Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Ophthalmol Sci ; 4(3): 100439, 2024.
Article in English | MEDLINE | ID: mdl-38361912

ABSTRACT

Purpose: The murine oxygen-induced retinopathy (OIR) model is one of the most widely used animal models of ischemic retinopathy, mimicking hallmark pathophysiology of initial vaso-obliteration (VO) resulting in ischemia that drives neovascularization (NV). In addition to NV and VO, human ischemic retinopathies, including retinopathy of prematurity (ROP), are characterized by increased vascular tortuosity. Vascular tortuosity is an indicator of disease severity, need to treat, and treatment response in ROP. Current literature investigating novel therapeutics in the OIR model often report their effects on NV and VO, and measurements of vascular tortuosity are less commonly performed. No standardized quantification of vascular tortuosity exists to date despite this metric's relevance to human disease. This proof-of-concept study aimed to apply a previously published semi-automated computer-based image analysis approach (iROP-Assist) to develop a new tool to quantify vascular tortuosity in mouse models. Design: Experimental study. Subjects: C57BL/6J mice subjected to the OIR model. Methods: In a pilot study, vasculature was manually segmented on flat-mount images of OIR and normoxic (NOX) mice retinas and segmentations were analyzed with iROP-Assist to quantify vascular tortuosity metrics. In a large cohort of age-matched (postnatal day 12 [P12], P17, P25) NOX and OIR mice retinas, NV, VO, and vascular tortuosity were quantified and compared. In a third experiment, vascular tortuosity in OIR mice retinas was quantified on P17 following intravitreal injection with anti-VEGF (aflibercept) or Immunoglobulin G isotype control on P12. Main Outcome Measures: Vascular tortuosity. Results: Cumulative tortuosity index was the best metric produced by iROP-Assist for discriminating between OIR mice and NOX controls. Increased vascular tortuosity correlated with disease activity in OIR. Treatment of OIR mice with aflibercept rescued vascular tortuosity. Conclusions: Vascular tortuosity is a quantifiable feature of the OIR model that correlates with disease severity and may be quickly and accurately quantified using the iROP-Assist algorithm. Financial Disclosures: Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.

2.
Ophthalmol Sci ; 4(1): 100338, 2024.
Article in English | MEDLINE | ID: mdl-37869029

ABSTRACT

Objective: To develop a generative adversarial network (GAN) to segment major blood vessels from retinal flat-mount images from oxygen-induced retinopathy (OIR) and demonstrate the utility of these GAN-generated vessel segmentations in quantifying vascular tortuosity. Design: Development and validation of GAN. Subjects: Three datasets containing 1084, 50, and 20 flat-mount mice retina images with various stains used and ages at sacrifice acquired from previously published manuscripts. Methods: Four graders manually segmented major blood vessels from flat-mount images of retinas from OIR mice. Pix2Pix, a high-resolution GAN, was trained on 984 pairs of raw flat-mount images and manual vessel segmentations and then tested on 100 and 50 image pairs from a held-out and external test set, respectively. GAN-generated and manual vessel segmentations were then used as an input into a previously published algorithm (iROP-Assist) to generate a vascular cumulative tortuosity index (CTI) for 20 image pairs containing mouse eyes treated with aflibercept versus control. Main Outcome Measures: Mean dice coefficients were used to compare segmentation accuracy between the GAN-generated and manually annotated segmentation maps. For the image pairs treated with aflibercept versus control, mean CTIs were also calculated for both GAN-generated and manual vessel maps. Statistical significance was evaluated using Wilcoxon signed-rank tests (P ≤ 0.05 threshold for significance). Results: The dice coefficient for the GAN-generated versus manual vessel segmentations was 0.75 ± 0.27 and 0.77 ± 0.17 for the held-out test set and external test set, respectively. The mean CTI generated from the GAN-generated and manual vessel segmentations was 1.12 ± 0.07 versus 1.03 ± 0.02 (P = 0.003) and 1.06 ± 0.04 versus 1.01 ± 0.01 (P < 0.001), respectively, for eyes treated with aflibercept versus control, demonstrating that vascular tortuosity was rescued by aflibercept when quantified by GAN-generated and manual vessel segmentations. Conclusions: GANs can be used to accurately generate vessel map segmentations from flat-mount images. These vessel maps may be used to evaluate novel metrics of vascular tortuosity in OIR, such as CTI, and have the potential to accelerate research in treatments for ischemic retinopathies. Financial Disclosures: The author(s) have no proprietary or commercial interest in any materials discussed in this article.

3.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35639473

ABSTRACT

Disruption of the neurovascular unit (NVU) underlies the pathophysiology of various CNS diseases. One strategy to repair NVU dysfunction uses stem/progenitor cells to provide trophic support to the NVU's functionally coupled and interdependent vasculature and surrounding CNS parenchyma. A subset of endothelial progenitor cells, endothelial colony-forming cells (ECFCs) with high expression of the CD44 hyaluronan receptor (CD44hi), provides such neurovasculotrophic support via a paracrine mechanism. Here, we report that bioactive extracellular vesicles from CD44hi ECFCs (EVshi) are paracrine mediators, recapitulating the effects of intact cell therapy in murine models of ischemic/neurodegenerative retinopathy; vesicles from ECFCs with low expression levels of CD44 (EVslo) were ineffective. Small RNA sequencing comparing the microRNA cargo from EVshi and EVslo identified candidate microRNAs that contribute to these effects. EVshi may be used to repair NVU dysfunction through multiple mechanisms to stabilize hypoxic vasculature, promote vascular growth, and support neural cells.


Subject(s)
Endothelial Progenitor Cells , Extracellular Vesicles , MicroRNAs , Retinal Diseases , Animals , Endothelial Progenitor Cells/metabolism , Extracellular Vesicles/metabolism , Ischemia/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Physiologic/physiology , Retinal Diseases/metabolism , Retinal Diseases/therapy
4.
Glia ; 70(9): 1762-1776, 2022 09.
Article in English | MEDLINE | ID: mdl-35611927

ABSTRACT

Retinal neovascularization (NV) is the major cause of severe visual impairment in patients with ischemic eye diseases. While it is known that retinal microglia contribute to both physiological and pathological angiogenesis, the molecular mechanisms by which these glia regulate pathological NV have not been fully elucidated. In this study, we utilized a retinal microglia-specific Transforming Growth Factor-ß (Tgfß) receptor knock out mouse model and human iPSC-derived microglia to examine the role of Tgfß signaling in activated microglia during retinal NV. Using a tamoxifen-inducible, microglia-specific Tgfß receptor type 2 (Tgfßr2) knockout mouse [Tgfßr2 KO (ΔMG)] we show that Tgfß signaling in microglia actively represses leukostasis in retinal vessels. Furthermore, we show that Tgfß signaling represses expression of the pro-angiogenic factor, Insulin-like growth factor 1 (Igf1), independent of Vegf regulation. Using the mouse model of oxygen-induced retinopathy (OIR) we show that Tgfß signaling in activated microglia plays a role in hypoxia-induced NV where a loss in Tgfß signaling microglia exacerbates and prolongs retinal NV in OIR. Using human iPSC-derived microglia cells in an in vitro assay, we validate the role of Transforming Growth Factor-ß1 (Tgfß1) in regulating Igf1 expression in hypoxic conditions. Finally, we show that Tgfß signaling in microglia is essential for microglial homeostasis and that the disruption of Tgfß signaling in microglia exacerbates retinal NV in OIR by promoting leukostasis and Igf1 expression.


Subject(s)
Leukostasis , Retinal Diseases , Retinal Neovascularization , Animals , Disease Models, Animal , Hypoxia/complications , Hypoxia/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Leukostasis/complications , Leukostasis/metabolism , Leukostasis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Neovascularization, Pathologic/metabolism , Oxygen/metabolism , Retinal Diseases/metabolism , Retinal Neovascularization/etiology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Transforming Growth Factor beta/metabolism
5.
Cytokine ; 143: 155542, 2021 07.
Article in English | MEDLINE | ID: mdl-33926775

ABSTRACT

Interferon-γ (IFNG) is one of the key cytokines that regulates both innate and adaptive immune responses in the body. However, the role of IFNG in the regulation of vascularization, especially in the context of Vascular endothelial growth factor A (VEGFa)-induced angiogenesis is not clarified. Here, we report that IFNG shows potent anti-angiogenic potential against VEGFa-induced angiogenesis. IFNG significantly inhibited proliferation, migration, and tube formation of Human umbilical vein endothelial cells (HUVECs) both under basal and VEGFa-treated conditions. Intriguingly, Knockdown (KD) of STAT1 abolished the inhibitory effect of IFNG on VEGFa-induced angiogenic processes in HUVECs. Furthermore, IFNG exhibited potent anti-angiogenic efficacy in the mouse model of oxygen-induced retinopathy (OIR), an in vivo model for hypoxia-induced retinal neovascularization, without induction of functional side effects. Taken together, these results show that IFNG plays a crucial role in the regulation of VEGFa-dependent angiogenesis, suggesting its potential therapeutic applicability in neovascular diseases.


