wound healing, and Matrigel plug assays. Additionally, FMOD enhanced vascular sprouting during normal retinal development. Deletion of Fmod in albino mice resulted in a marked reduction in the amount of neovascularization induced by retinal vein occlusion, corneal growth factor pellets, and Matrigel plugs. Our data implicate the melanocyte-secreted factor FMOD as a key regulator of angiogenesis and suggest an underlying mechanism for epidemiological differences between light-skinned individuals of mixed European descent and African-Americans. Furthermore, inhibition of FMOD in humans has potential as a therapeutic strategy for treating angiogenesis-dependent diseases.


Subject(s)
Extracellular Matrix Proteins/metabolism , Melanocytes/metabolism , Neovascularization, Physiologic , Proteoglycans/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibromodulin , Humans , Mice , Mice, Inbred C57BL , Skin Pigmentation , Transforming Growth Factor beta1/metabolism
13.
PLoS One ; 8(6): e66219, 2013.
Article in English | MEDLINE | ID: mdl-23785488

ABSTRACT

Chronic and recurrent uveitis account for approximately 10% of legal blindness in the western world. Autoimmune uveitis is driven by activated CD4(+) T cells that differentiate into effector T helper cells (Th1, Th2, and Th17) which release proinflammatory cytokines that damage the retina. In this study we investigated the effect of the methionine aminopeptidase 2 (MetAP2) inhibitor, Lodamin, on T cell activation and differentiation. MetAp2 is an enzyme which regulates cellular protein synthesis and is highly expressed in T cells. Lodamin was found to suppress T cell receptor (TCR) mediated T cell proliferation and reduced the production of Th1 and Th17 cells. Further, Lodamin suppressed overall inflammation in the mouse model of experimental autoimmune uveitis (EAU) by a six fold. This effect was attributed in part to a reduction in retinal proinflammatory cytokines, down regulation of MetAP2 expression in purified lymph node CD4(+) T cells, and a general normalization of the systemic immune reaction.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Polyesters/pharmacology , Retinitis/immunology , Retinitis/metabolism , Sesquiterpenes/pharmacology , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Animals , Autoimmune Diseases/drug therapy , CD3 Complex/metabolism , Cell Differentiation/drug effects , Cytokines/biosynthesis , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Mice , Models, Biological , Polyesters/administration & dosage , Retinitis/drug therapy , Sesquiterpenes/administration & dosage , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Uveitis/drug therapy , Uveitis/immunology
14.
J Med Chem ; 56(5): 1940-5, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23394144

ABSTRACT

Capillary morphogenesis gene 2 (CMG2) is a transmembrane extracellular matrix binding protein that is also an anthrax toxin receptor. We have shown that high-affinity CMG2 binders can inhibit angiogenesis and tumor growth. We recently described a high-throughput FRET assay to identify CMG2 inhibitors. We now report the serendipitous discovery that PGG (1,2,3,4,6-penta-O-galloyl-ß-D-glucopyranose) is a CMG2 inhibitor with antiangiogenic activity. PGG is a gallotannin produced by a variety of medicinal plants that exhibits a wide variety of antitumor and other activities. We find that PGG inhibits CMG2 with a submicromolar IC50 and it also inhibits the migration of human dermal microvascular endothelial cells at similar concentrations in vitro. Finally, oral or intraperitoneal administration of PGG inhibits angiogenesis in the mouse corneal micropocket assay in vivo. Together, these results suggest that a portion of the in vivo antitumor activity of PGG may be the result of antiangiogenic activity mediated by inhibition of CMG2.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Hydrolyzable Tannins/pharmacology , Neovascularization, Pathologic/drug therapy , Receptors, Peptide/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Humans , Mice , Receptors, Peptide/physiology
15.
PLoS One ; 7(6): e39911, 2012.
Article in English | MEDLINE | ID: mdl-22768167

ABSTRACT

Anti-angiogenic therapies are effective for the treatment of cancer, a variety of ocular diseases, and have potential benefits in cardiovascular disease, arthritis, and psoriasis. We have previously shown that anthrax protective antigen (PA), a non-pathogenic component of anthrax toxin, is an inhibitor of angiogenesis, apparently as a result of interaction with the cell surface receptors capillary morphogenesis gene 2 (CMG2) protein and tumor endothelial marker 8 (TEM8). Hence, molecules that bind the anthrax toxin receptors may be effective to slow or halt pathological vascular growth. Here we describe development and testing of an effective homogeneous steady-state fluorescence resonance energy transfer (FRET) high throughput screening assay designed to identify molecules that inhibit binding of PA to CMG2. Molecules identified in the screen can serve as potential lead compounds for the development of anti-angiogenic and anti-anthrax therapies. The assay to screen for inhibitors of this protein-protein interaction is sensitive and robust, with observed Z' values as high as 0.92. Preliminary screens conducted with a library of known bioactive compounds identified tannic acid and cisplatin as inhibitors of the PA-CMG2 interaction. We have confirmed that tannic acid both binds CMG2 and has anti-endothelial properties. In contrast, cisplatin appears to inhibit PA-CMG2 interaction by binding both PA and CMG2, and observed cisplatin anti-angiogenic effects are not mediated by interaction with CMG2. This work represents the first reported high throughput screening assay targeting CMG2 to identify possible inhibitors of both angiogenesis and anthrax intoxication.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays/methods , Membrane Proteins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cisplatin/pharmacology , Immobilized Proteins/metabolism , Inhibitory Concentration 50 , Kinetics , Membrane Proteins/antagonists & inhibitors , Mice , Pilot Projects , Protein Binding/drug effects , Receptors, Peptide , Reproducibility of Results , Surface Plasmon Resonance , Tannins/pharmacology , Time Factors
16.
PLoS One ; 5(9)2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20824139

