Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
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
2.
Proc Natl Acad Sci U S A ; 119(45): e2204795119, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36322719

ABSTRACT

Abnormal neovascularization is an important cause of blindness in many ocular diseases, for which the etiology and pathogenic mechanisms remain incompletely understood. Recent studies have revealed the diverse roles of noncoding RNAs in various biological processes and facilitated the research and development of the clinical application of numerous RNA drugs, including microRNAs. Here, we report the antiangiogenic activity of microRNA-29a (miR-29a) in three animal models of ocular neovascularization. The miR-29a knockout (KO) mice displayed enhanced vessel pruning, resulting in a decreased vascularized area during retinal development. In contrast, miR-29a deletion in adult mice accelerated angiogenesis in preclinical disease models, including corneal neovascularization, oxygen-induced retinopathy, and choroidal neovascularization, while the administration of agomir-29a ameliorated pathological neovascularization. Furthermore, miR-29a exerted inhibitory effects on endothelial cell proliferation, migration, and tube formation capacities. RNA sequencing analysis of retinas from miR-29a KO mice and RNA interference experiments identified platelet-derived growth factor C and several extracellular matrix genes as downstream targets of miR-29a involved in regulating ocular angiogenesis. Our data suggest that miR-29a may be a promising clinical candidate for the treatment of neovascular diseases.


Subject(s)
Choroidal Neovascularization , MicroRNAs , Mice , Animals , MicroRNAs/metabolism , Cell Proliferation , RNA Interference , Eye/metabolism , Choroidal Neovascularization/metabolism , Mice, Knockout
3.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068392

ABSTRACT

Myocardial infarction (MI) remains the leading cause of death in the western world. Despite advancements in interventional revascularization technologies, many patients are not candidates for them due to comorbidities or lack of local resources. Non-invasive approaches to accelerate revascularization within ischemic tissues through angiogenesis by providing Vascular Endothelial Growth Factor (VEGF) in protein or gene form has been effective in animal models but not in humans likely due to its short half-life and systemic toxicity. Here, we tested the hypothesis that PR1P, a small VEGF binding peptide that we developed, which stabilizes and upregulates endogenous VEGF, could be used to improve outcome from MI in rodents. To test this hypothesis, we induced MI in mice and rats via left coronary artery ligation and then treated animals with every other day intraperitoneal PR1P or scrambled peptide for 14 days. Hemodynamic monitoring and echocardiography in mice and echocardiography in rats at 14 days showed PR1P significantly improved multiple functional markers of heart function, including stroke volume and cardiac output. Furthermore, molecular biology and histological analyses of tissue samples showed that systemic PR1P targeted, stabilized and upregulated endogenous VEGF within ischemic myocardium. We conclude that PR1P is a potential non-invasive candidate therapeutic for MI.


Subject(s)
AC133 Antigen/metabolism , Disease Models, Animal , Ischemia/complications , Myocardial Infarction/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Female , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Ventricular Function, Left/drug effects
4.
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
5.
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
6.
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
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.
Front Pharmacol ; 11: 276, 2020.
Article in English | MEDLINE | ID: mdl-32210827

ABSTRACT

Pathologic ocular neovascularization commonly results in visual impairment or even blindness in numerous fundus diseases, including proliferative diabetic retinopathy (PDR), retinopathy of prematurity (ROP), and age-related macular degeneration (AMD). MicroRNAs regulate angiogenesis through modulating target genes and disease progression, making them a new class of targets for drug discovery. In this study, we investigated the potential role of miR-18a-5p in retinal neovascularization using a mouse model of oxygen-induced proliferative retinopathy (OIR). We found that miR-18a-5p was highly expressed in the retina of pups as well as retinal endothelial cells, and was consistently down-regulated during retinal development. On the other hand, miR-18a-5p was increased significantly during pathologic neovascularization in the retinas of OIR mice. Moreover, intravitreal administration of miRNA mimic, agomiR-18a-5p, significantly suppressed retinal neovascularization in OIR models. Accordingly, agomir-18a-5p markedly suppressed human retinal microvascular endothelial cell (HRMEC) function including proliferation, migration, and tube formation ability. Additionally, we demonstrated that miR-18a-5p directly down-regulated known vascular growth factors, fibroblast growth factor 1 (FGF1) and hypoxia-inducible factor 1-alpha (HIF1A), as the target genes. In conclusion, miR-18a-5p may be a useful drug target for pathologic ocular neovascularization.

9.
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
10.
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
11.
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
12.
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
13.
Nanomedicine ; 11(8): 1993-2002, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26247432

ABSTRACT

Oral delivery of poorly soluble and permeable drugs represents a significant challenge in drug development. The oral delivery of drugs remains to be the ultimate route of any drugs. However, in many cases, drugs are not absorbed well in the gastrointestinal tract, or they lose their activity. Polymer micelles were recognized as an effective carrier system for drug encapsulation, and are now studied as a vehicle for oral delivery of insoluble compounds. We characterized the properties of monomethoxy polyethylene glycol-poly lactic acid (mPEG-PLA) micelles, and visualized their internalization in mouse small intestine. Using Caco-2 cells as a cellular model, we studied the kinetics of particle uptake, their transport, and the molecular mechanism of their intestinal absorption. Moreover, by inhibiting specific endocytosis pathways, pharmacologically and genetically, we found that mPEG-PLA nanoparticle endocytosis is mediated by clathrin in an energy-dependent manner, and that the low-density lipoprotein receptor is involved. FROM THE CLINICAL EDITOR: Many current drugs used are non-water soluble and indeed, the ability to deliver these drugs via the gastrointestinal tract remains the holy grail for many researchers. The authors in this paper developed monomethoxy polyethylene glycol-poly lactic acid (mPEG-PLA) micelles as a drug nanocarrier, and studied the mechanism of uptake across intestinal cells. The findings should improve our current understanding and point to the development of more nanocarriers.


Subject(s)
Drug Carriers/pharmacokinetics , Intestine, Small/metabolism , Lactic Acid/pharmacokinetics , Micelles , Polyethylene Glycols/pharmacokinetics , Polymers/pharmacokinetics , Administration, Oral , Animals , Caco-2 Cells , Drug Carriers/chemistry , Endocytosis , Humans , Intestinal Absorption , Lactic Acid/chemistry , Mice, Inbred C57BL , Polyesters , Polyethylene Glycols/chemistry , Polymers/chemistry
14.
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
15.
PLoS One ; 9(9): e106507, 2014.
Article in English | MEDLINE | ID: mdl-25188381

ABSTRACT

BACKGROUND: Vision loss due to vascular disease of the retina is a leading cause of blindness in the world. Retinal angiomatous proliferation (RAP) is a subgroup of neovascular age-related macular degeneration (AMD), whereby abnormal blood vessels develop in the retina leading to debilitating vision loss and eventual blindness. The novel mouse strain, neoretinal vascularization 2 (NRV2), shows spontaneous fundus changes associated with abnormal neovascularization. The purpose of this study is to characterize the induction of pathologic angiogenesis in this mouse model. METHODS: The NRV2 mice were examined from postnatal day 12 (p12) to 3 months. The phenotypic changes within the retina were evaluated by fundus photography, fluorescein angiography, optical coherence tomography, and immunohistochemical and electron microscopic analysis. The pathological neovascularization was imaged by confocal microscopy and reconstructed using three-dimensional image analysis software. RESULTS: We found that NRV2 mice develop multifocal retinal depigmentation in the posterior fundus. Depigmented lesions developed vascular leakage observed by fluorescein angiography. The spontaneous angiogenesis arose from the retinal vascular plexus at postnatal day (p)15 and extended toward retinal pigment epithelium (RPE). By three months of age, histological analysis revealed encapsulation of the neovascular lesion by the RPE in the photoreceptor cell layer and subretinal space. CONCLUSIONS: The NRV2 mouse strain develops early neovascular lesions within the retina, which grow downward towards the RPE beginning at p15. This retinal neovascularization model mimics early stages of human retinal angiomatous proliferation (RAP) and will likely be a useful in elucidating targeted therapeutics for patients with ocular neovascular disease.


Subject(s)
Retinal Neovascularization/pathology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescein Angiography , Longitudinal Studies , Mice , Pigment Epithelium of Eye/pathology , Retina/pathology , Tomography, Optical Coherence
16.
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
17.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...