Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Am Soc Nephrol ; 33(6): 1120-1136, 2022 06.
Article in English | MEDLINE | ID: mdl-35292439

ABSTRACT

BACKGROUND: Glomerular endothelial cell (GEnC) fenestrations are recognized as an essential component of the glomerular filtration barrier, yet little is known about how they are regulated and their role in disease. METHODS: We comprehensively characterized GEnC fenestral and functional renal filtration changes including measurement of glomerular Kf and GFR in diabetic mice (BTBR ob-/ob- ). We also examined and compared human samples. We evaluated Eps homology domain protein-3 (EHD3) and its association with GEnC fenestrations in diabetes in disease samples and further explored its role as a potential regulator of fenestrations in an in vitro model of fenestration formation using b.End5 cells. RESULTS: Loss of GEnC fenestration density was associated with decreased filtration function in diabetic nephropathy. We identified increased diaphragmed fenestrations in diabetes, which are posited to increase resistance to filtration and further contribute to decreased GFR. We identified decreased glomerular EHD3 expression in diabetes, which was significantly correlated with decreased fenestration density. Reduced fenestrations in EHD3 knockdown b.End5 cells in vitro further suggested a mechanistic role for EHD3 in fenestration formation. CONCLUSIONS: This study demonstrates the critical role of GEnC fenestrations in renal filtration function and suggests EHD3 may be a key regulator, loss of which may contribute to declining glomerular filtration function through aberrant GEnC fenestration regulation. This points to EHD3 as a novel therapeutic target to restore filtration function in disease.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Urinary Tract Physiological Phenomena , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Endothelial Cells/metabolism , Kidney Glomerulus/metabolism , Mice
2.
Dev Biol ; 464(2): 137-144, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32565279

ABSTRACT

Tissue macrophages, which are ubiquitously present innate immune cells, play versatile roles in development and organogenesis. During development, macrophages prune transient or unnecessary synapses in neuronal development, and prune blood vessels in vascular development, facilitating appropriate tissue remodeling. In the present study, we identified that macrophages contributed to the development of pupillary morphology. Csf1op/op mutant mice, in which ocular macrophages are nearly absent, exhibited abnormal pupillary edges, with abnormal protrusions of excess iris tissue into the pupillary space. Macrophages located near the pupillary edge engulfed pigmented debris, which likely consisted of unnecessary iris protrusions that emerge during smoothening of the pupillary edge. Indeed, pupillary edge macrophages phenotypically possessed some features of M2 macrophages, consistent with robust tissue engulfment and remodeling activities. Interestingly, protruding irises in Csf1op/op mice were only detected in gaps between regressing blood vessels. Taken together, our findings uncovered a new role for ocular macrophages, demonstrating that this cell population is important for iris pruning during development.


Subject(s)
Macrophages/metabolism , Pupil , Animals , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Mice , Mice, Mutant Strains
3.
Cells ; 9(6)2020 06 03.
Article in English | MEDLINE | ID: mdl-32503129

ABSTRACT

Fenestrae are transcellular plasma membrane pores that mediate blood-tissue exchange in specialised vascular endothelia. The composition and biogenesis of the fenestra remain enigmatic. We isolated and characterised the protein composition of large patches of fenestrated plasma membrane, termed sieve plates. Loss-of-function experiments demonstrated that two components of the sieve plate, moesin and annexin II, were positive and negative regulators of fenestra formation, respectively. Biochemical analyses showed that moesin is involved in the formation of an actin-fodrin submembrane cytoskeleton that was essential for fenestra formation. The link between the fodrin cytoskeleton and the plasma membrane involved the fenestral pore protein PV-1 and Na,K-ATPase, which is a key regulator of signalling during fenestra formation both in vitro and in vivo. These findings provide a conceptual framework for fenestra biogenesis, linking the dynamic changes in plasma membrane remodelling to the formation of a submembrane cytoskeletal signalling complex.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Microfilament Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Actin Cytoskeleton/metabolism , Animals , Annexin A2/metabolism , Cell Line , Cell Membrane/ultrastructure , Cytoskeleton/ultrastructure , Endothelial Cells/ultrastructure , Male , Mice , Ouabain/pharmacology , Rats, Sprague-Dawley
4.
Circ Res ; 126(6): 767-783, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32078435

ABSTRACT

RATIONALE: Central nervous system has low vascular permeability by organizing tight junction (TJ) and limiting endothelial transcytosis. While TJ has long been considered to be responsible for vascular barrier in central nervous system, suppressed transcytosis in endothelial cells is now emerging as a complementary mechanism. Whether transcytosis regulation is independent of TJ and its dysregulation dominantly causes diseases associated with edema remain elusive. Dll4 signaling is important for various vascular contexts, but its role in the maintenance of vascular barrier in central nervous system remains unknown. OBJECTIVE: To find a TJ-independent regulatory mechanism selective for transcytosis and identify its dysregulation as a cause of pathological leakage. METHODS AND RESULTS: We studied transcytosis in the adult mouse retina with low vascular permeability and employed a hypertension-induced retinal edema model for its pathological implication. Both antibody-based and genetic inactivation of Dll4 or Notch1 induce hyperpermeability by increasing transcytosis without junctional destabilization in arterial endothelial cells, leading to nonhemorrhagic leakage predominantly in the superficial retinal layer. Endothelial Sox17 deletion represses Dll4 in retinal arteries, phenocopying Dll4 blocking-driven vascular leakage. Ang II (angiotensin II)-induced hypertension represses arterial Sox17 and Dll4, followed by transcytosis-driven retinal edema, which is rescued by a gain of Notch activity. Transcriptomic profiling of retinal endothelial cells suggests that Dll4 blocking activates SREBP1 (sterol regulatory element-binding protein 1)-mediated lipogenic transcription and enriches gene sets favorable for caveolae formation. Profiling also predicts the activation of VEGF (vascular endothelial growth factor) signaling by Dll4 blockade. Inhibition of SREBP1 or VEGF-VEGFR2 (VEGF receptor 2) signaling attenuates both Dll4 blockade-driven and hypertension-induced retinal leakage. CONCLUSIONS: In the retina, Sox17-Dll4-SREBP1 signaling axis controls transcytosis independently of TJ in superficial arteries among heterogeneous regulations for the whole vessels. Uncontrolled transcytosis via dysregulated Dll4 underlies pathological leakage in hypertensive retina and could be a therapeutic target for treating hypertension-associated retinal edema.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blood-Retinal Barrier/metabolism , Calcium-Binding Proteins/metabolism , Hypertensive Retinopathy/metabolism , Transcytosis , Adaptor Proteins, Signal Transducing/genetics , Animals , Arteries/metabolism , Calcium-Binding Proteins/genetics , Caveolae/metabolism , Endothelial Cells/metabolism , HMGB Proteins/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , SOXF Transcription Factors/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Diabetologia ; 61(11): 2422-2432, 2018 11.
Article in English | MEDLINE | ID: mdl-30094465