ABSTRACT

UNLABELLED: Pathological neovascularization is a hallmark of late stage neovascular (wet) age-related macular degeneration (AMD) and the leading cause of blindness in people over the age of 50 in the western world. The treatments focus on suppression of choroidal neovascularization (CNV), while current approved therapies are limited to inhibiting vascular endothelial growth factor (VEGF) exclusively. However, this treatment does not address the underlying cause of AMD, and the loss of VEGF's neuroprotective can be a potential side effect. Therapy which targets the key processes in AMD, the pathological neovascularization, vessel leakage and inflammation could bring a major shift in the approach to disease treatment and prevention. In this study we have demonstrated the efficacy of such broad spectrum antiangiogenic therapy on mouse model of AMD. METHODS AND FINDINGS: Lodamin, a polymeric formulation of TNP-470, is a potent broad-spectrum antiangiogenic drug. Lodamin significantly reduced key processes involved in AMD progression as demonstrated in mice and rats. Its suppressive effects on angiogenesis, vascular leakage and inflammation were studied in a wide array of assays including; a Matrigel, delayed-type hypersensitivity (DTH), Miles assay, laser-induced CNV and corneal micropocket assay. Lodamin significantly suppressed the secretion of various pro-inflammatory cytokines in the CNV lesion including monocyte chemotactic protein-1 (MCP-1/Ccl2). Importantly, Lodamin was found to regress established CNV lesions, unlike soluble fms-like tyrosine kinase-1 (sFlk-1). The drug was found to be safe in mice and have little toxicity as demonstrated by electroretinography (ERG) assessing retinal and by histology. CONCLUSIONS: Lodamin, a polymer formulation of TNP-470, was identified as a first in its class, broad-spectrum antiangiogenic drug that can be administered orally or locally to treat corneal and retinal neovascularization. Several unique properties make Lodamin especially beneficial for ophthalmic use. Our results support the concept that broad spectrum antiangiogenic drugs are promising agents for AMD treatment and prevention.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cyclohexanes/therapeutic use , Macular Degeneration/drug therapy , Sesquiterpenes/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/toxicity , Animals , Cyclohexanes/administration & dosage , Cyclohexanes/toxicity , Cytokines/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Macular Degeneration/immunology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , O-(Chloroacetylcarbamoyl)fumagillol , Rats , Rats, Inbred Lew , Sesquiterpenes/administration & dosage , Sesquiterpenes/toxicity
17.
Invest Ophthalmol Vis Sci ; 49(8): 3666-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18408184

ABSTRACT

PURPOSE: Dendritic cells (DCs) are innate immune cells that have recently been shown to support angiogenesis in tumors, endometriosis, and lymph nodes. A major cause of legal blindness is wet age-related macular degeneration (wet ARMD), wherein abnormal blood vessels grow under the retina, an abnormality also referred to as choroidal neovascularization (CNV). The purpose of the present study was to investigate the role of DCs in the development of CNV. METHODS: Laser photocoagulation was used to induce CNV in C57BL/6J mice. The authors analyzed CNV lesions for the presence of DCs using flow cytometry and immunostaining at designated times. They also analyzed the effects of intravenous DC transplantation on CNV development by measuring the lesion area using confocal microscopy 1 week after laser injury. RESULTS: The authors analyzed CNV lesions for the presence of DCs by flow cytometry and observed that CD11c(+) major histocompatibility complex (MHC) class II(+) DCs transiently infiltrated the CNV lesions, reaching a peak at 2 to 4 days after laser injury. These DCs were mostly immature (CD11c(+) MHCII(low)) and expressed vascular endothelial growth factor receptor 2. Immunostaining of laser-induced CNV lesions confirmed that DCs are located at the sites of newly formed blood vessels. Intravenously injected DCs incorporated into the CNV lesions. However, only immature DCs enhanced CNV size. CONCLUSIONS: These results suggest a role for DCs in promoting angiogenesis and lesion growth in laser-induced CNV. The present data suggest that DCs may represent potential cellular targets for therapeutic intervention in wet ARMD.


Subject(s)
Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Dendritic Cells/physiology , Animals , CD11c Antigen/metabolism , Cell Movement , Cell Transplantation , Cells, Cultured , Choroid/blood supply , Dendritic Cells/transplantation , Disease Models, Animal , Endothelium, Vascular/metabolism , Flow Cytometry , Histocompatibility Antigens Class II/metabolism , Laser Coagulation , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Nat Biotechnol ; 26(7): 799-807, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18587385

ABSTRACT

Targeting angiogenesis, the formation of blood vessels, is an important modality for cancer therapy. TNP-470, a fumagillin analog, is among the most potent and broad-spectrum angiogenesis inhibitors. However, a major clinical limitation is its poor oral availability and short half-life, necessitating frequent, continuous parenteral administration. We have addressed these issues and report an oral formulation of TNP-470, named Lodamin. TNP-470 was conjugated to monomethoxy-polyethylene glycol-polylactic acid to form nanopolymeric micelles. This conjugate can be absorbed by the intestine and selectively accumulates in tumors. Lodamin significantly inhibits tumor growth, without causing neurological impairment in tumor-bearing mice. Using the oral route of administration, it first reaches the liver, making it especially efficient in preventing the development of liver metastasis in mice. We show that Lodamin is an oral nontoxic antiangiogenic drug that can be chronically administered for cancer therapy or metastasis prevention.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Cyclohexanes/administration & dosage , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Polyesters/administration & dosage , Sesquiterpenes/administration & dosage , Administration, Oral , Animals , Cell Line, Tumor , Humans , Mice , O-(Chloroacetylcarbamoyl)fumagillol , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL