Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Diabetes ; 73(2): 280-291, 2024 Feb 01.
Article En | MEDLINE | ID: mdl-37986627

Vascular inflammation is known to cause degeneration of retinal capillaries in early diabetic retinopathy (DR), a major microvascular complication of diabetes. Past studies investigating these diabetes-induced retinal vascular abnormalities have focused primarily on the role of molecular or biochemical cues. Here we show that retinal vascular inflammation and degeneration in diabetes are also mechanically regulated by the increase in retinal vascular stiffness caused by overexpression of the collagen-cross-linking enzyme lysyl oxidase (LOX). Treatment of diabetic mice with LOX inhibitor ß-aminopropionitrile (BAPN) prevented the increase in retinal capillary stiffness, vascular intracellular adhesion molecule-1 overexpression, and leukostasis. Consistent with these anti-inflammatory effects, BAPN treatment of diabetic mice blocked the upregulation of proapoptotic caspase-3 in retinal vessels, which concomitantly reduced retinal capillary degeneration, pericyte ghost formation, and the diabetes-induced loss of contrast sensitivity in these mice. Finally, our in vitro studies indicate that retinal capillary stiffening is sufficient to increase the adhesiveness and neutrophil elastase-induced death of retinal endothelial cells. By uncovering a link between LOX-dependent capillary stiffening and the development of retinal vascular and functional defects in diabetes, these findings offer a new insight into DR pathogenesis that has important translational potential.


Diabetes Mellitus, Experimental , Diabetic Retinopathy , Retinal Degeneration , Mice , Animals , Endothelial Cells , Diabetes Mellitus, Experimental/complications , Aminopropionitrile/pharmacology , Retina/pathology , Diabetic Retinopathy/pathology , Inflammation/pathology , Retinal Vessels/pathology , Mice, Inbred C57BL
2.
Proc Natl Acad Sci U S A ; 120(19): e2221045120, 2023 05 09.
Article En | MEDLINE | ID: mdl-37126699

Chronic, progressive retinal diseases, such as age-related macular degeneration (AMD), diabetic retinopathy, and retinitis pigmentosa, arise from genetic and environmental perturbations of cellular and tissue homeostasis. These disruptions accumulate with repeated exposures to stress over time, leading to progressive visual impairment and, in many cases, legal blindness. Despite decades of research, therapeutic options for the millions of patients suffering from these disorders remain severely limited, especially for treating earlier stages of pathogenesis when the opportunity to preserve the retinal structure and visual function is greatest. To address this urgent, unmet medical need, we employed a systems pharmacology platform for therapeutic development. Through integrative single-cell transcriptomics, proteomics, and phosphoproteomics, we identified universal molecular mechanisms across distinct models of age-related and inherited retinal degenerations, characterized by impaired physiological resilience to stress. Here, we report that selective, targeted pharmacological inhibition of cyclic nucleotide phosphodiesterases (PDEs), which serve as critical regulatory nodes that modulate intracellular second messenger signaling pathways, stabilized the transcriptome, proteome, and phosphoproteome through downstream activation of protective mechanisms coupled with synergistic inhibition of degenerative processes. This therapeutic intervention enhanced resilience to acute and chronic forms of stress in the degenerating retina, thus preserving tissue structure and function across various models of age-related and inherited retinal disease. Taken together, these findings exemplify a systems pharmacology approach to drug discovery and development, revealing a new class of therapeutics with potential clinical utility in the treatment or prevention of the most common causes of blindness.


Diabetic Retinopathy , Macular Degeneration , Retinal Degeneration , Retinitis Pigmentosa , Humans , Retina/metabolism , Retinal Degeneration/metabolism , Retinitis Pigmentosa/metabolism , Macular Degeneration/pathology , Diabetic Retinopathy/metabolism
3.
Diabetologia ; 66(3): 590-602, 2023 03.
Article En | MEDLINE | ID: mdl-36698021

AIMS/HYPOTHESIS: Accumulating evidence suggests that leucocytes play a critical role in diabetes-induced vascular lesions and other abnormalities that characterise the early stages of diabetic retinopathy. However, the role of monocytes has yet to be fully investigated; therefore, we used Ccr2-/- mice to study the role of CCR2+ inflammatory monocytes in the pathogenesis of diabetes-induced degeneration of retinal capillaries. METHODS: Experimental diabetes was induced in wild-type and Ccr2-/- mice using streptozotocin. After 2 months, superoxide levels, expression of inflammatory genes, leucostasis, leucocyte- and monocyte-mediated cytotoxicity against retinal endothelial cell death, retinal thickness and visual function were evaluated. Retinal capillary degeneration was determined after 8 months of diabetes. Flow cytometry of peripheral blood for differential expression of CCR2 in monocytes was assessed. RESULTS: In nondiabetic mice, CCR2 was highly expressed on monocytes, and Ccr2-/- mice lack CCR2+ monocytes in the peripheral blood. Diabetes-induced retinal superoxide, expression of proinflammatory genes Inos and Icam1, leucostasis and leucocyte-mediated cytotoxicity against retinal endothelial cells were inhibited in diabetic Ccr2-deficient mice and in chimeric mice lacking Ccr2 only from myeloid cells. In order to focus on monocytes, these cells were immuno-isolated after 2 months of diabetes, and they significantly increased monocyte-mediated endothelial cell cytotoxicity ex vivo. Monocytes from Ccr2-deficient mice caused significantly less endothelial cell death. The diabetes-induced retinal capillary degeneration was inhibited in Ccr2-/- mice and in chimeric mice lacking Ccr2 only from myeloid cells. CONCLUSIONS/INTERPRETATION: CCR2+ inflammatory monocytes contribute to the pathogenesis of early lesions of diabetic retinopathy.


Diabetes Mellitus, Experimental , Diabetic Retinopathy , Retinal Degeneration , Animals , Mice , Diabetic Retinopathy/metabolism , Monocytes/metabolism , Endothelial Cells/metabolism , Superoxides/metabolism , Retinal Degeneration/metabolism , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred C57BL , Retinal Vessels/pathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
4.
Diabetologia ; 65(10): 1734-1744, 2022 10.
Article En | MEDLINE | ID: mdl-35852587

AIMS/HYPOTHESIS: Induction of intercellular adhesion molecule-1 (ICAM-1) has been implicated in the development of macrovascular and microvascular diseases such as diabetic retinopathy. Lesions of diabetic retinopathy are unique to the retina but the reason for this is unclear, as all tissues are exposed to the same hyperglycaemic insult. We tested whether diabetes induces ICAM-1 on the luminal surface of endothelial cells to a greater extent in the retina than in other tissues and the role of vision itself in that induction. METHODS: Experimental diabetes was induced in C57Bl/6J, P23H opsin mutant and Gnat1-/- × Gnat2-/- double knockout mice using streptozotocin. The relative abundance of ICAM-1 on the luminal surface of endothelial cells in retina and other tissues was determined by conjugating anti-ICAM-1 antibodies to fluorescent microspheres (2 µm), injecting them intravenously and allowing them to circulate for 30 min. After transcardial perfusion, quantification of microspheres adherent to the endothelium in tissues throughout the body was carried out by fluorescent microscopy or flow cytometry. Mice injected with lipopolysaccharide (LPS) were used as positive controls. The difference in leucostasis between retinal and non-retinal vasculature was evaluated. RESULTS: Diabetes significantly increased ICAM-1-mediated adherence of microspheres to retinal microvessels by almost threefold, independent of sex. In contrast, diabetes had a much smaller effect on endothelial ICAM-1 in other tissues, and more tissues showed a significant induction of endothelial ICAM-1 with LPS than with diabetes. The diabetes-induced increase in endothelial ICAM-1 in retinal vasculature was inhibited by blocking phototransduction in photoreceptor cells. Diabetes significantly increased leucostasis in the retina by threefold compared with a non-ocular tissue (cremaster). CONCLUSIONS/INTERPRETATION: The diabetes-induced upregulation of ICAM-1 on the luminal surface of the vascular endothelium varies considerably among tissues and is highest in the retina. Induction of ICAM-1 on retinal vascular endothelial cells in diabetes is influenced by vision-related processes in photoreceptor cells. The unique presence of photoreceptors in the retina might contribute to the greater susceptibility of this tissue to vascular disease in diabetes.


Diabetes Mellitus, Experimental , Diabetic Retinopathy , Intercellular Adhesion Molecule-1/metabolism , Animals , Endothelial Cells , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Opsins , Streptozocin
5.
Nat Nanotechnol ; 17(9): 1004-1014, 2022 09.
Article En | MEDLINE | ID: mdl-35851383

Targeted drug delivery to disease-associated activated neutrophils can provide novel therapeutic opportunities while avoiding systemic effects on immune functions. We created a nanomedicine platform that uniquely utilizes an α1-antitrypsin-derived peptide to confer binding specificity to neutrophil elastase on activated neutrophils. Surface decoration with this peptide enabled specific anchorage of nanoparticles to activated neutrophils and platelet-neutrophil aggregates, in vitro and in vivo. Nanoparticle delivery of a model drug, hydroxychloroquine, demonstrated significant reduction of neutrophil activities in vitro and a therapeutic effect on murine venous thrombosis in vivo. This innovative approach of cell-specific and activation-state-specific targeting can be applied to several neutrophil-driven pathologies.


Leukocyte Elastase , alpha 1-Antitrypsin Deficiency , Animals , Humans , Hydroxychloroquine/pharmacology , Leukocyte Elastase/metabolism , Mice , Nanomedicine , Neutrophils
6.
Invest Ophthalmol Vis Sci ; 62(13): 7, 2021 10 04.
Article En | MEDLINE | ID: mdl-34643662

Purpose: Previous studies indicate that leukocytes, notably neutrophils, play a causal role in the capillary degeneration observed in diabetic retinopathy (DR), however, the mechanism by which they cause such degeneration is unknown. Neutrophil elastase (NE) is a protease released by neutrophils which participates in a variety of inflammatory diseases. In the present work, we investigated the potential involvement of NE in the development of early DR. Methods: Experimental diabetes was induced in NE-deficient mice (Elane-/-), in mice treated daily with the NE inhibitor, sivelestat, and in mice overexpressing human alpha-1 antitrypsin (hAAT+). Mice were assessed for diabetes-induced retinal superoxide generation, inflammation, leukostasis, and capillary degeneration. Results: In mice diabetic for 2 months, deletion of NE or selective inhibition of NE inhibited diabetes-induced retinal superoxide levels and inflammation, and inhibited leukocyte-mediated cytotoxicity of retinal endothelial cells. In mice diabetic for 8 months, genetic deletion of NE significantly inhibited diabetes-induced retinal capillary degeneration. Conclusions: These results suggest that a protease released from neutrophils contributes to the development of DR, and that blocking NE activity could be a novel therapy to inhibit DR.


Diabetes Mellitus, Experimental/complications , Diabetic Retinopathy/metabolism , Neutrophils/enzymology , Peptide Hydrolases/blood , Retina/metabolism , Animals , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/etiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Retina/diagnostic imaging
7.
Am J Pathol ; 191(10): 1805-1821, 2021 10.
Article En | MEDLINE | ID: mdl-34214506

This study tested the hypothesis that diabetes promotes a greater than normal cytosolic calcium level in rod cells that activates a Ca2+-sensitive protease, calpain, resulting in oxidative stress and inflammation, two pathogenic factors of early diabetic retinopathy. Nondiabetic and 2-month diabetic C57Bl/6J and calpain1 knockout (Capn1-/-) mice were studied; subgroups were treated with a calpain inhibitor (CI). Ca2+ content was measured in photoreceptors using Fura-2. Retinal calpain expression was studied by quantitative RT-PCR and immunohistochemistry. Superoxide and expression of inflammatory proteins were measured using published methods. Proteomic analysis was conducted on photoreceptors isolated from untreated diabetic mice or treated daily with CI for 2 months. Cytosolic Ca2+ content was increased twofold in photoreceptors of diabetic mice as compared with nondiabetic mice. Capn1 expression increased fivefold in photoreceptor outer segments of diabetic mice. Pharmacologic inhibition or genetic deletion of Capn1 significantly suppressed diabetes-induced oxidative stress and expression of proinflammatory proteins in retina. Proteomics identified a protein (WW domain-containing oxidoreductase [WWOX]) whose expression was significantly increased in photoreceptors from mice diabetic for 2 months and was inhibited with CI. Knockdown of Wwox using specific siRNA in vitro inhibited increase in superoxide caused by the high glucose. These results suggest that reducing Ca2+ accumulation, suppressing calpain activation, and/or reducing Wwox up-regulation are novel targets for treating early diabetic retinopathy.


Calcium/metabolism , Calpain/metabolism , Diabetic Retinopathy/pathology , Inflammation/pathology , Oxidative Stress , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Animals , Calpain/genetics , Cell Line , Diabetic Retinopathy/complications , Diabetic Retinopathy/genetics , Diabetic Retinopathy/physiopathology , Enzyme Activation/drug effects , Gene Deletion , Gene Expression Regulation/drug effects , Glycoproteins/pharmacology , Inflammation/complications , Inflammation/genetics , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Proteome/metabolism , Retina/pathology , Severity of Illness Index , Superoxides/metabolism , Up-Regulation/drug effects , Vision, Ocular/drug effects , WW Domain-Containing Oxidoreductase/metabolism
8.
PLoS One ; 15(9): e0238727, 2020.
Article En | MEDLINE | ID: mdl-32941450

PURPOSE: Female mice have been found to be resistant to streptozotocin (STZ)-induced diabetes, and pre-clinical research related to diabetic complications commonly omits females. The purpose of this study was to develop a method to induce diabetes in female mice, and to determine if retinas of diabetic female mice develop molecular changes and histopathological abnormalities comparable to those which develop in male diabetic mice. METHODS: To induce diabetes, animals of both sexes received daily intraperitoneal (i.p.) injection of STZ for 5 consecutive days at 55 mg/kg BW (a dose that is known to induce diabetes in male mice) or for females, 75 mg/kg BW of STZ. Retinal abnormalities that have been implicated in the development of the retinopathy (superoxide generation and expression of inflammatory proteins, iNOS and ICAM-1) were evaluated at 2 months of diabetes, and retinal capillary degeneration was evaluated at 8 months of diabetes. RESULTS: Daily i.p. injection of STZ for 5 consecutive days at a concentration of 55 mg/kg BW was sufficient to induce diabetes in 100% of male mice, but only 33% of female mice. However, females did become hyperglycemic when the dose of STZ administered was increased to 75 mg/kg BW. The resulting STZ-induced hyperglycemia in female and male mice was sustained for at least 8 months. After induction of the diabetes, both sexes responded similarly with respect to the oxidative stress, expression of iNOS, and degeneration of retinal capillaries, but differed in the limited population evaluated with respect to expression of ICAM-1. CONCLUSIONS: The resistance of female mice to STZ-induced diabetes can be overcome by increasing the dose of STZ used. Female mice can, and should, be included in pre-clinical studies of diabetes and its complications.


Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Disease Models, Animal , Sex Characteristics , Animals , Capillaries/drug effects , Capillaries/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Gene Expression Regulation/drug effects , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Retina/drug effects , Retina/pathology , Streptozocin/pharmacology
9.
Diabetologia ; 62(12): 2365-2374, 2019 12.
Article En | MEDLINE | ID: mdl-31612267

AIMS/HYPOTHESIS: Levels of neutrophil elastase, a serine protease secreted by neutrophils, are elevated in diabetes. The purpose of this study was to determine whether neutrophil elastase (NE) contributes to the diabetes-induced increase in retinal vascular permeability in mice with streptozotocin-induced diabetes, and, if so, to investigate the potential role of IL-17 in this process. METHODS: In vivo, diabetes was induced in neutrophil elastase-deficient (Elane-/-), Il-17a-/- and wild-type mice. After 8 months of diabetes, Elane-/- mice and wild-type age-matched control mice were injected with FITC-BSA. Fluorescence microscopy was used to assess leakage of FITC-BSA from the retinal vasculature into the neural retina. The level of NE in Il-17a-/- diabetic retina and sera were determined by ELISA. In vitro, the effect of NE on the permeability and viability of human retinal endothelial cells and the expression of junction proteins and adhesion molecules were studied. RESULTS: Eight months of diabetes resulted in increased retinal vascular permeability and levels of NE in retina and plasma of wild-type animals. All of these abnormalities were significantly inhibited in mice lacking the elastase. The diabetes-induced increase in NE was inhibited in mice lacking IL-17. In vitro, NE increased retinal endothelial cell permeability, which was partially inhibited by a myeloid differentiation primary response 88 (MyD88) inhibitor, NF-κB inhibitor, and protease-activated receptor (PAR)2 inhibitor. NE degraded vascular endothelial-cadherin (VE-cadherin) in a concentration-dependent manner. CONCLUSIONS/INTERPRETATION: IL-17 regulates NE expression in diabetes. NE contributes to vascular leakage in diabetic retinopathy, partially through activation of MyD88, NF-κB and PAR2 and degradation of VE-cadherin.


Blood-Retinal Barrier/metabolism , Diabetic Retinopathy/metabolism , Leukocyte Elastase/metabolism , Retina/metabolism , Retinal Vessels/metabolism , Animals , Blood-Retinal Barrier/pathology , Capillary Permeability/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Interleukin-17/genetics , Interleukin-17/metabolism , Leukocyte Elastase/genetics , Male , Mice , Mice, Knockout , Retina/pathology , Retinal Vessels/pathology
10.
PLoS One ; 14(4): e0213960, 2019.
Article En | MEDLINE | ID: mdl-30970040

Mutations in the gene Centrosomal Protein 290 kDa (CEP290) result in multiple ciliopathies ranging from the neonatal lethal disorder Meckel-Gruber Syndrome to multi-systemic disorders such as Joubert Syndrome and Bardet-Biedl Syndrome to nonsyndromic diseases like Leber Congenital Amaurosis (LCA) and retinitis pigmentosa. Results from model organisms and human genetics studies, have suggest that mutations in genes encoding protein components of the transition zone (TZ) and other cilia-associated proteins can function as genetic modifiers and be a source for CEP290 pleiotropy. We investigated the zebrafish cep290fh297/fh297 mutant, which encodes a nonsense mutation (p.Q1217*). This mutant is viable as adults, exhibits scoliosis, and undergoes a slow, progressive cone degeneration. The cep290fh297/fh297 mutants showed partial mislocalization of the transmembrane protein rhodopsin but not of the prenylated proteins rhodopsin kinase (GRK1) or the rod transducin subunit GNB1. Surprisingly, photoreceptor degeneration did not trigger proliferation of Müller glia, but proliferation of rod progenitors in the outer nuclear layer was significantly increased. To determine if heterozygous mutations in other cilia genes could exacerbate retinal degeneration, we bred cep290fh297/fh297 mutants to arl13b, ahi1, and cc2d2a mutant zebrafish lines. While cep290fh297/fh297 mutants lacking a single allele of these genes did not exhibit accelerated photoreceptor degeneration, loss of one alleles of arl13b or ahi1 reduced visual performance in optokinetic response assays at 5 days post fertilization. Our results indicate that the cep290fh297/fh297 mutant is a useful model to study the role of genetic modifiers on photoreceptor degeneration in zebrafish and to explore how progressive photoreceptor degeneration influences regeneration in adult zebrafish.


Cilia/pathology , Microtubule-Associated Proteins/genetics , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Visual Acuity/genetics , Zebrafish Proteins/genetics , ADP-Ribosylation Factors/genetics , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Cell Survival/genetics , Cilia/genetics , Disease Models, Animal , Humans , Mutation , Retinal Cone Photoreceptor Cells/cytology , Retinal Degeneration/pathology , Vesicular Transport Proteins/genetics , Zebrafish
11.
Invest Ophthalmol Vis Sci ; 58(1): 448-460, 2017 01 01.
Article En | MEDLINE | ID: mdl-28118669

Purpose: Joubert syndrome (JBTS) is an autosomal recessive ciliopathy with considerable phenotypic variability. In addition to central nervous system abnormalities, a subset of JBTS patients exhibit retinal dystrophy and/or kidney disease. Mutations in the AHI1 gene are causative for approximately 10% of all JBTS cases. The purpose of this study was to generate ahi1 mutant alleles in zebrafish and to characterize the retinal phenotypes. Methods: Zebrafish ahi1 mutants were generated using transcription activator-like effector nucleases (TALENs). Expression analysis was performed by whole-mount in situ hybridization. Anatomic and molecular characterization of photoreceptors was investigated by histology, electron microscopy, and immunohistochemistry. The optokinetic response (OKR) behavior assay was used to assess visual function. Kidney cilia were evaluated by whole-mount immunostaining. Results: The ahi1lri46 mutation in zebrafish resulted in shorter cone outer segments but did not affect visual behavior at 5 days after fertilization (dpf). No defects in rod morphology or rhodopsin localization were observed at 5 dpf. By 5 months of age, cone degeneration and rhodopsin mislocalization in rod photoreceptors was observed. The connecting cilium formed normally and Cc2d2a and Cep290 localized properly. Distal pronephric duct cilia were absent in mutant fish; however, only 9% of ahi1 mutants had kidney cysts by 5 dpf, suggesting that the pronephros remained largely functional. Conclusions: The results indicate that Ahi1 is required for photoreceptor disc morphogenesis and outer segment maintenance in zebrafish.


Carrier Proteins/genetics , Cilia/ultrastructure , Ciliopathies/genetics , Morphogenesis , Mutation , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Zebrafish Proteins/genetics , Animals , Carrier Proteins/metabolism , Cell Survival , Cilia/metabolism , Ciliopathies/metabolism , Ciliopathies/pathology , DNA Mutational Analysis , Genotype , Immunohistochemistry , In Situ Hybridization , Microscopy, Electron, Transmission , Proto-Oncogene Proteins , Retinal Photoreceptor Cell Outer Segment/metabolism , Zebrafish , Zebrafish Proteins/metabolism
12.
Exp Eye Res ; 153: 65-78, 2016 Dec.
Article En | MEDLINE | ID: mdl-27720860

Non-invasive imaging is an invaluable diagnostic tool in ophthalmology. Two imaging devices, the scanning laser ophthalmoscope (SLO) and spectral domain optical coherence tomography (SDOCT), emerged from the clinical realm to provide research scientists with a real-time view of ocular morphology in living animals. We utilized these two independent imaging modalities in a complementary manner to perform in vivo optical sectioning of the adult zebrafish retina. Due to the very high optical power of the zebrafish lens, the confocal depth of field is narrow, allowing for detailed en face views of specific retinal layers, including the cone mosaic. Moreover, we demonstrate that both native reflectance, as well as fluorescent features observed by SLO, can be combined with axial in-depth information obtained by SDOCT. These imaging approaches can be used to screen for ocular phenotypes and monitor retinal pathology in a non-invasive manner.


Ophthalmoscopy/methods , Retinal Ganglion Cells/pathology , Tomography, Optical Coherence/methods , Animals , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Reproducibility of Results , Retinal Diseases/diagnosis , Zebrafish
...