ABSTRACT

AIMS/HYPOTHESIS: Diabetic retinopathy is increasing in prevalence worldwide and is fast becoming a global epidemic and a leading cause of visual loss. Current therapies are limited, and the development of effective treatments for diabetic retinopathy requires a greater in-depth knowledge of disease progression and suitable modelling of diabetic retinopathy in animals. The aim of this study was to assess the early pathological changes in retinal morphology and neuronal, inflammatory and vascular features consistent with diabetic retinopathy in the ob/ob mouse model of type 2 diabetes, to investigate whether features similar to those in human diabetic retinopathy were present. METHODS: Male and female wild-type (+/+), heterozygous (+/-) and homozygous (-/-) BTBR ob/ob mice were examined at 6, 10, 15 and 20 weeks of age. Animals were weighed and blood glucose was measured. TUNEL and brain-specific homeobox/POU domain protein 3A (BRN3A) markers were used to examine retinal ganglion cells. We used immunostaining (collagen IV and platelet endothelial cell adhesion molecule [PECAM]/CD31) to reveal retinal vessel degeneration. Spectral domain optical coherence tomography was used to reveal changes in the thickness and structure of the retinal layer. Vitreous fluorophotometry was used to investigate vascular permeability. A-waves, b-waves and oscillatory potentials were measured under photopic and scotopic conditions. Concanavalin A leucostasis and immunostaining with glial fibrillary acidic protein (GFAP) and ionised calcium-binding adapter molecule 1 (IBA-1) identified differences in inflammatory status. Paraffin sections and transmission electron microscopy were used to reveal changes in the thickness and structure of the retinal layer. RESULTS: Following the development of obesity and hyperglycaemia in 2-week-old and 3-week-old ob-/ob- mice, respectively (p < 0.001), early functional deficits (p < 0.001) and thinning of the inner retina (p < 0.001) were identified. Glial activation, leucostasis (p < 0.05) and a shift in microglia/macrophage phenotype were observed before microvascular degeneration (p < 0.05) and elevated vascular permeability occurred (p < 0.05). CONCLUSIONS/INTERPRETATION: The present characterisation of the development of diabetic retinopathy in the ob/ob mouse represents a platform that will enable the development of new therapies, particularly for the early stages of disease.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Retina/metabolism , Retina/pathology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Female , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Obesity/metabolism , Obesity/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology
7.
J Exp Med ; 215(3): 963-983, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29444818

ABSTRACT

High-grade glioma (HGG) is highly angiogenic, but antiangiogenic therapy has transient clinical benefit in only a fraction of patients. Vascular regulators of these heterogeneous responses remain undetermined. We found up-regulation of Sox7 and down-regulation of Sox17 in tumor endothelial cells (tECs) in mouse HGG. Sox7 deletion suppressed VEGFR2 expression, vascular abnormality, hypoxia-driven invasion, regulatory T cell infiltration, and tumor growth. Conversely, Sox17 deletion exacerbated these phenotypes by up-regulating Sox7 in tECs. Anti-VEGFR2 antibody treatment delayed tumor growth by normalizing Sox17-deficient abnormal vessels with high Sox7 levels but promoted it by regressing Sox7-deficient vessels, recapitulating variable therapeutic responses to antiangiogenic therapy in HGG patients. Our findings establish that Sox7 promotes tumor growth via vessel abnormalization, and its level determines the therapeutic outcome of VEGFR2 inhibition in HGG. In 189 HGG patients, Sox7 expression was heterogeneous in tumor vessels, and high Sox7 levels correlated with poor survival, early recurrence, and impaired vascular function, emphasizing the clinical relevance of Sox7 in HGG.


Subject(s)
Blood Vessels/abnormalities , Glioma/metabolism , Glioma/pathology , SOXF Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Calcium-Binding Proteins , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Deletion , Glioma/blood supply , Glioma/immunology , Humans , Immunity , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Neoplasm Grading , Prognosis , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
9.
EMBO Mol Med ; 8(11): 1265-1288, 2016 11.
Article in English | MEDLINE | ID: mdl-27742718

ABSTRACT

Anti-angiogenic therapies using biological molecules that neutralize vascular endothelial growth factor-A (VEGF-A) have revolutionized treatment of retinal vascular diseases including age-related macular degeneration (AMD). This study reports preclinical assessment of a strategy to enhance anti-VEGF-A monotherapy efficacy by targeting both VEGF-A and angiopoietin-2 (ANG-2), a factor strongly upregulated in vitreous fluids of patients with retinal vascular disease and exerting some of its activities in concert with VEGF-A. Simultaneous VEGF-A and ANG-2 inhibition was found to reduce vessel lesion number, permeability, retinal edema, and neuron loss more effectively than either agent alone in a spontaneous choroidal neovascularization (CNV) model. We describe the generation of a bispecific domain-exchanged (crossed) monoclonal antibody (CrossMAb; RG7716) capable of binding, neutralizing, and depleting VEGF-A and ANG-2. RG7716 showed greater efficacy than anti-VEGF-A alone in a non-human primate laser-induced CNV model after intravitreal delivery. Modification of RG7716's FcRn and FcγR binding sites disabled the antibodies' Fc-mediated effector functions. This resulted in increased systemic, but not ocular, clearance. These properties make RG7716 a potential next-generation therapy for neovascular indications of the eye.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Antibodies, Monoclonal/administration & dosage , Eye Diseases/drug therapy , Immunologic Factors/administration & dosage , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Eye Diseases/pathology , Immunologic Factors/isolation & purification , Immunologic Factors/pharmacology , Macaca fascicularis , Treatment Outcome
10.
Am J Pathol ; 185(9): 2534-49, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26188133