Subject(s)
Interferon-gamma/therapeutic use , Ischemia/complications , Retinal Neovascularization/complications , Retinal Neovascularization/drug therapy , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia/complications , Interferon-gamma/administration & dosage , Interferon-gamma/pharmacology , Intravitreal Injections , Mice , Neovascularization, Physiologic/drug effects , Retina/drug effects , Retina/pathology , Retina/physiopathology , Retinal Neovascularization/physiopathology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
6.
Proc Natl Acad Sci U S A ; 117(45): 28297-28306, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33106407

ABSTRACT

Retinal neovascularization (NV), a leading cause of vision loss, results from localized hypoxia that stabilizes the hypoxia-inducible transcription factors HIF-1α and HIF-2α, enabling the expression of angiogenic factors and genes required to maintain homeostasis under conditions of oxygen stress. HIF transcriptional activity depends on the interaction between its intrinsically disordered C-terminal domain and the transcriptional coactivators CBP/p300. Much effort is currently directed at disrupting protein-protein interactions between disease-associated transcription factors like HIF and their cellular partners. The intrinsically disordered protein CITED2, a direct product of HIF-mediated transcription, functions as a hypersensitive negative regulator that attenuates the hypoxic response by competing allosterically with HIF-1α for binding to CBP/p300. Here, we show that a peptide fragment of CITED2 is taken up by retinal cells and efficiently regulates pathological angiogenesis in murine models of ischemic retinopathy. Both vaso-obliteration (VO) and NV were significantly inhibited in an oxygen-induced retinopathy (OIR) model following intravitreal injection of the CITED2 peptide. The CITED2 peptide localized to retinal neurons and glia, resulting in decreased expression of HIF target genes. Aflibercept, a commonly used anti-VEGF therapy for retinal neovascular diseases, rescued NV but not VO in OIR. However, a combination of the CITED2 peptide and a reduced dose of aflibercept significantly decreased both NV and VO. In contrast to anti-VEGF agents, the CITED2 peptide can rescue hypoxia-induced retinal NV by modulating the hypoxic response through direct competition with HIF for CBP/p300, suggesting a dual targeting strategy for treatment of ischemic retinal diseases and other neovascular disorders.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia/metabolism , Peptides/metabolism , Repressor Proteins/metabolism , Retinal Neovascularization/metabolism , Trans-Activators/metabolism , Animals , E1A-Associated p300 Protein/metabolism , Gene Expression , HEK293 Cells , Humans , Intrinsically Disordered Proteins/metabolism , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Protein Interaction Domains and Motifs , Repressor Proteins/genetics , Trans-Activators/genetics , p300-CBP Transcription Factors/metabolism
7.
Invest Ophthalmol Vis Sci ; 61(10): 20, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32780864

ABSTRACT

Purpose: Ciliary neurotrophic factor (CNTF) is a well-characterized neurotrophic factor currently in clinical trials for the treatment of macular telangiectasia type II. Our previous work showed that CNTF-induced STAT3 signaling is a potent inhibitor of pathologic preretinal neovascular tuft formation in the mouse model of oxygen-induced retinopathy. In this study, we investigated the effect of CNTF on outer retinal and choroidal angiogenesis and the mechanisms that underpin the observed decrease in outer retinal neovascularization following CNTF treatment. Methods: In the Vldlr-/- and laser-CNV mouse models, mice received a one-time injection (on postnatal day [P] 12 in the Vldlr-/- model and 1 day after laser in the Choroidal Neovascularization (CNV) model) of recombinant CNTF or CxCl10, and the extent of neovascular lesions was assessed 6 days posttreatment. STAT3 downstream targets affected by CNTF treatment were identified using quantitative PCR analysis. A proteome array was used to compare media conditioned by CNTF-treated and control-treated primary Müller cells to screen for CNTF-induced changes in secreted angiogenic factors. Results: Intravitreal treatment with recombinant CNTF led to significant reduction in neovascularization in the Vldlr-/- and laser-CNV mouse models. Treatment effect in the Vldlr-/- was long-lasting but time sensitive, requiring intravitreal treatment before P19. Mechanistic workup in vitro as well as in vivo confirmed significant activation of the STAT3-signaling pathway in Müller cells in response to CNTF treatment and upregulation of CxCl10. Intravitreal injections of recombinant CxCl10 significantly reduced outer retinal neovascularization in vivo in both the Vldlr-/- and laser-CNV mouse models. Conclusions: CNTF treatment indirectly affects outer retinal and choroidal neovascularization by inducing CxCl10 secretion from retinal Müller cells.


Subject(s)
Chemokine CXCL10/metabolism , Ciliary Neurotrophic Factor/therapeutic use , Retinal Neovascularization/prevention & control , Animals , Blotting, Western , Cells, Cultured , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/prevention & control , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Ependymoglial Cells , Immunohistochemistry , Laser Coagulation , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , STAT3 Transcription Factor/metabolism , Up-Regulation
8.
JCI Insight ; 5(12)2020 06 18.
Article in English | MEDLINE | ID: mdl-32437334

ABSTRACT

Abnormal subretinal neovascularization is a characteristic of vision-threatening retinal diseases, including macular telangiectasia (MacTel) and retinal angiomatous proliferation (RAP). Subretinal neovascular tufts and photoreceptor dysfunction are observed in very-low-density lipoprotein receptor (Vldlr-/-) mutant mice. These changes mirror those observed in patients with MacTel and RAP, but the pathogenesis is largely unknown. In this study, we show that retinal microglia were closely associated with retinal neovascular tufts in Vldlr-/- mice and retinal tissue from patients with MacTel; ablation of microglia/macrophages dramatically prevented formation of retinal neovascular tufts and improved neuronal function, as assessed by electroretinography. Vldlr-/- mice with retinal pigmented epithelium-specific (RPE-specific) Vegfa had greatly reduced subretinal infiltration of microglia/macrophages, subsequently reducing neovascular tufts. These findings highlight the contribution of microglia/macrophages to the pathogenesis of neovascularization, provide valuable clues regarding potential causative cellular mechanisms for subretinal neovascularization in patients with MacTel and RAP and suggest that targeting microglia activation may be a therapeutic option in these diseases.


Subject(s)
Macular Degeneration/pathology , Microglia/pathology , Retinal Neovascularization/pathology , Retinal Pigment Epithelium/pathology , Animals , Disease Models, Animal , Mice, Knockout , Neovascularization, Pathologic/pathology , Retina/pathology , Retinal Vessels/pathology
9.
Neurochem Int ; 129: 104489, 2019 10.
Article in English | MEDLINE | ID: mdl-31199961

ABSTRACT

Mutations in dystrophin are the major cause of muscular dystrophies. Continuous muscular degeneration and late stage complications, including cardiomyopathy and respiratory insufficiency, dominate the clinical phenotype. Gene expression and regulation of the dystrophin gene outside of muscular tissue is far more complex. Multiple tissue-specific dystrophin gene products are widely expressed throughout the body, including the central nervous system and eye, predisposing affected patients to secondary complications in non-muscular tissues. In this study, we evaluated the impact of the full-length dystrophin gene product, Dp427, on retinal homeostasis and angiogenesis. Based on the clinical case of a Duchenne muscular dystrophy (DMD) patient who developed severe fibrovascular changes in the retina in response to hypoxic stress, we hypothesized that defects in Dp427 make the retina more susceptible to stresses such as ageing and ischemia. To further study this, a mouse strain lacking Dp427 expression (Mdx) was studied during retinal development, ageing and in the oxygen-induced retinopathy (OIR) model. While retinal vascular morphology was normal during development and ageing, retinal function measured by electroretinography (ERG) was slightly reduced in young adult Mdx mice and deteriorated with age. Mdx mice also had increased retinal neovascularization in response to OIR and more pronounced long-term deterioration in retinal function following OIR. Based on these results, we suggest that DMD patients with a mutation in Dp427 may experience disturbed retinal homeostasis with increasing age and therefore be prone to develop excessive retinal neovascular changes in response to hypoxic stress. DMD patients in late disease stages should, thus, be regularly examined to detect asymptomatic retinal abnormalities and prevent visual impairment.


