Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
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
2.
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
3.
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
4.
Angiogenesis ; 20(3): 399-408, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28397127

ABSTRACT

Therapeutic angiogenesis is an experimental frontier in vascular biology that seeks to deliver angiogenic growth factors to ischemic or injured tissues to promote targeted formation of new blood vessels as an alternative approach to surgical revascularization procedures. Vascular endothelial growth factor (VEGF) is a potent angiogenic signal protein that is locally upregulated at sites of tissue injury. However, therapies aimed at increasing VEGF levels experimentally by injecting VEGF gene or protein failed to improve outcomes in human trials in part due to its short half-life and systemic toxicity. We recently designed a novel 12-amino acid peptide (PR1P) whose sequence was derived from an extracellular VEGF-binding domain of the pro-angiogenic glycoprotein prominin-1. In this study, we characterized the molecular binding properties of this novel potential therapeutic for targeted angiogenesis and provided the foundation for its use as an angiogenic molecule that can potentiate endogenous VEGF. We showed that PR1P bound VEGF directly and enhanced VEGF binding to endothelial cells and to VEGF receptors VEGFR2 and neuropilin-1. PR1P increased angiogenesis in the murine corneal micropocket assay when combined with VEGF, but had no activity without added VEGF. In addition, PR1P also enhanced angiogenesis in murine choroidal neovascularization and wound-healing models and augmented reperfusion in a murine hind-limb ischemia model. Together our data suggest that PR1P enhanced angiogenesis by potentiating the activity of endogenous VEGF. In so doing, this novel therapy takes advantage of endogenous VEGF gradients generated in injured tissues and may improve the efficacy of and avoid systemic toxicity seen with previous VEGF therapies.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Peptides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Female , Humans , Ischemia/pathology , Mice, Inbred C57BL , Perfusion , Protein Binding/drug effects
5.
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
6.
Angiogenesis ; 16(3): 639-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23423728

ABSTRACT

We have observed substantial differences in angiogenic responsiveness in mice and have mapped the genetic loci responsible for these differences. We have found that the albino mutation is one of the loci responsible for such differences. Using B6.A consomic strains, we determined that chromosome 7 bears a locus that inhibits VEGF-induced corneal neovascularization. F2 crosses between B6.A consomic mice and C57BL/6J parents along with AXB and BXA recombinant inbred strains demonstrated highest linkage near the tyrosinase gene. This region was named AngVq4. Congenic animals confirmed this locus, but could not demonstrate that the classical tyrosinase albino (c) mutation was causative because of the existence of additional linked loci in the congenic region. However, in 1970, a second tyrosinase albino mutation (c-2J) arose in the C57BL/6J background at Jackson Labs. Testing this strain (C57BL/6J) demonstrated that the albino mutation is sufficient to completely explain the alteration in angiogenic response that we observed in congenic animals. Thus, we conclude that the classical tyrosinase mutation is responsible for AngVq4. In contrast to the cornea, where pigmented animals exhibit increased angiogenic responsiveness, iris neovascularization was inhibited in pigmented animals. These results may partially explain increased aggressiveness in amelanotic melanoma, as well as ethnic differences in diabetic retinopathy and macular degeneration.


Subject(s)
Albinism, Oculocutaneous/genetics , Corneal Neovascularization/enzymology , Monophenol Monooxygenase/genetics , Mutation/genetics , Animals , Corneal Neovascularization/genetics , Crosses, Genetic , Endothelial Cells/physiology , Genetic Linkage , Genotype , Humans , Mice , Mice, Inbred Strains
7.
Angiogenesis ; 16(3): 541-51, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23334337