ABSTRACT

Choroidal neovascularization (CNV) is a defining feature of wet age-related macular degeneration. We examined the functional role of CCR3 in the development of CNV in mice and primates. CCR3 was associated with spontaneous CNV lesions in the newly described JR5558 mice, whereas CCR3 ligands localized to CNV-associated macrophages and the retinal pigment epithelium/choroid complex. Intravitreal injection of neutralizing antibodies against vascular endothelial growth factor receptor 2, CCR3, CC chemokine ligand 11/eotaxin-1, and CC chemokine ligand 24/eotaxin-2 all reduced CNV area and lesion number in these mice. Systemic administration of the CCR3 antagonists GW766994X and GW782415X reduced spontaneous CNV in JR5558 mice and laser-induced CNV in mouse and primate models in a dose-dependent fashion. Combination treatment with antivascular endothelial growth factor receptor 2 antibody and GW766994X yielded additive reductions in CNV area and hyperpermeability in mice. Interestingly, topical GW766994X and intravitreal anti-CCR3 antibody yielded strong systemic effects, reducing CNV in the untreated, contralateral eye. Contrarily, ocular administration of GW782415X in primates failed to substantially elevate plasma drug levels or to reduce the development of grade IV CNV lesions. These findings suggest that CCR3 signaling may be an attractive therapeutic target for CNV, utilizing a pathway that is at least partly distinct from that of vascular endothelial growth factor receptor. The findings also demonstrate that systemic exposure to CCR3 antagonists may be crucial for CNV-targeted activity.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Capillary Permeability/drug effects , Choroidal Neovascularization/drug therapy , Receptors, CCR3/antagonists & inhibitors , Wet Macular Degeneration/drug therapy , Animals , Capillary Permeability/immunology , Choroid/pathology , Choroidal Neovascularization/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/metabolism , Wet Macular Degeneration/pathology
11.
Dev Cell ; 30(5): 541-52, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25175707

ABSTRACT

The vascular endothelium operates in a highly polarized environment, but to date there has been little exploration of apicobasal polarization of its signaling. We show that VEGF-A, histamine, IGFBP3, and LPA trigger unequal endothelial responses when acting from the circulation or the parenchymal side at blood-neural barriers. For VEGF-A, highly polarized receptor distribution contributed to distinct signaling patterns: VEGFR2, which was found to be predominantly abluminal, mediated increased permeability via p38; in contrast, luminal VEGFR1 led to Akt activation and facilitated cytoprotection. Importantly, such differential apicobasal signaling and VEGFR distribution were found in the microvasculature of brain and retina but not lung, indicating that endothelial cells at blood-neural barriers possess specialized signaling compartments that assign different functions depending on whether an agonist is tissue or blood borne.


Subject(s)
Blood-Brain Barrier/physiology , Neurons/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Endothelium, Vascular/metabolism , Male , Mice , Mice, Inbred C57BL , Microcirculation , Permeability , Rats , Rats, Inbred Lew , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Invest Ophthalmol Vis Sci ; 55(6): 3709-19, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24845632

ABSTRACT

PURPOSE: Characterization of a mouse model of spontaneous choroidal neovascularization (sCNV) and its effect on retinal architecture and function. METHODS: The sCNV mouse phenotype was characterized by using fundus photography, fluorescein angiography, confocal scanning laser ophthalmoscopy (SLO), optical coherence tomography (OCT), ERG, immunostaining, biochemistry, and electron microscopy. A role for VEGF-A signaling in sCNV was investigated by using neutralizing antibodies and a role for macrophages explored by cell-depletion studies. RESULTS: The sCNV starts between postnatal day 10 and 15 (P10-P15), increasing in number and severity causing RPE disruption and dysfunction. Various morphological methods confirmed the choroidal origin and subretinal position of the angiogenic vessels. At approximately P25, vessels were present in the outer retina with instances of anastomosis of some sCNV lesions with the retinal vasculature. The number of CNV lesions was significantly decreased by systemic blockade of the VEGF-A pathway. Choroidal neovascularization size also was significantly modulated by reducing the number of lesion-associated macrophages. Later stages of sCNV were associated with edema, neuronal loss, and dysfunction. CONCLUSIONS: The sCNV mouse is a new model for the study of both early and late events associated with choroidal neovascularization. Pharmacological reduction in sCNV with VEGF-A antagonists and an anti-inflammatory strategy suggests the model may be useful for investigating novel targets for treating human ocular neovascular disease.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Edema/metabolism , Retinal Pigment Epithelium/ultrastructure , Vascular Endothelial Growth Factor A/metabolism , Animals , Choroid/ultrastructure , Choroidal Neovascularization/pathology , Disease Models, Animal , Edema/pathology , Electroretinography , Enzyme-Linked Immunosorbent Assay , Fluorescein Angiography , Fundus Oculi , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Confocal , Microscopy, Electron , Ophthalmoscopy , Phenotype , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiopathology , Retinal Vessels/metabolism , Retinal Vessels/ultrastructure , Signal Transduction , Tomography, Optical Coherence
13.
PLoS One ; 8(6): e67263, 2013.
Article in English | MEDLINE | ID: mdl-23840644

ABSTRACT

AIMS: With ageing extracellular material is deposited in Bruch's membrane, as drusen. Lipofuscin is deposited in retinal pigment epithelial cells. Both of these changes are associated with age related macular degeneration, a disease now believed to involve chronic inflammation at the retinal-choroidal interface. We hypothesise that these molecules may act as danger signals, causing the production of inflammatory chemokines and cytokines by the retinal pigment epithelium, via activation of pattern recognition receptors. METHODS: ARPE-19 cells were stimulated in vitro with the following reported components of drusen: amyloid-ß (1-42), Carboxyethylpyrrole (CEP) modified proteins (CEP-HSA), Nε-(Carboxymethyl)lysine (CML) modified proteins and aggregated vitronectin. The cells were also stimulated with the major fluorophore of lipofuscin: N-retinylidene-N-retinylethanolamine (A2E). Inflammatory chemokine and cytokine production was assessed using Multiplex assays and ELISA. The mechanistic evaluation of the NLRP3 inflammasome pathway was assessed in a stepwise fashion. RESULTS: Of all the molecules tested only A2E induced inflammatory chemokine and cytokine production. 25 µM A2E induced the production of significantly increased levels of the chemokines IL-8, MCP-1, MCG and MIP-1α, the cytokines IL-1ß, IL-2, IL-6, and TNF-α, and the protein VEGF-A. The release of IL-1ß was studied further, and was determined to be due to NLRP3 inflammasome activation. The pathway of activation involved endocytosis of A2E, and the three inflammasome components NLRP3, ASC and activated caspase-1. Immunohistochemical staining of ABCA4 knockout mice, which show progressive accumulation of A2E levels with age, showed increased amounts of IL-1ß proximal to the retinal pigment epithelium. CONCLUSIONS: A2E has the ability to stimulate inflammatory chemokine and cytokine production by RPE cells. The pattern recognition receptor NLRP3 is involved in this process. This provides further evidence for the link between A2E, inflammation, and the pathogenesis of AMD. It also supports the recent discovery of NLRP3 inflammasome activation in AMD.


Subject(s)
Carrier Proteins/metabolism , Epithelial Cells/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Retinoids/physiology , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Animals , Carrier Proteins/genetics , Cell Line , Cytoplasm/metabolism , Endocytosis , Epithelial Cells/drug effects , Epithelial Cells/immunology , Gene Knockdown Techniques , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Mice , Mice, 129 Strain , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Precursors/biosynthesis , Protein Precursors/genetics , Protein Transport , RNA, Small Interfering/genetics , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction , Up-Regulation
14.
Invest Ophthalmol Vis Sci ; 54(2): 1490-500, 2013 Feb 27.
Article in English | MEDLINE | ID: mdl-23385800

ABSTRACT

PURPOSE: Two noninvasive delivery strategies for VEGF/PDGF receptor tyrosine kinase inhibitors (RTKI) were explored that exploited uveal retention as a means for establishing an ocular drug depot: a single oral "loading" dose and topical administration. METHODS: Melanin binding was confirmed by centrifugation and mass spectrometry. Ocular retention was examined in pigmented and albino rats. Ocular release kinetics were measured 3 to 28 days postdosing in pigmented rats. Microautoradiography was used to demonstrate retention of RTKI in the uveal tract. A uveal drug depot of pazopanib was created by a single oral dose prior to induction of laser choroidal neovascularization (CNV). Choroid/retinal pigmented epithelium (RPE) retention of a related RTKI with enhanced topical bioavailability, GW771806, was confirmed by bioanalytics, and its ability to regress CNV compared with pazopanib. RESULTS: Pazopanib and GW771806 directly bound melanin and were retained within the uveal tract of pigmented rats for weeks following a single oral dose. Pazopanib was undetectable systemically following a single oral administration prior to CNV induction, and reduced CNV as well as twice daily dosing. Topical ocular delivery of GW771806 at 5 mg/mL led to high choroidal/RPE exposure and significantly regressed CNV lesions; 2 mg/mL prevented lesion progression. CONCLUSIONS: Uveal retention of drugs such as pazopanib can be used to create a sustained-release depot. Topical GW771806 regressed CNV. These data indicate that topical or infrequent oral loading dose treatment with VEGF/PDGF RTKI retained in the choroid/RPE might allow noninvasive treatments for ocular neovascular disease.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Choroidal Neovascularization/drug therapy , Drug Delivery Systems , Indazoles/administration & dosage , Pyrimidines/administration & dosage , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Sulfonamides/administration & dosage , Sulfones/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Administration, Oral , Administration, Topical , Angiogenesis Inhibitors/pharmacokinetics , Animals , Autoradiography , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/metabolism , Female , Fluorescein Angiography , Half-Life , Indazoles/pharmacokinetics , Melanins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pyrimidines/pharmacokinetics , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , Sulfones/pharmacokinetics , Uvea/metabolism
15.
Am J Pathol ; 182(4): 1379-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416159

ABSTRACT

Vascular endothelial growth factor A (VEGF-A) is a validated therapeutic target in several angiogenic- and vascular permeability-related pathological conditions, including certain cancers and potentially blinding diseases, such as age-related macular degeneration and diabetic retinopathy. We and others have shown that VEGF-A also plays an important role in neuronal development and neuroprotection, including in the neural retina. Antagonism of VEGF-A function might therefore present a risk to neuronal survival as a significant adverse effect. Herein, we demonstrate that VEGF-A acts directly on retinal ganglion cells (RGCs) to promote survival. VEGF receptor-2 signaling via the phosphoinositide-3-kinase/Akt pathway was required for the survival response in isolated RGCs. These results were confirmed in animal models of staurosporine-induced RGC death and experimental hypertensive glaucoma. Importantly, we observed that VEGF-A blockade significantly exacerbated neuronal cell death in the hypertensive glaucoma model. Our findings highlight the need to better define the risks associated with use of VEGF-A antagonists in the ocular setting.


Subject(s)
Glaucoma/drug therapy , Glaucoma/pathology , Neuroprotective Agents/therapeutic use , Retina/pathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Death/drug effects , Cells, Cultured , Cytoprotection/drug effects , Disease Models, Animal , Glaucoma/enzymology , Neuropilins/metabolism , Neuroprotective Agents/pharmacology , Neutralization Tests , Ocular Hypertension/drug therapy , Ocular Hypertension/enzymology , Ocular Hypertension/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/enzymology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Signal Transduction/drug effects , Toxicity Tests, Acute , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
PLoS One ; 7(5): e35231, 2012.
Article in English | MEDLINE | ID: mdl-22567098

ABSTRACT

Splicing of the human vascular endothelial growth factor-A (VEGF-A) gene has been reported to generate angiogenic (VEGFxxx) and anti-angiogenic (VEGFxxxb) isoforms. Corresponding VEGFxxxb isoforms have also been reported in rat and mouse. We examined VEGFxxxb expression in mouse fibrosarcoma cell lines expressing all or individual VEGF isoforms (VEGF120, 164 or 188), grown in vitro and in vivo, and compared results with those from normal mouse and human tissues. Importantly, genetic construction of VEGF164 and VEGF188 expressing fibrosarcomas, in which exon 7 is fused to the conventional exon 8, precludes VEGFxxxb splicing from occurring. Thus, these two fibrosarcoma cell lines provided endogenous negative controls. Using RT-PCR we show that primers designed to simultaneously amplify VEGFxxx and VEGFxxxb isoforms amplified only VEGFxxx variants in both species. Moreover, only VEGFxxx species were generated when mouse podocytes were treated with TGFß-1, a reported activator of VEGFxxxb splice selection in human podocytes. A VEGF164/120 heteroduplex species was identified as a PCR artefact, specifically in mouse. VEGFxxxb isoform-specific PCR did amplify putative VEGFxxxb species in mouse and human tissues, but unexpectedly also in VEGF188 and VEGF164 fibrosarcoma cells and tumours, where splicing to produce true VEGFxxxb isoforms cannot occur. Moreover, these products were only consistently generated using reverse primers spanning more than 5 bases across the 8b/7 or 8b/5 splice junctions. Primer annealing to VEGFxxx transcripts and amplification of exon 8b primer 'tails' explained the artefactual generation of VEGFxxxb products, since the same products were generated when the PCR reactions were performed with cDNA from VEGF164/VEGF188 'knock-in' vectors used in the generation of single VEGF isoform-expressing transgenic mice from which the fibrosarcoma lines were developed. Collectively, our results highlight important pitfalls in data interpretation associated with detecting VEGFxxxb isoforms using current methods, and demonstrate that anti-angiogenic isoforms are not commonly expressed in mouse or human tissues.


Subject(s)
Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Exons/genetics , Humans , In Vitro Techniques , Male , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
17.
Proc Natl Acad Sci U S A ; 109(9): E553-62, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22323600

ABSTRACT

The generation of diverse neuronal types and subtypes from multipotent progenitors during development is crucial for assembling functional neural circuits in the adult central nervous system. During mouse retinogenesis, early retinal progenitors give rise to several cell types, including ganglion, amacrine, horizontal, cone, and rod cells. It is unknown at present how each of these fates is selected from the multiple neuronal fates available to the early progenitor. By using a combination of bioinformatic, genetic, and biochemical approaches, we investigated the mechanism by which Foxn4 selects the amacrine and horizontal cell fates from multipotential retinal progenitors. These studies indicate that Foxn4 has an intrinsic activity to suppress the alternative photoreceptor cell fates of early retinal progenitors by selectively activating Dll4-Notch signaling. Gene expression and conditional ablation analyses reveal that Dll4 is directly activated by Foxn4 via phylogenetically conserved enhancers and that Dll4 can partly mediate the Foxn4 function by serving as a major Notch ligand to expand the progenitor pool and limit photoreceptor production. Our data together define a Foxn4-mediated molecular and signaling pathway that underlies the suppression of alternative cell fates of early retinal progenitors.