Subject(s)
Aging/physiology , Dystrophin/physiology , Ischemia/physiopathology , Muscular Dystrophy, Duchenne/pathology , Oxygen/toxicity , Retina/physiology , Retinal Diseases/physiopathology , Retinal Neovascularization/etiology , Retinal Vessels/ultrastructure , Aging/pathology , Animals , Cell Hypoxia , Dystrophin/genetics , Exons/genetics , Fibrosis , Gene Duplication , Humans , Ischemia/pathology , Male , Mice , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology , Retina/diagnostic imaging , Retinal Diseases/chemically induced , Retinal Diseases/pathology , Retinal Neovascularization/physiopathology , Sepsis/complications , Young Adult
10.
Glia ; 67(2): 332-344, 2019 02.
Article in English | MEDLINE | ID: mdl-30484883

ABSTRACT

Ischemia-induced angiogenesis contributes to various neuronal and retinal diseases, and often results in neurodegeneration and visual impairment. Current treatments involve the use of anti-VEGF agents but are not successful in all cases. In this study we determined that miR-30a-5p is another important mediator of retinal angiogenesis. Using a rodent model of ischemic retinopathy, we show that inhibiting miR-30a-5p reduces neovascularization and promotes tissue repair, through modulation of microglial and endothelial cell cross-talk. miR-30a-5p inhibition results in increased expression of the death receptor Fas and CCL2, to decrease endothelial cell survival and promote microglial migration and phagocytic function in focal regions of ischemic injury. Our data suggest that miR-30a-5p inhibition accelerates tissue repair by enhancing FasL-Fas crosstalk between microglia and endothelial cells, to promote endothelial cell apoptosis and removal of dead endothelial cells. Finally, we found that miR-30a levels were increased in the vitreous of patients with proliferative diabetic retinopathy. Our study identifies a role for miR-30a in the pathogenesis of neovascular retinal disease by modulating microglial and endothelial cell function, and suggests it may be a therapeutic target to treat ischemia-mediated conditions.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/metabolism , Microglia/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , fas Receptor/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , RNA Interference/physiology , RNA, Messenger/metabolism
11.
JCI Insight ; 2(24)2017 12 21.
Article in English | MEDLINE | ID: mdl-29263301

ABSTRACT

Oxygen-induced retinopathy (OIR) is a widely used model to study ischemia-driven neovascularization (NV) in the retina and to serve in proof-of-concept studies in evaluating antiangiogenic drugs for ocular, as well as nonocular, diseases. The primary parameters that are analyzed in this mouse model include the percentage of retina with vaso-obliteration (VO) and NV areas. However, quantification of these two key variables comes with a great challenge due to the requirement of human experts to read the images. Human readers are costly, time-consuming, and subject to bias. Using recent advances in machine learning and computer vision, we trained deep learning neural networks using over a thousand segmentations to fully automate segmentation in OIR images. While determining the percentage area of VO, our algorithm achieved a similar range of correlation coefficients to that of expert inter-human correlation coefficients. In addition, our algorithm achieved a higher range of correlation coefficients compared with inter-expert correlation coefficients for quantification of the percentage area of neovascular tufts. In summary, we have created an open-source, fully automated pipeline for the quantification of key values of OIR images using deep learning neural networks.


Subject(s)
Deep Learning , Retinal Neovascularization/diagnosis , Algorithms , Animals , Disease Models, Animal , Female , Image Processing, Computer-Assisted/methods , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal/methods , Observer Variation , Oxygen , Retinal Neovascularization/etiology , Retinal Neovascularization/pathology
12.
Circulation ; 136(2): 180-195, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28356444

ABSTRACT

BACKGROUND: Anti-angiogenic biologicals represent an important concept for the treatment of vasoproliferative diseases. However, the need for continued treatment, the presence of nonresponders, and the risk of long-term side effects limit the success of existing therapeutic agents. Although Tspan12 has been shown to regulate retinal vascular development, nothing is known about its involvement in neovascular disease and its potential as a novel therapeutic target for the treatment of vasoproliferative diseases. METHODS: Rodent models of retinal neovascular disease, including the mouse model of oxygen-induced retinopathy and the very low density lipoprotein receptor knockout mouse model were analyzed for Tspan/ß-catenin regulation. Screening of a phage display of a human combinatorial antibody (Ab) library was used for the development of a high-affinity Ab against Tspan12. Therapeutic effects of the newly developed Ab on vascular endothelial cells were tested in vitro and in vivo in the oxygen-induced retinopathy and very low density lipoprotein receptor knockout mouse model. RESULTS: The newly developed anti-Tspan12 Ab exhibited potent inhibitory effects on endothelial cell migration and tube formation. Mechanistic studies confirmed that the Ab inhibited the interaction between Tspan12 and Frizzled-4 and effectively modulates ß-catenin levels and target genes in vascular endothelial cells. Tspan12/ß-catenin signaling was activated in response to acute and chronic stress in the oxygen-induced retinopathy and very low density lipoprotein receptor mouse model of proliferative retinopathy. Intravitreal application of the Ab showed significant therapeutic effects in both models without inducing negative side effects on retina function. Moreover, combined intravitreal injection of the Ab with a known vascular endothelial growth factor inhibitor, Aflibercept, resulted in significant enhancement of the therapeutic efficacy of each monotherapy. Combination therapy with the Tspan12 blocking antibody can be used to reduce anti-vascular endothelial growth factor doses, thus decreasing the risk of long-term off-target effects. CONCLUSIONS: Tspan12/ß-catenin signaling is critical for the progression of vasoproliferative disease. The newly developed anti-Tspan12 antibody has therapeutic effects in vasoproliferative retinopathy and can enhance the potency of existing anti- vascular endothelial growth factor agents.


Subject(s)
Retinal Neovascularization/metabolism , Signal Transduction/physiology , Tetraspanins/antagonists & inhibitors , Tetraspanins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies/pharmacology , Antibodies/therapeutic use , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Intravitreal Injections , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Retinal Neovascularization/drug therapy , Signal Transduction/drug effects
13.
JCI Insight ; 2(2): e89906, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138561

ABSTRACT

Vascular abnormalities are a common component of eye diseases that often lead to vision loss. Vaso-obliteration is associated with inherited retinal degenerations, since photoreceptor atrophy lowers local metabolic demands and vascular support to those regions is no longer required. Given the degree of neurovascular crosstalk in the retina, it may be possible to use one cell type to rescue another cell type in the face of severe stress, such as hypoxia or genetically encoded cell-specific degenerations. Here, we show that intravitreally injected human endothelial colony-forming cells (ECFCs) that can be isolated and differentiated from cord blood in xeno-free media collect in the vitreous cavity and rescue vaso-obliteration and neurodegeneration in animal models of retinal disease. Furthermore, we determined that a subset of the ECFCs was more effective at anatomically and functionally preventing retinopathy; these cells expressed high levels of CD44, the hyaluronic acid receptor, and IGFBPs (insulin-like growth factor-binding proteins). Injection of cultured media from ECFCs or only recombinant human IGFBPs also rescued the ischemia phenotype. These results help us to understand the mechanism of ECFC-based therapies for ischemic insults and retinal neurodegenerative diseases.


Subject(s)
Endothelial Progenitor Cells/metabolism , Hyaluronan Receptors/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Ischemia/pathology , Retinal Diseases/pathology , Retinal Neurons/pathology , Retinal Vessels/pathology , Animals , Cell Differentiation , Endothelial Cells/metabolism , Endothelial Progenitor Cells/transplantation , Fetal Blood , Humans , Hyaluronic Acid/metabolism , Intravitreal Injections , Mice
14.
J Clin Invest ; 127(1): 199-214, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27918307

ABSTRACT

Outer retinal and renal glomerular functions rely on specialized vasculature maintained by VEGF that is produced by neighboring epithelial cells, the retinal pigment epithelium (RPE) and podocytes, respectively. Dysregulation of RPE- and podocyte-derived VEGF is associated with neovascularization in wet age-related macular degeneration (ARMD), choriocapillaris degeneration, and glomerular thrombotic microangiopathy (TMA). Since complement activation and genetic variants in inhibitory complement factor H (CFH) are also features of both ARMD and TMA, we hypothesized that VEGF and CFH interact. Here, we demonstrated that VEGF inhibition decreases local CFH and other complement regulators in the eye and kidney through reduced VEGFR2/PKC-α/CREB signaling. Patient podocytes and RPE cells carrying disease-associated CFH genetic variants had more alternative complement pathway deposits than controls. These deposits were increased by VEGF antagonism, a common wet ARMD treatment, suggesting that VEGF inhibition could reduce cellular complement regulatory capacity. VEGF antagonism also increased markers of endothelial cell activation, which was partially reduced by genetic complement inhibition. Together, these results suggest that VEGF protects the retinal and glomerular microvasculature, not only through VEGFR2-mediated vasculotrophism, but also through modulation of local complement proteins that could protect against complement-mediated damage. Though further study is warranted, these findings could be relevant for patients receiving VEGF antagonists.


Subject(s)
Complement Factor H/metabolism , Eye Proteins/metabolism , Podocytes/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/metabolism , Aged , Animals , Complement Factor H/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Eye Proteins/antagonists & inhibitors , Eye Proteins/genetics , Female , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mice , Mice, Knockout , Podocytes/pathology , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Retinal Pigment Epithelium/pathology , Thrombotic Microangiopathies/genetics , Thrombotic Microangiopathies/metabolism , Thrombotic Microangiopathies/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Sci Rep ; 6: 36659, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27834402

ABSTRACT

Macrophages, key cells of the innate immune system, are known to support angiogenesis but are not believed to directly form vessel walls. Here we show that macrophages structurally form primitive, NON-ENDOTHELIAL "vessels" or vascular mimicry (VM) channels in both tumor and angiogenesis in vivo models. These channels are functionally connected to the systemic vasculature as they are perfused by intravenously injected dye. Since both models share hypoxic micro-environments, we hypothesized that hypoxia may be an important mediator of VM formation. Indeed, conditional genetic depletion of myeloid-specific HIF-1α results in decreased VM network formation, dye perfusion and tumor size. Although the macrophage VM network shares some features with an endothelial vasculature, it is ultrastructurally different. Cancer stem cells have been shown to form vascular mimicry channels. Our data demonstrates that tumor-associated macrophages also form them. The identification of this novel type of vascular mimicry may help in the development of targeted cancer therapeutics.


Subject(s)
Blood Vessels/immunology , Macrophages/immunology , Neoplastic Stem Cells/immunology , Animals , Blood Vessels/pathology , Cell Hypoxia/immunology , Macrophages/pathology , Mice , Mice, Nude , Mice, Transgenic , Neoplastic Stem Cells/pathology
16.
Invest Ophthalmol Vis Sci ; 57(10): 4017-26, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27494343

ABSTRACT

PURPOSE: Retinal vascular disease represents a major cause for vision loss in the Western world. Recent research has shown that neuronal and vascular damage are closely related in retinal disease. Ciliary neurotrophic factor (CNTF) is a well-studied neurotrophic factor that is currently being tested in clinical trials for the treatment of retinal degenerative diseases and macular telangiectasia. However, little is known about its effect on retinal vasculature. In this study, we investigate the effects of CNTF in retinal neovascular disease using the mouse model of oxygen-induced retinopathy (OIR). METHODS: Newborn pups were exposed to 75% oxygen from postnatal day (P)7 to P12 and subsequently returned to room air. Ciliary neurotrophic factor was injected intravitreally at OIR P12 and the vaso-obliterated and neovascular areas were quantified at OIR P17. Immunohistochemistry, RNA, and protein analysis were used to identify CNTF-responsive cells. In vitro experiments were performed to analyze the effect of CNTF on endothelial and astroglial cells. RESULTS: In the OIR model, CNTF facilitated capillary regrowth and attenuated preretinal neovascularization in a dose-dependent manner. The protective effect of CNTF was mediated via activation of the JAK/STAT3/SOCS3 signaling pathway. Immunohistochemical studies identified endothelial cells among others as CNTF-responsive cells in the retina. In vitro studies confirmed the anti-angiogenic effect of CNTF on endothelial cell sprouting. CONCLUSIONS: This study provides evidence for a therapeutic potential of CNTF beyond degenerative retinal disease. Vasoproliferative retinopathies may benefit from a CNTF-dependent and SOCS3-mediated angiomodulatory effect.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Gene Expression Regulation , Retinal Degeneration/drug therapy , Retinal Neovascularization/drug therapy , Retinal Vessels/pathology , Suppressor of Cytokine Signaling 3 Protein/genetics , Up-Regulation , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice, Inbred C57BL , Oxygen/toxicity , Polymerase Chain Reaction , RNA/genetics , Recombinant Proteins/pharmacology , Retinal Degeneration/chemically induced , Retinal Degeneration/diagnosis , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Vessels/drug effects , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/biosynthesis
18.
Elife ; 52016 03 15.
Article in English | MEDLINE | ID: mdl-26978795

ABSTRACT

Photoreceptors are the most numerous and metabolically demanding cells in the retina. Their primary nutrient source is the choriocapillaris, and both the choriocapillaris and photoreceptors require trophic and functional support from retinal pigment epithelium (RPE) cells. Defects in RPE, photoreceptors, and the choriocapillaris are characteristic of age-related macular degeneration (AMD), a common vision-threatening disease. RPE dysfunction or death is a primary event in AMD, but the combination(s) of cellular stresses that affect the function and survival of RPE are incompletely understood. Here, using mouse models in which hypoxia can be genetically triggered in RPE, we show that hypoxia-induced metabolic stress alone leads to photoreceptor atrophy. Glucose and lipid metabolism are radically altered in hypoxic RPE cells; these changes impact nutrient availability for the sensory retina and promote progressive photoreceptor degeneration. Understanding the molecular pathways that control these responses may provide important clues about AMD pathogenesis and inform future therapies.


Subject(s)
Epithelial Cells/physiology , Hypoxia , Macular Degeneration/physiopathology , Photoreceptor Cells/physiology , Retinal Pigment Epithelium/physiology , Stress, Physiological , Animals , Disease Models, Animal , Mice
19.
Nat Med ; 22(4): 439-45, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26974308

ABSTRACT

Tissues with high metabolic rates often use lipids, as well as glucose, for energy, conferring a survival advantage during feast and famine. Current dogma suggests that high-energy-consuming photoreceptors depend on glucose. Here we show that the retina also uses fatty acid ß-oxidation for energy. Moreover, we identify a lipid sensor, free fatty acid receptor 1 (Ffar1), that curbs glucose uptake when fatty acids are available. Very-low-density lipoprotein receptor (Vldlr), which is present in photoreceptors and is expressed in other tissues with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acid. In the retinas of Vldlr(-/-) mice with low fatty acid uptake but high circulating lipid levels, we found that Ffar1 suppresses expression of the glucose transporter Glut1. Impaired glucose entry into photoreceptors results in a dual (lipid and glucose) fuel shortage and a reduction in the levels of the Krebs cycle intermediate α-ketoglutarate (α-KG). Low α-KG levels promotes stabilization of hypoxia-induced factor 1a (Hif1a) and secretion of vascular endothelial growth factor A (Vegfa) by starved Vldlr(-/-) photoreceptors, leading to neovascularization. The aberrant vessels in the Vldlr(-/-) retinas, which invade normally avascular photoreceptors, are reminiscent of the vascular defects in retinal angiomatous proliferation, a subset of neovascular age-related macular degeneration (AMD), which is associated with high vitreous VEGFA levels in humans. Dysregulated lipid and glucose photoreceptor energy metabolism may therefore be a driving force in macular telangiectasia, neovascular AMD and other retinal diseases.


Subject(s)
Fatty Acids/metabolism , Macular Degeneration/metabolism , Photoreceptor Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, LDL/metabolism , Retina/metabolism , Animals , Gene Expression Regulation , Glucose/metabolism , Humans , Ketoglutaric Acids/metabolism , Lipid Metabolism/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Oxidation-Reduction , Photoreceptor Cells/pathology , Receptors, G-Protein-Coupled/biosynthesis , Receptors, LDL/genetics , Retina/pathology , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Stem Cells Int ; 2016: 8470263, 2016.
Article in English | MEDLINE | ID: mdl-26880994

ABSTRACT

Phototransduction is accomplished in the retina by photoreceptor neurons and retinal pigment epithelium (RPE) cells. Photoreceptors rely heavily on the RPE, and death or dysfunction of RPE is characteristic of age-related macular degeneration (AMD), a very common neurodegenerative disease for which no cure exists. RPE replacement is a promising therapeutic intervention for AMD, and large numbers of RPE cells can be generated from pluripotent stem cells. However, questions persist regarding iPSC-derived RPE (iPS-RPE) viability, immunogenicity, and tumorigenesis potential. We showed previously that iPS-RPE prevent photoreceptor atrophy in dystrophic rats up until 24 weeks after implantation. In this follow-up study, we longitudinally monitored the same implanted iPS-RPE, in the same animals. We observed no gross abnormalities in the eyes, livers, spleens, brains, and blood in aging rats with iPSC-RPE grafts. iPS-RPE cells that integrated into the subretinal space outlived the photoreceptors and survived for as long as 2 1/2 years while nonintegrating RPE cells were ingested by host macrophages. Both populations could be distinguished using immunohistochemistry and electron microscopy. iPSC-RPE could be isolated from the grafts and maintained in culture; these cells also phagocytosed isolated photoreceptor outer segments. We conclude that iPS-RPE grafts remain viable and do not induce any obvious associated pathological changes.

SELECTION OF CITATIONS
SEARCH DETAIL
...