ABSTRACT

Endometriosis is an angiogenesis-dependent disease. Many studies demonstrated inhibition of angiogenesis leads to inhibition of endometriotic growth, however underlying mechanism is still not fully understood. Our previous study suggested vascular endothelial growth factor C (VEGF-C) as a target of anti-angiogenesis therapy for endometriosis. In this study, VEGF-C in endometrium and its role in angiogenesis of endometriosis were studied. Human endometrium were obtained from women with and without endometriosis for molecular studies. VEGF-A, VEGF-B, VEGF-C and VEGF-D mRNA and proteins in eutopic and ectopic endometrium were measured. Human endothelial cells were transfected with VEGF-C siRNA in vitro, effects of VEGF-C on endothelial cell migration, invasion and tube formation were investigated in vitro. Angiogenesis was inhibited in wild type mice, vascular permeability in dermal skin was determined in vivo. Transplanted endometrium were inhibited by VEGF-C siRNA in immunocompromised mice, development, growth and angiogenesis of the experimental endometriosis were compared in vivo. The results showed that VEGF-C mRNA and protein were increased in eutopic and ectopic endometrium of endometriosis patients. VEGF-C siRNA significantly inhibited endothelial cell migration and tube formation. VEGF-C siRNA significantly inhibited development and angiogenesis of the experimental endometriotic lesions in mice. Supplementation and over-expression of VEGF-C significantly reversed the inhibitory effects on the endothelial functions, vascular permeability and endometriotic growth. In conclusion, VEGF-C is increased in endometrium and it promotes endothelial functions, vascular permeability and development of experimental endometriosis. VEGF-C is important for angiogenesis in endometriosis.


Subject(s)
Capillary Permeability/physiology , Endometriosis/metabolism , Endometrium/metabolism , Endothelial Cells/physiology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor C/metabolism , Analysis of Variance , Animals , Cell Movement/drug effects , Cell Movement/physiology , Endometrium/cytology , Endothelial Cells/metabolism , Female , Humans , Mice , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology
8.
J Clin Invest ; 132(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36453543

ABSTRACT

Glaucoma is a highly heritable disease that is a leading cause of blindness worldwide. Here, we identified heterozygous thrombospondin 1 (THBS1) missense alleles altering p.Arg1034, a highly evolutionarily conserved amino acid, in 3 unrelated and ethnically diverse families affected by congenital glaucoma, a severe form of glaucoma affecting children. Thbs1R1034C-mutant mice had elevated intraocular pressure (IOP), reduced ocular fluid outflow, and retinal ganglion cell loss. Histology revealed an abundant, abnormal extracellular accumulation of THBS1 with abnormal morphology of juxtacanalicular trabecular meshwork (TM), an ocular tissue critical for aqueous fluid outflow. Functional characterization showed that the THBS1 missense alleles found in affected individuals destabilized the THBS1 C-terminus, causing protein misfolding and extracellular aggregation. Analysis using a range of amino acid substitutions at position R1034 showed that the extent of aggregation was correlated with the change in protein-folding free energy caused by variations in amino acid structure. Extracellular matrix (ECM) proteins, especially fibronectin, which bind to THBS1, also accumulated within THBS1 deposits. These results show that missense variants altering THBS1 p.Arg1034 can cause elevated IOP through a mechanism involving impaired TM fluid outflow in association with accumulation of aggregated THBS1 in the ECM of juxtacanalicular meshwork with altered morphology.


Subject(s)
Glaucoma , Trabecular Meshwork , Animals , Mice , Extracellular Matrix Proteins , Thrombospondin 1/genetics , Alleles , Glaucoma/genetics , Amino Acids
9.
FASEB J ; 23(7): 2235-43, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19237505

ABSTRACT

Angiogenesis is controlled by a balance between stimulators and inhibitors. We propose that the balance, as well as the general sensitivity of the endothelium to these factors, varies from individual to individual. Indeed, we have found that individual mouse strains have dramatically different responses to growth factor-induced neovascularization. Quantitative trait loci (QTLs), which influence the extent of corneal angiogenesis induced by vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF2), were previously identified by our laboratory. To investigate the genetic contribution to choroidal neovascularization (CNV), a leading cause of blindness, we have undertaken a similar mapping approach to identify QTLs that influence laser-induced CNV in the BXD series of recombinant inbred mouse strains. Composite interval mapping identified new angiogenic QTLs on chromosomes 2 and 19, in addition to confirming our previous corneal neovascularization QTLs of AngVq1 and AngFq2. The new QTLs are named AngCNVq1 and AngCNVq2. The newly mapped regions contain several candidate genes involved in the angiogenic process, including thrombospondin 1, delta-like 4, BclII modifying factor, phospholipase C, beta 2, adrenergic receptor, beta 1, actin-binding LIM protein 1 and colony stimulating factor 2 receptor, alpha. Differences in these regions may control individual susceptibility to CNV.


Subject(s)
Choroidal Neovascularization/genetics , Lasers/adverse effects , Quantitative Trait Loci/genetics , Animals , Chromosome Mapping , Chromosomes , Genes , Genetic Predisposition to Disease , Mice , Mice, Inbred Strains , Neovascularization, Pathologic/genetics
10.
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.

11.
Sci Rep ; 10(1): 18034, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093573

ABSTRACT

Amiodarone is an anti-arrhythmic drug that was approved by the US Food and Drug Administration (FDA) in 1985. Pre-clinical studies suggest that Amiodarone induces cytotoxicity in several types of cancer cells, thus making it a potential candidate for use as an anti-cancer treatment. However, it is also known to cause a variety of severe side effects. We hypothesized that in addition to the cytotoxic effects observed in cancer cells Amiodarone also has an indirect effect on angiogensis, a key factor in the tumor microenvironment. In this study, we examined Amiodarone's effects on a murine tumor model comprised of U-87 MG glioblastoma multiforme (GBM) cells, known to form highly vascularized tumors. We performed several in vitro assays using tumor and endothelial cells, along with in vivo assays utilizing three murine models. Low dose Amiodarone markedly reduced the size of GBM xenograft tumors and displayed a strong anti-angiogenic effect, suggesting dual cancer fighting properties. Our findings lay the ground for further research of Amiodarone as a possible clinical agent that, used in safe doses, maintains its dual properties while averting the drug's harmful side effects.


Subject(s)
Amiodarone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Neovascularization, Pathologic/prevention & control , Vasodilator Agents/pharmacology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Dose-Response Relationship, Drug , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Cancer Res ; 67(5): 2040-5, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332332

ABSTRACT

Radiation can potentially suppress neovascularization by inhibiting the incorporation of hematopoietic precursors as well as damaging mature endothelial cells. The purpose of these studies was to quantify the effect of radiation on angiogenesis and to examine the relationship between bone marrow reconstitution and neovascularization. Immune competent, severe combined immunodeficient, RAG1-deficient, and green fluorescence protein transgenic mice in the C57 genetic background, as well as the highly angiogenic 129S1/SvlmJ strain of mice, underwent whole-body or localized exposure to radiation. The hematopoietic systems in the irradiated recipients were restored by bone marrow transfer. Hematopoietic reconstitution was assessed by doing complete blood counts. Angiogenesis was induced in the mouse cornea using 80 ng of purified basic fibroblast growth factor, and the neovascular response was quantified using a slit lamp biomicroscope. Following whole-body exposure and bone marrow transplantation, the hematopoietic system was successfully reconstituted over time, but the corneal angiogenic response was permanently and significantly blunted up to 66%. Localized exposure of the eyes to radiation suppressed corneal angiogenesis comparably to whole-body exposure. Whole-body irradiation with ocular shielding induced bone marrow suppression but did not inhibit corneal neovascularization. In mice exposed to radiation before tumor implantation, the reduced local angiogenic response correlated with significantly reduced growth of tumor cells in vivo. These results indicate that bone marrow suppression does not suppress neovascularization in the mouse cornea and that although hematopoietic stem cells can readily reconstitute peripheral blood, they do not restore a local radiation-induced deficit in neovascular response.


Subject(s)
Corneal Neovascularization , Hematopoiesis/radiation effects , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Cells/physiology , Carcinoma, Lewis Lung/pathology , Corneal Neovascularization/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Tumor Cells, Cultured , Whole-Body Irradiation/adverse effects
13.
Cancer Res ; 67(20): 9980-5, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17942931

ABSTRACT

Bacillus anthracis protective antigen (PA), the B subunit of the binary anthrax toxin, binds to the cellular receptors capillary morphogenesis gene 2 protein and tumor endothelial marker 8 with high affinity. Both receptors are expressed on endothelial cells during angiogenesis. We sought to determine whether one could inhibit angiogenesis by interfering with the binding of these receptors to their endogenous ligands. Here, we show that wild-type PA inhibits both vascular endothelial growth factor-induced and basic fibroblast growth factor-induced angiogenesis at moderate but statistically significant levels. Structure-activity studies identified a PA mutant that exhibited markedly enhanced inhibition of angiogenesis and also inhibited tumor growth in vivo. This mutant, PASSSR, is unable to undergo normal cellular processing and, thus, remains bound to the surface receptor. Further mutation of PASSSR so that it does not bind to these cell surface receptors abolished its ability to inhibit angiogenesis. We conclude that high-affinity anthrax toxin receptor (ATR) ligands, such as PA and PASSSR, are angiogenesis inhibitors and that ATRs are useful targets for antiangiogenic therapy. These results also suggest that endothelial cell-binding proteins from additional pathogens may inhibit angiogenesis and raise the question of the role of such inhibition in pathogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Growth Processes/drug effects , Cornea/blood supply , Corneal Neovascularization/chemically induced , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins , Mutation , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/metabolism , Receptors, Peptide , Vascular Endothelial Growth Factor A/antagonists & inhibitors
14.
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
15.
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
16.
Cancer Res ; 65(9): 3656-63, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867360

ABSTRACT

The first recombinant endostatin that elicited strong antitumor activity was expressed in Escherichia coli and administered as a suspension. Under these conditions, the protein retained its full antiangiogenic activity. Lack of requirement for a folded structure prompted us to investigate antitumor properties of synthetic peptides corresponding to different regions of endostatin. Here, we show that the entire antitumor, antimigration, and antipermeability activities of endostatin are mimicked by a 27-amino-acid peptide corresponding to the NH2-terminal domain of endostatin. This peptide contains three histidines that are responsible for zinc binding. Mutations of the zinc-binding histidines abolished its antitumor and antimigration activities, but not antipermeability properties.


Subject(s)
Endostatins/pharmacology , Peptide Fragments/pharmacology , Zinc/metabolism , Adenocarcinoma/drug therapy , Amino Acid Sequence , Animals , Carcinoma, Lewis Lung/drug therapy , Cell Movement/drug effects , Endostatins/chemistry , Endothelial Cells/cytology , Endothelial Cells/drug effects , Histidine/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Pancreatic Neoplasms/drug therapy , Peptide Fragments/chemistry , Protein Conformation , Protein Structure, Tertiary , Structure-Activity Relationship , Xenograft Model Antitumor Assays , Zinc/chemistry
17.
Cancer Res ; 62(8): 2300-5, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11956087

ABSTRACT

Thalidomide has recently been shown to be useful in the treatment of multiple myeloma and may also be useful in the treatment of other hematological malignancies. We have identified a new derivative of thalidomide, S-3-[3-amino-phthalimido]-glutarimide (S-3APG) with dual activity against B-cell neoplasias. S-3APG was able to directly inhibit the proliferation of myeloma and Burkitt's lymphoma cell lines in vitro without showing toxicity to normal bone marrow stromal cells or hematopoietic progenitor cells. In vivo, S-3APG treatment of drug resistant myeloma cell tumors in mice was able to produce complete and sustained regressions without any observed toxicity. Additionally, S-3APG induced complete regressions of Burkitt's lymphoma cell tumors. Furthermore, S-3APG inhibited angiogenesis more potently than thalidomide in the murine corneal micropocket model. We conclude that S-3APG is a powerful anti-myeloma and anti-B-cell-lymphoma agent that has both antiproliferative and antiangiogenic effects.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Burkitt Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Thalidomide/pharmacology , 3T3 Cells , Animals , Burkitt Lymphoma/pathology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Cell Division/drug effects , Growth Inhibitors/pharmacology , Humans , Interleukin-6/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Multiple Myeloma/pathology , Neovascularization, Physiologic/drug effects , Thalidomide/analogs & derivatives , Xenograft Model Antitumor Assays
18.
FASEB J ; 17(14): 2112-4, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12958152