Subject(s)
Eye Proteins/physiology , Forkhead Transcription Factors/physiology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Photoreceptor Cells, Vertebrate/cytology , Receptors, Notch/physiology , Adaptor Proteins, Signal Transducing , Amacrine Cells/cytology , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Lineage , Conserved Sequence , Enhancer Elements, Genetic , Eye Proteins/genetics , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Recombinant Fusion Proteins/physiology , Retina/embryology , Retina/growth & development , Retinal Rod Photoreceptor Cells/cytology , Signal Transduction/physiology
18.
Drug Discov Today ; 15(7-8): 272-82, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20184967

ABSTRACT

Angiogenic diseases of the retina are the leading cause of blindness in the developed world. The development of anti-angiogenic molecular therapies has transformed the prognosis of these conditions, especially age-related macular degeneration. With these new treatments comes the new challenge of delivering an effective dosage to the retina, over a prolonged period of time and in a safe and cost-effective manner. A range of new anti-angiogenics are on the horizon, offering new and varied modes of drug delivery. In addition, a range of new sustained-release drug delivery technologies are being developed.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Retinal Diseases/drug therapy , Absorbable Implants , Animals , Drug Delivery Systems , Drug Implants , Genetic Therapy , Humans , Injections , Vitreous Body
19.
Biochem Soc Trans ; 37(Pt 6): 1201-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19909247

ABSTRACT

The longer splice isoforms of VEGF (vascular endothelial growth factor)-A, including VEGF(164(165)), contain a highly basic HBD (heparin-binding domain). This domain allows these isoforms to interact with and localize to the HS (heparan sulfate)-rich extracellular matrix, and bind to the co-receptor Nrp-1 (neuropilin-1). Heparin-binding VEGF-A isoforms are critical for survival: mice engineered to express exclusively the non-heparin-binding VEGF(120) have diminished vascular branching during embryonic development and die from postnatal angiogenesis defects shortly after birth. Although it is thought that the HBD contributes to the diverse functions of VEGF-A in both physiological and pathological processes, little is known about the molecular features within this domain that enable these functions. In the present paper, we discuss the roles of the VEGF HBD in normal and disease conditions, with a particular focus on the VEGF(164(165)) isoform.


Subject(s)
Heparin/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism , Alternative Splicing , Animals , Eye Diseases/metabolism , Eye Diseases/pathology , Heparan Sulfate Proteoglycans/metabolism , Humans , Mice , Models, Molecular , Neoplasms/metabolism , Neovascularization, Pathologic , Protein Binding , Protein Conformation , Protein Isoforms/genetics , Protein Structure, Tertiary , Vascular Endothelial Growth Factor A/genetics
20.
J Exp Med ; 205(11): 2507-13, 2008 Oct 27.
Article in English | MEDLINE | ID: mdl-18824583

ABSTRACT

The thymic microenvironment is required for T cell development in vivo. However, in vitro studies have shown that when hematopoietic progenitors acquire Notch signaling via Delta-like (Dll)1 or Dll4, they differentiate into the T cell lineage in the absence of a thymic microenvironment. It is not clear, however, whether the thymus supports T cell development specifically by providing Notch signaling. To address this issue, we generated mice with a loxP-flanked allele of Dll4 and induced gene deletion specifically in thymic epithelial cells (TECs). In the thymus of mutant mice, the expression of Dll4 was abrogated on the epithelium, and the proportion of hematopoietic cells bearing the intracellular fragment of Notch1 (ICN1) was markedly decreased. Corresponding to this, CD4 CD8 double-positive or single-positive T cells were not detected in the thymus. Further analysis showed that the double-negative cell fraction was lacking T cell progenitors. The enforced expression of ICN1 in hematopoietic progenitors restored thymic T cell differentiation, even when the TECs were deficient in Dll4. These results indicate that the thymus-specific environment for determining T cell fate indispensably requires Dll4 expression to induce Notch signaling in the thymic immigrant cells.


Subject(s)
Cell Differentiation/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Gene Deletion , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Receptors, Notch/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...