ABSTRACT

Angiogenesis is regulated by the balance between angiogenic stimulators and inhibitors. Numerous reports have demonstrated that tumors induce aggressive angiogenesis by up-regulating the production of angiogenesis stimulating growth factors to overcome the baseline levels of endogenous inhibitors. However, the possibility of large differences in the host's responsiveness to angiogenic factors has been largely overlooked. Using the corneal micropocket neovascularization assay, we have observed >10-fold differences in responsiveness to either basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF) among various mouse strains. The inheritance pattern observed for these traits supported a QTL (quantitative trait locus) approach to mapping the genes responsible for the differences in angiogenic responsiveness. To overcome variability in the assay, we used recombinant inbred lines to map this phenotype. In the BXD series of recombinant inbred mouse strains, we have mapped the regions responsible for regulating VEGF-induced angiogenesis using both composite interval mapping and multiple interval mapping. Both approaches link VEGF responsiveness to regions on chromosomes 2 (near D2Mit6) and 10 (near D10Mit20). Candidate angiogenesis-related genes in these regions include those for collagen XVIII/endostatin, matrix metalloproteinase 11, integrin beta2, prostaglandin D2 synthase, and interleukin-1 receptor antagonist.


Subject(s)
Mice/genetics , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor A/pharmacology , Animals , Chromosome Mapping , Fibroblast Growth Factor 2/pharmacology , Genes, Dominant , Genetic Linkage , Genetic Variation , Mice/physiology , Mice, Inbred Strains , Models, Biological , Quantitative Trait Loci , Species Specificity
19.
FASEB J ; 18(10): 1050-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15226265

ABSTRACT

Angiogenesis is controlled by a balance between stimulatory growth factors and endogenous inhibitors. We propose that the balance of stimulators and inhibitors, as well as the general sensitivity of the endothelium to these factors, varies from individual to individual. Indeed, we have found that individual mouse strains have dramatically different responses to growth factor-induced neovascularization. Quantitative trait loci (QTLs), which influence the extent of angiogenesis induced by vascular endothelial growth factor (VEGF), were previously identified by our laboratory. Since genetic susceptibility may vary according to the angiogenic stimulator, we have undertaken a similar mapping approach to identify QTLs that influence basic fibroblast growth factor (FGF2) induced neovascularization in the BXD series of recombinant inbred mouse strains. Composite and multiple interval mapping identified areas of chromosomes 4, 13, 15, and 18. These new angiogenesis QTLs, named AngFq1 through AngFq4 (for angiogenesis due to FGF2), are different from previously identified VEGF QTLs. The mapped regions contain several genes involved in the angiogenic process including matrix metalloproteinase 16, eph receptor A7, angiopoetin 1, endothelial lipase, and autotaxin. Differences in these regions may influence individual susceptibility to angiogenesis related diseases such as cancer, macular degeneration, atherosclerosis, and arthritis.


Subject(s)
Corneal Neovascularization/genetics , Fibroblast Growth Factor 2/pharmacology , Mice, Inbred Strains/genetics , Quantitative Trait Loci , Animals , Chromosome Mapping , Genotype , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Species Specificity , Vascular Endothelial Growth Factor A/pharmacology
20.
J Vis Exp ; (90)2014 Aug 16.
Article in English | MEDLINE | ID: mdl-25177860

ABSTRACT

The mouse corneal micropocket assay is a robust and quantitative in vivo assay for evaluating angiogenesis. By using standardized slow-release pellets containing specific growth factors that trigger blood vessel growth throughout the naturally avascular cornea, angiogenesis can be measured and quantified. In this assay the angiogenic response is generated over the course of several days, depending on the type and dose of growth factor used. The induction of neovascularization is commonly triggered by either basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF). By combining these growth factors with sucralfate and hydron (poly-HEMA (poly(2-hydroxyethyl methacrylate))) and casting the mixture into pellets, they can be surgically implanted in the mouse eye. These uniform pellets slowly-release the growth factors over five or six days (bFGF or VEGF respectively) enabling sufficient angiogenic response required for vessel area quantification using a slit lamp. This assay can be used for different applications, including the evaluation of angiogenic modulator drugs or treatments as well as comparison between different genetic backgrounds affecting angiogenesis. A skilled investigator after practicing this assay can implant a pellet in less than 5 min per eye.


Subject(s)
Cornea/blood supply , Corneal Neovascularization , Neovascularization, Physiologic/physiology , Animals , Cornea/drug effects , Delayed-Action Preparations , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacology , Mice , Models, Animal , Neovascularization, Physiologic/drug effects , Polyhydroxyethyl Methacrylate/chemistry , Sucralfate/chemistry , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL